Echinacoside‑induced nitric oxide production in endothelial cells: Roles of androgen receptor and the PI3K‑Akt pathway

  • Authors:
    • Li Gu
    • Danhong Lian
    • Yimei Zheng
    • Wei Zhou
    • Jinlei Gu
    • Xin Liu
  • View Affiliations

  • Published online on: January 27, 2020     https://doi.org/10.3892/ijmm.2020.4476
  • Pages: 1195-1202
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Echinacoside (ECH) is a natural compound with an endothelium‑dependent vasodilatory effect. Nitric oxide (NO) is an important vasorelaxant released from endothelial cells. In order to examine the molecular mechanism of ECH‑induced NO production in endothelial cells, the present study investigated the involvement of androgen receptor (AR) and the phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (Akt) pathway in the phosphorylation of endothelial nitric oxide synthase (eNOS) in human umbilical vein endothelial cells (HUVECs). Using the fluorescent probe DAF‑FM, the production of NO was found to be significantly increased, and eNOS was phosphorylated at Ser1177 in a concentration‑​dependent manner under 0.01‑10 µM ECH treatment in HUVECs. In addition, NO production and eNOS phosphorylation induced by ECH were diminished when pretreated with the AR antagonist nilutamide, or when transfected with AR small interfering RNAs. Furthermore, the ECH‑induced phosphorylation of the Akt at Ser473 was abrogated by 5 µM wortmannin (a PI3K inhibitor). These data indicated that ECH stimulated NO production via the AR‑dependent activation of eNOS in HUVECs, and that the PI3K/Akt pathway may be involved in eNOS phosphorylation induced by ECH.

Introduction

Cistanche Hoffmg. Et Link is a perennial parasite herb of a genus of the Orobanchaceae family. Herba Cistanche, the stem of Cistanche deserticola Y. C. MA and Cistanche tubulosa (Schenk) R. Weight, have been used as a tonic agent to treat kidney deficiency, impotence, morbid leukorrhea and senile constipation (1), which may be due to its androgen-like or sex hormone regulatory effect (2). Echinacoside (ECH; C35H46O20; molecular weight, 786.73; Fig. 1), is one of the phenylethanoid glycosides (PhGs) isolated from the stems of Herba Cistanche, and exhibits multiple biological properties, including antioxidant, antiaging, anti-inflammatory, hepatoprotective and neuroprotective effects (3). Modern pharmacology investigations have demonstrated that various constituents of Herba Cistanche, including ECH, acteoside, kankanose, kankanoside F and cistanoside F, exhibit vasorelaxant activity (4).

The endothelium is a critical regulator of the vasculature, and nitric oxide (NO) is an important relaxing factor released by endothelial cells. By diffusing into smooth muscle cells, NO activates guanylate cyclase, increases the levels of cyclic guanosine monophosphate (cGMP), and then activates cGMP-dependent protein kinases (PKGs) to promote smooth muscle relaxation (5). It was previously demonstrated that ECH at 350-400 µM directly acts on vascular smooth muscle and inhibits hypoxia-induced proliferation in rat pulmonary artery smooth muscle cells (6), whereas 30-300 µM ECH caused acute vasorelaxation in endothelium-intact rings in a concentration-dependent manner, and enhanced cGMP production in the corpus cavernosum smooth muscle of aortic rings contracted by phenylephrine (7). By opening the NO-cGMP-PKG-BKCa channels in smooth muscle cells, 100 or 300 µM ECH suppressed noradrenaline-induced contraction in the pulmonary arteries of rats, particularly in endothelium-denuded rings (8). Endothelial cells are key regulators of cardiovascular function and, in several cases, it has been found that the endothelial-dependent relaxation was due to a transferrable substance, such as NO, released from the endothelium (9). However, the upstream molecular mechanism of ECH-induced NO production in vascular endothelial cells requires further investigation.

