MicroRNA-138 inhibits hypoxia-induced proliferation of endothelial progenitor cells via inhibition of HIF-1α-mediated MAPK and AKT signaling

Retraction in: /10.3892/etm.2021.10428

  • Authors:
    • Wei Zhou
    • Weimin Zhou
    • Qingzhong Zeng
    • Jixin Xiong
  • View Affiliations

  • Published online on: January 27, 2017     https://doi.org/10.3892/etm.2017.4091
  • Pages: 1017-1024
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endothelial progenitor cells (EPCs) participate in angiogenesis by differentiating into endothelial cells (ECs) and may be developed to treat ischemia/reperfusion injury. MicroRNAs (miRs) are a type of non‑coding RNA that are 18-25 nucleotides in length and serve a role in angiogenesis. It has been demonstrated that miR‑138 regulates hypoxia-induced EC dysfunction. However, to the best of our knowledge, the exact role of miR‑138 in the regulation of hypoxia-induced EPCs has not previously been reported. In the present study, data collected from an MTT assay indicated that hypoxia treatment enhanced EPC proliferation, which was accompanied by an upregulation of hypoxia‑inducible factor 1α (HIF-1α) expression. miR‑138 overexpression inhibited hypoxia‑induced EPC proliferation and induced cell cycle arrest at the G1 stage. A mechanistic investigation revealed that miR‑138 negatively regulated HIF‑1α protein levels but did not affect HIF‑1α mRNA levels in EPCs. Moreover, results from a dual luciferase reporter assay demonstrated that HIF‑1α was a direct target of miR‑138 in EPCs. Furthermore, upregulation of miR‑138 suppressed the hypoxia‑induced upregulation of HIF‑1α. Downstream factors of HIF‑1α were also investigated and it was observed that the upregulation of miR‑138 inhibited the hypoxia‑induced upregulation of vascular endothelial growth factor, as well as the activity of mitogen-activated protein kinase and AKT signaling in EPCs. In summary, the present study suggested that miR‑138 inhibits hypoxia-induced EPC proliferation, possibly by inhibiting HIF-1α-mediated signaling.

Introduction

It has been demonstrated that endothelial progenitor cells (EPCs) are strongly associated with angiogenesis due to their differentiation into endothelial cells (ECs) (1). During hypoxia, EPCs are mobilized from the bone marrow into the peripheral blood and subsequently migrate to hypoxic tissues (2). Furthermore, hypoxia inhibits cellular senescence to restore the therapeutic potential of EPCs from elderly individuals (3). Therefore, EPCs may be developed as a promising novel treatment for ischemia/reperfusion injury.

MicroRNAs (miRs), are a type of non-coding RNA that are 18–25 nucleotides long and can induce mRNA degradation or suppress protein translation by binding to the 3′-untranslated region (3′UTR) of mRNA of specific genes (4). Previous studies have shown that miRs are involved in angiogenesis (5,6). It has been demonstrated that miR-487b promotes proliferation, migration, invasion and tube formation in human umbilical vein ECs by regulating thrombospondin 1 (7). miR-214, which is upregulated in heart failure patients, was found to serve a suppressive role in the regulation of Xbox-binding protein 1-mediated EC angiogenesis (8). In addition, it has been observed that miR-138 regulates hypoxia-induced EC dysfunction by targeting S100A1 (9). Following stimulation with vascular endothelial growth factor (VEGF), it was observed that ECs overexpressing miR-138 exhibited reduced tube formation and expressed lower levels of nitric oxide (NO) (9). Furthermore, miR-138 is involved in hypoxic pulmonary vascular remodeling by targeting macrophage stimulating protein 1 (10). However, to the best of our knowledge, there have been no studies investigating the exact role of miR-138 in the regulation of hypoxia-induced EPCs.

Hypoxia-inducible factor-1α (HIF-1α) is involved in the hypoxia-induced proliferation, migration and differentiation of EPCs (11), and it has been identified as a target gene of miR-138 in human cancer cells (12,13). Yeh et al (12) found that miR-138 suppressed ovarian cancer cell invasion and metastasis by targeting HIF-1α (12). Song et al (13) reported that miR-138 induced apoptosis and reduced the migration of clear cell renal cell carcinoma cells by inhibiting HIF-1α. However, it has remained elusive whether HIF-1α is directly targeted by miR-138 in EPCs.