In the vascular endothelium, NO generation is primarily mediated by endothelial NO synthase (eNOS). Several groups have demonstrated that the phosphatidylinositol 3-kinase (PI3K) pathway activates the serine threonine protein kinase B (Akt), which causes direct eNOS phosphorylation at serine 1177 (Ser1177) (10). The androgen receptor (AR) is a member of the nuclear receptor subfamily s3, which canonically modifies gene expression. AR localization to the caveolae in the cell membrane is involved in the non-genomic regulation of endothelial cell function or gene expression by triggering the c-Src/PI3K/Akt cascade, which ultimately results in eNOS phosphorylation and NO production (11). In human aortic endothelial cells, testosterone has been reported to activate PI3K/Akt signaling and rapidly induce NO production due to the direct interaction of AR and the p85α subunit of PI3K on the cardiovascular system (12). It was reported that ECH aggravates hormone deficiency-related symptoms and exerts its androgen-like effects due to competitive binding to the AR instead of testosterone (2). Therefore, the present study hypothesized that the PI3K/Akt pathway may be involved in NO production through AR-dependent eNOS phosphorylation induced by ECH. The aim of the present study was to evaluate the following effects of ECH: i) Induction of NO production and eNOS phosphorylation; ii) involvement of AR in eNOS phosphorylation; and iii) activation of the PI3K/Akt pathway in human umbilical vein endothelial cells (HUVECs), a well-known experimental model for studying the regulation of endothelial cell functions and angiogenesis (13).

Materials and methods

Chemicals and reagents

ECH (purity, 92.5%) was obtained from the National Drug Reference Standards in National Institute for the Food and Drug Control. Antibodies against p-Akt (Ser473; cat. no. ab8805), Akt (cat. no. ab81283), p-eNOS (Ser1177; cat. no. ab184154) and eNOS (cat. no. ab76198) were purchased from Abcam. The inhibitors of nilutamide and ICI 182780 were obtained from Sigma-Aldrich; Merck KGaA. L-NAME was purchased from Adamas-Beta, Ltd., and wortmannin was purchased from Pribolab.

Cell culture and drug treatment

HUVECs were obtained from ScienCell Research Laboratories Inc. and cultured in endothelial cell medium (ECM; ScienCell Research Laboratories) with 5% (v/v) fetal bovine serum (FBS; Gibco, Thermo Fisher Scientific, Inc.) and 1% endothelial cell growth supplement (ECGS) at 37°C (5% CO2 and 95% humidity) (14). Upon reaching confluence, the cells were digested with trypsin and plated in ECM with 1% FBS and 1% ECGS. For all experiments, HUVECs were plated at a concentration of 1×104/ml and grown until reaching confluence. Prior to treatment with ECH or other stimulators, the cells were incubated in phenol red-free ECM without FBS and ECGS for 6 h to induce growth arrest. In the inhibitory experiments, HUVECs were pre-incubated with various antagonists or inhibitors, including 10 µM nilutamide, 10 µM ICI 182780, 0.5 mM L-NAME or 5 µM wortmannin, for 30 min, with or without ECH, at 37°C. In all groups, including the control, DMSO was used as a solvent at equal concentrations of 0.001%.

Measurement of intracellular NO production

Relative changes in cytosolic NO concentration in HUVECs were monitored using the fluorescent NO probe DAF-FM (Cayman Chemical Company), as previously reported (12). Briefly, the cells were loaded with 5 µM DAF-FM diacetate for 20 min at 37°C in a black microtiter plate and rinsed several times with PBS (pH 7.4). The fluorescence was determined at excitation and emission wavelengths of 495 and 515 nm, respectively, using a fluorescent microplate reader (Biotek Synergy H4; BioTek Instruments, Inc.) and a compact inverted microscope (Nikon eclipse Ts2R; Nikon Corporation).

Western blot analysis

As previously reported (15), confluent monolayers of cells were washed twice in ice-cold PBS and lysed with RIPA buffer (P0013D; Beyotime Institute of Biotechnology). The protein concentration in the supernatant was measured using the bicinchoninic acid assay method (16). Subsequently, 30 µg protein were loaded per lane, separated using 10% polyacrylamide gels and transferred onto polyvi-nylidene difluoride membranes. The membranes were blocked with 5% skimmed milk for 1 h at 25°C. After incubation with monoclonal antibodies against Akt (1:1,000 dilution), p-Akt (1:500 dilution), eNOS (1:2,000 dilution), or p-eNOS (1:1,000 dilution) at 4°C overnight, the membranes were washed with TBST (containing 0.1% Tween-20) 4 times for ~15 min per wash at 25°C. Subsequently, the membranes were incubated with horseradish peroxidase-conjugated secondary antibodies (1:5,000 dilution; anti-mouse antibody, cat. no. A0216, or anti-rabbit antibody, cat. no. A0208, Beyotime Institute of biotechnology) for 1 h at 25°C, and detected using an enhanced chemiluminescence kit (cat. no. 32209, Thermo Fisher Scientific, Inc.). Band intensities were quantified using Image J gel analysis software, version 1.8.0_112 (National Institutes of Health).

Small interfering (si) RNA preparation and transfection

The long double-stranded RNAs were synthesized by the target mRNA of the AR and estrogen receptor (ER) with sequences shown in Table I. The conditions for siRNA knockdown involved transfecting HUVECs at 70% confluence maintained in non-antimicrobial culture medium in 60 mm collagen-coated culture dishes. Transfection of 5 nM AR-siRNA or 10 nM ERα-siRNA with Lipofectamine 3000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was performed separately, according to the manufacturer's protocols. Transfection efficiency was evaluated by reverse transcription quantitative-PCR analysis (RT-qPCR), as previously reported (17). The thermo-cycling conditions were as follows: 10 min at 95°C; 40 cycles of 95°C for 5 sec and 60°C for 1 min, and a melting curve at 95°C for 15 sec, 60°C for 1 min and 95°C for 15 sec; three independent biological replicates were performed for each sample. The subsequent experiments were conducted 48 h after transfection. Primer pairs were designed using Primer Premier v5.0 software (PREMIER Biosoft) with the following sequences: AR forward, 5′-GGT TAC ACC AAA GGG CTA GAA-3′ and reverse, 5′-GAC TTG TAG AGA GAC AGG GTA GA-3′; ERα forward, 5′-CCA GTA CCA ATG ACA AGG GAA G-3′ and reverse, 5′-TCA CAG GAC CAG ACT CCA TAA-3′; and GAPDH forward, 5′-CAG GGC TGC TTT TAA CTC TGG TAA-3′ and reverse, 5′-GGG TGG AAT CAT ATT GGA ACA TGT-3′.

Table I

The sequences of the sense RNA strand targeting AR and ER in siRNA experiments.

Table I

The sequences of the sense RNA strand targeting AR and ER in siRNA experiments.

Target geneNameSequences (5′-3′)
ARsiRNA-1 AAACAGGUACUUCUGUUUCCC
siRNA-2 AAUGCAAAGGUUCUCUGCUAG
siRNA-3 AAGGUCUUCUUCAAAAGAGCC
ERαsiRNA-1 GAUCAAACGCUCUAAGAAG
siRNA-2 GAAUGUGCCUGGCUAGAGA
siRNA-3 GAUGAAAGGUGGGAUACGA
ERβsiRNA-1 GGAAAUGCGUAGAAGGAAU
siRNA-2 UUCAAGGUUUCGAGAGUUA
siRNA-3 GCACGGCUCCAUAUACAUA
ControlsiRNA-random AAGUGCGAUCUAACUGACCUA

[i] AR, androgen receptor; ER, estrogen receptor.

Statistical analysis

All data are presented as the mean ± standard deviation. Statistical comparisons between groups were performed using the Kruskal-Wallis test or two-way ANOVA with Tukey's post hoc test for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

ECH induces NO production and eNOS phosphorylation

As shown in Fig. 2A and B, 1 µM ECH significantly increased intracellular NO production in HUVECs compared with the negative control cells, while the stimulatory effect of ECH was attenuated to the control level by pretreatment with L-NAME. To obtain the optimal concentration, eNOS phosphorylation was tested at 60 min after treatment with ECH at concentrations of 0, 0.01, 0.1, 1 and 10 µM. The results revealed that ECH at concentrations of 0.01-10 µM may significantly induced eNOS phosphorylation at Ser 1177 in a concentration-dependent manner, with the maximal eNOS phosphorylation observed at 10 µM ECH induction (Fig. 2C and D). Furthermore, eNOS phosphorylation at Ser1177 was examined by western blot analysis at 0, 5, 15, 30, 60 and 120 min after incubation with 1 µM ECH. It was observed that eNOS phosphorylation was rapidly triggered by ECH at 5 min, and continued to increase until 30 min of incubation (Fig. 2E and F). Subsequently, despite ECH treatment, the relative magnitude of eNOS phosphorylation remained stable from 30 min onwards; therefore, ECH did not affect total eNOS expression (Fig. 2C and E).