Therefore, the primary aim of the present study was to investigate the role of miR-138 in the regulation of hypoxia-induced EPC proliferation. In addition, the underlying mechanism was investigated, focusing on HIF-1α-mediated signaling pathways.

Materials and methods

Reagents

EGM-2 medium, fetal bovine serum (FBS), Lipofectamine® 2000, MTT, dimethyl sulfoxide (DMSO), TRIzol® reagent and a miRNA Reverse Transcription kit were purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). The All-in-One™ miRNA qRT-PCR Detection kit was purchased from GeneCopoeia, Inc. (Rockville, MD, USA). miR-138 mimics, miR-138 inhibitor and scramble miR mimics were purchased from Genechem, Co., Ltd. (Shanghai, China). Rabbit anti-HIF-1α polyclonal antibody (ab69836), rabbit anti-VEGF polyclonal antibody (ab69479), rabbit anti-phosphorylated (p)-p38 mitogen-activated protein kinase (MAPK) polyclonal antibody (ab60999), rabbit anti-p38 MAPK polyclonal antibody (ab31828), rabbit anti-p-AKT monoclonal antibody (ab81283), rabbit anti-AKT monoclonal antibody (ab32505), rabbit anti-glyceraldehyde-3-phosphate dehydrogenase (GAPDH) polyclonal antibody (ab9485) and mouse monoclonal 2A9 Anti-Rabbit IgG heavy chain (HRP) (ab99702) were all purchased from Abcam (Cambridge, MA, USA). The Pierce ECL Western Blotting KIT and polyvinylidene difluoride membrane were purchased from Pierce; Thermo Fisher Scientific, Inc. pRL-TK plasmid, lysis buffer and the Dual-Luciferase® Reporter assay system were purchased from Promega Corp. (Madison, WI, USA).

Cell isolation and culture

An umbilical cord (UBC) was donated by a patient that had just given birth in the Second Affiliated Hospital of Nangchang University (Nanchang, China) and informed consent was obtained. The UBC was homogenized and immersed in Dulbecco's phosphate-buffered saline (DPBS; Thermo Fisher Scientific, Inc.) at a ratio of 1:1 and overlaid onto 1.077 g/ml Ficoll (GE Healthcare Bio-Sciences, Pittsburgh, PA, USA), followed by centrifugation at 4°C for 30 min at 400 × g. UBC-monocytes were collected and seeded into tissue culture plates coated with fibronectin (EMD Millipore, Billerica, MA, USA) and cultured in EGM-2 (Lonza Group, Basel, Switzerland) at 37°C, in a humidified incubator with 5% CO2. The culture medium was changed every 2 days until EPC colonies appeared. Typically, colonies appeared between day 5 and 10, and were passaged at sub-confluence.

Hypoxia treatment of EPCs

EPCs (5×107) were cultured in serum-free EGM-2 at 37°C and 5% CO2 for 12 h prior to hypoxia treatment. Hypoxia treatment consisted of EPCs undergoing culture in EGM-2 containing 20% fetal bovine serum at 37°C, in a humidified incubator containing 1% O2, 94% N2 and 5% CO2. Hypoxia treatment lasted for 3 h prior to subsequent analyses.

Cell proliferation assay

An MTT assay was performed to determine cell proliferation. In brief, 10 mg/ml MTT was added to the medium. Following 4 h of incubation, the reaction was terminated by removal of the supernatant and addition of 100 µl DMSO to dissolve the formazan product. After 30 min, the optical density of each well was measured at 570 nm using an ELx808 absorbance microplate reader (Bio-Tek Instruments, Inc., Winooski, VT, USA).

Cell cycle analysis

EPCs were resuspended in 70% ethanol. Following fixation overnight at −20°C, cells were pelleted, washed twice in 1X PBS with 3% bovine serum albumin (BSA; Thermo Fisher Scientific, Inc.) and pelleted at 1,000 × g for 5 min at 4°C. Subsequently, EPCs were resuspended and incubated for 30 min at room temperature in propidium iodide (PI) staining buffer containing 3% BSA, 40 µg/ml PI and 0.2 mg/ml RNase in 1X PBS. DNA content analyses were performed using flow cytometry (FACSCalibur; BD Biosciences, San Jose, CA, USA).