AR mediates ECH-induced NO production and eNOS phosphorylation

RT-qPCR assay demonstrated that the interfering effects of AR-siRNA-1 and ERα-siRNA-2 were the most successful in inhibiting the expression of AR and ER in HUVECs in the present study (Fig. 3). Subsequently, an antagonist for AR inhibition-of-function analysis and siRNA for AR loss-of-function analysis were applied to evaluate the involvement of AR in ECH-induced eNOS activation and NO production. As shown in Fig. 4A, 1 µM ECH significantly increased NO production in HUVECs (P<0.05). Pretreatment with the AR antagonist nilutamide (10 µM) abolished ECH-induced NO production, whereas ICI182789 (an ER antagonist) did not exert the same effects. Furthermore, the effects of siRNA-mediated AR knockdown on NO production were examined in cultured cells, indicating that NO production was diminished by transfection with AR siRNAs; however, it was not affected by ER siRNAs or control random siRNAs (Fig. 4B). Representative western blots and semi-quantitative analysis (Fig. 4C and D) revealed that the phosphorylation of eNOS induced by ECH was inhibited by nilutamide and ICI182789, and that the inhibitory effect of nilutamide on ECH-induced eNOS activation was significantly higher compared with that of ICI182789, which indicated that inhibition of AR function had a greater impact than ER on eNOS phosphorylation induced by ECH. Similarly, the phosphorylation of eNOS induced by ECH was reduced significantly in cells transfected with AR-siRNA and ER-siRNA compared with the control random siRNA; the inhibitory effect of AR-siRNA on ECH-induced eNOS activation was significantly stronger compared with that of ER-siRNA (Fig. 4E and F). Therefore, the aforementioned results suggested that ECH may cause AR-dependent activation of eNOS to induce NO production in HUVECs.

ECH activates the PI3K/Akt pathway

In the present study, Akt phosphorylation at Ser473 was tested 60 min after ECH incubation at concentrations of 0, 0.01, 0.1, 1 and 10 µM. The results indicated that ECH (0.01-10 µM) may significantly induce Akt phosphorylation. The relative expression levels of p-Akt peaked at 1 and 10 µM of ECH treatment (Fig. 5A and B). Furthermore, Akt phosphorylation was examined by western blot analysis at 0, 5, 15, 30, 60 and 120 min after the addition of 1 µM ECH to HUVEC cultures. As shown in Fig. 5C and D, ECH rapidly increased Akt phosphorylation after 5 min of incubation, and the maximum protein level of p-Akt was observed at 60 min. By contrast, ECH did not affect the total Akt expression (Fig. 5A and C). Moreover, to investigate the potential effect of PI3K pathway on eNOS phosphorylation, HUVECs were pre-treated with the PI3K inhibitor wortmannin prior to ECH application. Wortmannin was found to reduce ECH-induced NO production to baseline levels (Fig. 5E). A similar phenomenon was observed using fluorescence microscopy when examining ECH-induced DAF-FM fluorescence (Fig. 5F). Furthermore, wortmannin may have the ability to abolish rapid eNOS phosphorylation in HUVECs (Fig. 5G and H). The aforementioned results suggest that ECH may activate eNOS phosphorylation and NO production via the PI3K/Akt pathway.

Discussion

ECH is a natural compound isolated from Herba Cistanche, with various pharmacological properties. It was previously demonstrated that ECH exerted an endothelium-dependent vascular relaxation effect by opening NO-cGMP-PKG-BKCa channels in blood vessel smooth muscle cells (7,8). The current findings provide evidence that ECH exerted AR-dependent activation of eNOS to induce NO production with the involvement of the PI3K/Akt signaling pathway in vascular endothelial cells.

As the innermost layer of the vessel wall, the endothelium can quickly sense and respond to changes in blood flow, in turn resulting in signal transmission to the underlying smooth muscle cells in order to regulate vascular tone (18). It has been widely reported that multiple endothelial-derived vasodilatory compounds exist, with the most prototypical substance being NO, formed from the endothelial isoform of eNOS, which results in phosphorylation (19). Under normal conditions, eNOS remains inactive when bound to caveolin, and is activated with the following succession of events in endothelial cells: i) eNOS dissociates from caveolin-1 and associates with Ca2+/CaM; ii) heat shock protein (HSP)90 promotes eNOS dimerization and favors a steric formation to recruit Akt; and iii) calcineurin dephosphorylates Thr495 to reinforce the activation state (20). In the present study, NO production was significantly increased in HUVECs treated with 1 µM ECH. This phenomenon was similar to previous results, where ECH increased NO release and stimulated the synthesis of cGMP in rat thoracic aortic rings (7). Furthermore, two phosphorylation sites, Ser1177 and Thr495, appear to be particularly important for regulating eNOS activity (20), and the present study revealed that ECH induced acute eNOS phosphorylation at Ser1177 in a concentration-dependent manner. In addition, the stimuli associated with the effects on eNOS protein levels mainly determine eNOS mRNA stability, and if the stimulus is maintained for a longer period of time, eNOS mRNA transcription occurs via mitogen-activated protein kinase and nuclear factor κB (20). Taken together, these observations indicate that the activation of eNOS at Ser1177 is responsible for NO synthesis induced by ECH.

Furthermore, an increasing number of studies have demonstrated that AR is expressed in endothelial cells in a number of human tissues, which suggests a potential role for androgens and their analogues, that act through AR-mediated processes, in the modulation of human endothelial cell homeostasis (21). In the non-classical PI3K/Akt pathway, AR may activate PI3K by directly interacting with PI3K regulatory subunit p85α (22). The present study demonstrated that an AR antagonist or AR siRNA diminished the NO production and eNOS phosphorylation induced by ECH in HUVECs. It has been previously reported that estrogens induce eNOS phosphorylation and stimulate NO production via classic ER activation in endothelial cells (23), and the administration of ECH significantly enhances the expression of ER in the uterus (24). However, in the present study, the effect of AR was more prominent compared with that of ERα on ECH-induced eNOS activation and NO production. In addition, it was observed that ECH caused acute NO production within minutes via AR-involved eNOS activation in HUVECs, which is consistent with the non-genomic nature of the response in endothelial cells. The AR is associated with scaffolding proteins, including HSP90, HSP70 and kinase Src, in the cytoplasm, and it can be transported to the membrane from the AR complex within 5 min of testosterone treatment (23). Based on the 'target fishing' strategy, HSP90 was identified as the PhGs-coupled target, which indicated that ECH may facilitate the dissociation of AR from the scaffolding proteins (25). Another study suggested that, in the hypothalamus, ECH may combine with the AR pocket at amino acids Met-894 and Val-713 and inhibit the transport of cytoplasmic AR to the nucleus (26). However, the underlying mechanism through which ECH mediates cytoplasmic AR translocation to the membrane requires further investigation. Therefore, further research is required to elucidate the mechanism through which ECH achieves AR-dependent eNOS activation and how it may be associated with binding to HSP90 in vascular endothelial cells.