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

TRIzol reagent was used to extract total RNA from cells following the manufacturer's instructions. Total RNA was transcribed into complementary (c)DNA using the miRNA Reverse Transcription kit following the manufacturer's protocols. Reverse transcription was performed at 16°C for 30 min, followed by an incubation step at 42°C for 30 min and enzyme inactivation at 85°C for 5 min. miRNA levels were determined using the All-in-One™ miRNA qRT-PCR Detection kit on an ABI 7500 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.), in accordance with the manufacturer's instructions. For qPCR, 0.33 µl cDNA solution, 10 µl 1X TaqMan universal PCR master mix, 2 µl 1X gene-specific primer (Thermo Fisher Scientific, Inc.) and 7.67 µl H2O were mixed to obtain a final reaction volume of 20 µl. The reaction conditions were 95°C for 5 min, and 40 cycles of denaturation at 95°C for 15 sec and annealing/elongation step at 60°C for 60 sec. The U6 gene was used as an internal reference. The relative expression was analyzed by the 2−ΔΔCq method (14).

Transfection

Transfection was performed using Lipofectamine® 2000. In brief, EPCs were cultured to 70% confluence and re-suspended in EGM-2 medium without FBS. miR-138 mimics, miR-138 inhibitor and Lipofectamine® 2000 were all diluted with serum-free medium. The diluted Lipofectamine® 2000 was added to the diluted miR-138 mimics or miR-138 inhibitor and incubated for 20 min at room temperature, prior to addition to the cell suspension. Following 6 h of incubation at 37°C in 5% CO2, the medium was replaced by EGM-2 medium containing 20% FBS and EPCs were cultured for 24 h prior to further assays.

Western blot analysis

EPCs were solubilized in cold radioimmunoprecipitation assay lysis buffer (Thermo Fisher Scientific, Inc.). Proteins were separated by 10% SDS-PAGE and transferred onto a polyvinylidene difluoride membrane. The membrane was incubated with Tris-buffered saline containing Tween-20 and 5% skimmed milk at 4°C overnight. Subsequently, the membrane was incubated for 3 h at room temperature with rabbit anti-HIF-1α monoclonal antibody (1:50), rabbit anti-VEGF monoclonal antibody (1:100), rabbit anti-p-p38 MAPK monoclonal antibody (1:50), rabbit anti-p38 MAPK monoclonal antibody (1:50), rabbit anti-p-AKT monoclonal antibody (1:50), rabbit anti-AKT monoclonal antibody (1:50) and mouse anti-GAPDH monoclonal antibody (1:100). Following washing with PBS containing Tween-20 for three times, the membrane was incubated with the mouse anti-rabbit secondary antibody (1:5,000) at room temperature for 1 h. The ECL kit was used to detect chemiluminescence. Relative protein expression was determined using Image-Pro plus software 6.0 (Media Cybernetics, Inc., Rockville, MD, USA), represented as the density ratio vs. GAPDH.

Bioinformatics analysis

Targetscan 3.1 online software (http://www.targetscan.org) was used to predict the putative target genes of miR-138. The species was ‘Human’ and ‘miR-138’ was entered as the miR name.

Luciferase reporter gene assay

The wild-type (WT) and mutant (Mut) 3′UTR sequences of HIF-1α were designed by GeneCopoeia, Inc. and were inserted into a dual luciferase reporter vector (Promega Corp.). EPCs (5×107) were seeded into 24-well plates and co-transfected with 200 ng pMIR-HIF-1α or pMIR-HIF-1α-Mut vector and 100 ng miR-138 mimic or scramble miR mimic, and the pRL-TK plasmid for internal normalization. Cells were harvested after 36 h and lysed using the lysis buffer. A luciferase reporter gene assay was perfored using the Dual-Luciferase Reporter assay system, following the manufacturer's instructions.