The PI3K/Akt pathway is one of the most important signaling cascades, the activation of which is induced by producing phosphatidylinositol-3,4,5-trisphosphate to bind the N-terminal pleckstrin homology domain of the Ser/Thr kinase Akt. This facilitates Akt recruitment to the plasma membrane (27). The PI3K/Akt pathway may play an important role in the control of NO-dependent relaxation induced by ECH. The present study revealed that ECH-induced NO production was significantly reduced when the cells were incubated with the PI3K inhibitor wortmannin. A previous report demonstrated that 15 mg/kg ECH activated the PI3K/Akt signaling pathway in 5-fluorouracil-suppressed bone marrow cells (28). In the present study, PI3K inhibitors significantly decreased ECH-induced eNOS phosphorylation at Ser1177. Moreover, Akt activity was predominantly regulated by upstream regulatory pathways, particularly PI3K-dependent phosphorylation at Ser473 (20). In the present study, ECH induced Akt phosphorylation at Ser473 in a dose-dependent manner; similarly, a previous study demonstrated that 5, 10 or 20 µM ECH exerted a cardioprotective effect against anoxia/reperfusion treatment in a dose-dependent manner by potentially upregulating p-Akt and SLC8A3 (29). The transcriptional regulation of the Akt gene remains largely unknown (30); therefore, the present study focused on the post-transcriptional regulatory effects of ECH on Akt. Taking the aforementioned findings into consideration, it was inferred that the application of ECH to HUVECs may lead to the activation of the PI3K/Akt pathway, which phosphorylates eNOS and, subsequently, increases NO production.

In conclusion, ECH is a natural product that is mainly isolated from Herba Cistanche. The potential mechanism underlying ECH-induced NO production in endothelial cells may include the following (Fig. 6): i) ECH acts as a functional ligand of AR that is localized to the caveolae in the cell membrane; ii) PI3K binds to the hydrophobic domain of Akt at Ser473 and facilitates Akt recruitment to the cell membrane; iii) the recruitment of the PI3K/Akt cascades triggers AR-dependent eNOS phosphorylation; and iv) the generation of NO is mediated by eNOS in endothelial cells. The observation that ECH induces NO production through the AR-dependent phosphorylation of eNOS with the involvement of the PI3K/Akt pathway may contribute to further understanding of the vasorelaxant effects of ECH. Furthermore, the ECH targeting the endothelial-derived NO pathways may be due to non-genomic effects. Therefore, the present study may help elucidate the mechanisms through which ECH exerts its pharmacological effects to prevent cardiovascular disease.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81503333).

Availability of data and materials

The data sets generated and analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

LG and XL conceived and designed the experiments. DL and YZ performed the experiments. WZ and JG analyzed the data. LG drafted the manuscript. XL reviewed and edited the manuscript. All authors 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.

Abbreviations:

NO

nitric oxide

ECH

echinacoside

AR

androgen receptor

ER

estrogen receptor

PI3K

phosphatidylinositol 3-kinase

Akt

serine/threonine-protein Akt kinase (protein kinase B)

eNOS

endothelial isoform of nitric oxide synthase

siRNA

small interfering RNA

cGMP

cyclic guanosine monophosphate

PKG

cGMP-dependent protein kinase

HUVEC

human umbilical vein endothelial cell

ECM

endothelial cell medium

FBS

fetal bovine serum

ECGS

endothelial cell growth supplement

RT-qPCR

reverse transcription-quantitative PCR

DMSO

dimethylsulfoxide

Acknowledgments

Not applicable.

References

1 

Chinese Pharmacopoeia Commission: Cistanche Herba. Pharmacopoeia of the People's Repulic of China 1. Chin. Med. Sci. Press; Beijing: pp. 1352015

2 

Jiang Z, Wang J, Li X and Zhang X: Echinacoside and Cistanche tubulosa (Schenk) R. wight ameliorate bisphenol A-induced testicular and sperm damage in rats through gonad axis regulated steroidogenic enzymes. J Ethnopharmacol. 193:321–328. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Liu J, Yang L, Dong Y, Zhang B and Ma X: Echinacoside, an inestimable natural product in treatment of neurological and other disorders. Molecules. 23:pp. piiE12132018, View Article : Google Scholar

4 

Yoshikawa M, Matsuda H, Morikawa T, Xie H, Nakamura S and Muraoka O: Phenylethanoid oligoglycosides and acylated oligosugars with vasorelaxant activity from Cistanche tubulosa. Bioorg Med Chem. 14:7468–7475. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Arnold WP, Mittal CK, Katsuki S and Murad F: Nitric oxide activates guanylate cyclase and increases guanosine 3′:5′-cyclic monophosphate levels in various tissue preparations. Proc Natl Acad Sci USA. 74:3203–3207. 1977. View Article : Google Scholar

6 

Gai XY, Tang F, Ma J, Zeng KW, Wang SL, Wang YP, Wuren TN, Lu DX, Zhou Y and Ge RL: Antiproliferative effect of echinaco-side on rat pulmonary artery smooth muscle cells under hypoxia. J Pharmacol Sci. 126:155–163. 2014. View Article : Google Scholar

7 

He WJ, Fang TH, Ma X, Zhang K, Ma ZZ and Tu PF: Echinacoside elicits endothelium-dependent relaxation in rat aortic rings via an NO-cGMP pathway. Planta Med. 75:1400–1404. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Gai XY, Wei YH, Zhang W, Wuren TN, Wang YP, Li ZQ, Liu S, Ma L, Lu DX, Zhou Y and Ge RL: Echinacoside induces rat pulmonary artery vasorelaxation by opening the NO-cGMP-PKG-BKCa channels and reducing intracellular Ca2+ levels. Acta Pharmacol Sin. 36:587–596. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Maruhashi T, Kihara Y and Higashi Y: Assessment of endothelium-independent vasodilation: From methodology to clinical perspectives. J Hypertens. 36:1460–1467. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Ahmad KA, Ze H, Chen J, Khan FU, Chen X, Xu J and Ding Q: The protective effects of a novel synthetic β-elemene derivative on human umbilical vein endothelial cells against oxidative stress-induced injury: Involvement of antioxidation and PI3k/Akt/eNOS/NO signaling pathways. Biomed Pharmacother. 106:1734–1741. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Yu J, Akishita M, Eto M, Koizumi H, Hashimoto R, Ogawa S, Tanaka K, Ouchi Y and Okabe T: Src kinase-mediates androgen receptor-dependent non-genomic activation of signaling cascade leading to endothelial nitric oxide synthase. Biochem Bioph Res Commun. 424:538–543. 2012. View Article : Google Scholar

12 

Yu J, Akishita M, Eto M, Ogawa S, Son B, Kato S, Ouchi Y and Okabe T: Androgen receptor-dependent activation of endothelial nitric oxide synthase in vascular endothelial cells: Role of phosphatidylinositol 3-kinase/Akt pathway. Endocrinology. 151:1822–1828. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Pittarella P, Squarzanti DF, Molinari C, Invernizzi M, Uberti F and Reno F: NO-dependent proliferation and migration induced by vitamin D in HUVEC. J Steroid Biochem. 149:35–42. 2015. View Article : Google Scholar

14 

He Y, Luan Z, Fu X and Xu X: Overexpression of uncoupling protein 2 inhibits the high glucose-induced apoptosis of human umbilical vein endothelial cells. Int J Mol Med. 37:631–638. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Xiao-Hong D, Chang-Qin X, Jian-Hua H, Wen-Jiang Z and Bing S: Icariin delays homocysteine-induced endothelial cellular senescence involving activation of the PI3K/AKT-eNOS signaling pathway. Pharm Biol. 51:433–440. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Smith PK, Krohn RI, Hermanson GT, Mallia AK, Gartner FH, Provenzano MD, Fujimoto EK, Goeke NM, Olson BJ and Klenk DC: Measurement of protein using bicinchoninic acid. Anal Biochem. 150:76–85. 1985. View Article : Google Scholar : PubMed/NCBI

17 

Gu L, Zhong X, Lian D, Zheng Y, Wang H and Liu X: Triterpenoid biosynthesis and the transcriptional response elicited by nitric oxide in submerged fermenting Ganoderma lucidum. Process Biochem. 60:19–26. 2017. View Article : Google Scholar

18 

Ellinsworth DC, Sandow SL, Shukla N, Liu Y, Jeremy JY and Gutterman DD: Endothelium-derived hyperpolarization and coronary vasodilation: Diverse and integrated roles of epoxyeicosatrienoic acids, hydrogen peroxide, and gap junctions. Microcirculation. 23:15–32. 2016. View Article : Google Scholar :

19 

Freed JK and Gutterman DD: Communication is key: Mechanisms of intercellular signaling in vasodilation. J Cardiovasc Pharm. 69:264–272. 2017. View Article : Google Scholar

20 

Quillon A, Fromy B and Debret R: Endothelium microenvironment sensing leading to nitric oxide mediated vasodilation: A review of nervous and biomechanical signals. Nitric Oxide. 45:20–26. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Torres-Estay V, Carreno DV, Francisco IF, Sotomayor P, Godoy AS and Smith GJ: Androgen receptor in human endothelial cells. J Endocrinol. 224:131–137. 2015. View Article : Google Scholar

22 

Deng Q, Zhang Z, Wu Y, Yu WY, Zhang J, Jiang ZM, Zhang Y, Liang H and Gui YT: Non-genomic action of androgens is mediated by rapid phosphorylation and regulation of androgen receptor trafficking. Cell Physiol Biochem. 43:223–236. 2017. View Article : Google Scholar : PubMed/NCBI

23 

de Oliveira TS, de Oliveira LM, de Oliveira LP, Costa RMD, Tostes RC, Georg RC, Costa EA, Lobato NS, Filgueira FP and Ghedini PC: Activation of PI3K/Akt pathway mediated by estrogen receptors accounts for estrone-induced vascular activation of cGMP signaling. Vascul Pharmacol. 110:42–48. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Li F, Yang X, Yang Y, Guo C, Zhang C, Yang Z and Li P: Antiosteoporotic activity of echinacoside in ovariectomized rats. Phytomedicine. 20:549–557. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Zeng KW, Liao LX, Wan YJ, Jiang Y and Tu PF: Pharmacological targets identification and efficacy analysis of phenylethanoid glycosides from Cistanches Herba based on 'target fishing' strategy. Chin Tradit Herbal Drugs. 49:173–178. 2018.

26 

Jiang Z, Zhou B, Li X, Kirby GM and Zhang X: Echinacoside increases sperm quantity in rats by targeting the hypothalamic androgen receptor. Sci Rep. 8:3839–3850. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Coffer PJ, Jin J and Woodgett JR: Protein kinase B (c-Akt): A multifunctional mediator of phosphatidylinositol 3-kinase activation. Biochem J. 335:1–13. 1998. View Article : Google Scholar : PubMed/NCBI

28 

Wang S, Zheng G, Tian S, Zhang Y, Shen L, Pak Y, Shen Y and Qian J: Echinacoside improves hematopoietic function in 5-FU-induced myelosuppression mice. Life Sci. 123:86–92. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Chen M, Wang X, Hu B, Zhou J, Wang X, Wei W and Zhou H: Protective effects of echinacoside against anoxia/reperfusion injury in H9c2 cells via up-regulating p-AKT and SLC8A3. Biomed Pharmacother. 104:52–59. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Abeyrathna P and Su Y: The critical role of Akt in cardiovascular function. Vascul Pharmacol. 74:38–48. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2020
Volume 45 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gu L, Lian D, Zheng Y, Zhou W, Gu J and Liu X: Echinacoside‑induced nitric oxide production in endothelial cells: Roles of androgen receptor and the PI3K‑Akt pathway. Int J Mol Med 45: 1195-1202, 2020
APA
Gu, L., Lian, D., Zheng, Y., Zhou, W., Gu, J., & Liu, X. (2020). Echinacoside‑induced nitric oxide production in endothelial cells: Roles of androgen receptor and the PI3K‑Akt pathway. International Journal of Molecular Medicine, 45, 1195-1202. https://doi.org/10.3892/ijmm.2020.4476
MLA
Gu, L., Lian, D., Zheng, Y., Zhou, W., Gu, J., Liu, X."Echinacoside‑induced nitric oxide production in endothelial cells: Roles of androgen receptor and the PI3K‑Akt pathway". International Journal of Molecular Medicine 45.4 (2020): 1195-1202.
Chicago
Gu, L., Lian, D., Zheng, Y., Zhou, W., Gu, J., Liu, X."Echinacoside‑induced nitric oxide production in endothelial cells: Roles of androgen receptor and the PI3K‑Akt pathway". International Journal of Molecular Medicine 45, no. 4 (2020): 1195-1202. https://doi.org/10.3892/ijmm.2020.4476