Statistical analysis

Values are expressed as the mean ± standard deviation of three independent experiments. Statistical analysis of differences was performed using Student's t-test with SPSS version 17 software (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Hypoxia treatment enhances EPC proliferation and HIF-1α expression

The effect of hypoxia treatment on the proliferation capacity of EPCs was examined. An MTT assay was performed to determine EPC proliferation under hypoxia or normoxia. The proliferation of EPCs was significantly upregulated following culture under hypoxia for 12, 24, 48, 72 and 96 h, compared with the proliferation of cells cultured under normoxia (P<0.01; Fig. 1A), demonstrating that hypoxia enhances EPC proliferation. It has been suggested that HIF-1α is associated with EPC proliferation (11); therefore, the levels of HIF-1α expression in EPCs cultured under hypoxic or normoxic conditions were determined. Hypoxia was found to significantly enhance the levels of HIF-1α mRNA and protein in EPCs compared with the control group (P<0.01; Fig. 1B and C).

miR-138 inhibits hypoxia-induced EPC proliferation through inducing cell cycle arrest

The role of miR-138 in regulating hypoxia-induced EPC proliferation was investigated. EPCs were transfected with miR-138 mimics or scramble miR mimics as a negative control (miR-NC). Following transfection, RT-qPCR was performed to determine the expression of miR-138 in each group. The results demonstrated that miR-138 expression was significantly increased in EPCs transfected with miR-138 mimics, compared with those transfected with the miR-NC and the control group, indicating that the transfection was successful (P<0.01; Fig. 2A). EPCs in each group were cultured under hypoxic conditions for 12, 24, 48, 72 and 96 h. The results indicated that proliferation was significantly decreased in EPCs overexpressing miR-138 compared with that of the control group (Fig. 2B). These findings indicated that upregulation of miR-138 inhibits hypoxia-induced EPC proliferation.

Subsequently, the cell cycle distribution of EPCs in each group was examined. The results showed that the percentage of cells in the G1 stage was significantly higher in the miR-138 group, compared with that in the miR-NC and control groups (P<0.01; Fig. 3). This suggested that overexpression of miR-138 induces cell cycle arrest at G1, resulting in the decreased proliferation of EPCs.

miR-138 negatively mediates the expression of HIF-1α in EPCs

It has been demonstrated that the expression of HIF-1α is negatively regulated by miR-138 in ovarian cancer cells and clear cell renal cell carcinoma cells (12,13). However, it remains elusive whether miR-138 affects the expression of HIF-1α. Therefore, in the present study, EPCs were transfected with miR-138 mimics or miR-138 inhibitors. Following transfection, RT-qPCR was performed to determine the level of miR-138 expression in each group. Compared with the control group, the expression of miR-138 was significantly increased in EPCs transfected with miR-138 mimics but reduced in EPCs transfected with miR-138 inhibitor, compared with the control (P<0.01; Fig. 4A). Subsequently, RT-qPCR and western blotting analysis were performed to examine HIF-1α mRNA and protein expression, respectively, in each group. Neither miR-138 overexpression or miR-138 knockdown affected HIF-1α mRNA expression in EPCs (Fig. 4B). However, compared with the control group, overexpression of miR-138 led to a significant decrease in HIF-1α protein levels, while knockdown of miR-138 significantly increased HIF-1α protein levels (P<0.01; Fig. 4C). These findings indicated that miR-138 downregulated the expression of HIF-1α in EPCs at the post-transcriptional level.

miR-138 directly targets HIF-1α in EPCs

Bioinformatical analysis using Targetscan indicated that HIF-1α was a potential target gene of miR-138. To confirm this prediction, WT or Mut 3′UTR sequences of HIF-1α (Fig. 5A) were respectively inserted into the dual luciferase reporter vector (Fig. 5B). Subsequently, miR-138 mimic/miR-NC and the reporter plasmids driven by the WT/MUT sequence from the 3′UTR of HIF-1α were co-transfected into EPCs. The results of the dual luciferase reporter assay demonstrated that luciferase activity was significantly repressed in the cells co-transfected with miR-138 mimic and plasmid containing the WT 3′UTR sequence of HIF-1α, compared with that in the control group (P<0.01; Fig. 5C). However, the miR-138-mediated repression of luciferase activity was abolished in the group transfected with the reporter plasmid driven by the MUT 3′UTR sequence of HIF-1α (Fig. 5C). These results demonstrated that miR-138 directly targets the HIF-1α 3′UTR in EPCs.

miR-138 suppresses hypoxia-induced HIF-1α expression in EPCs

The effect of miR-138 upregulation on hypoxia-induced HIF-1α expression in EPCs was assessed. The expression of HIF-1α mRNA and protein was lower in miR-138-overexpressing EPCs cultured under hypoxia, when compared with non-transfected EPCs and EPCs transfected with miR-NC cultured under hypoxia (P<0.01; Fig. 6). These findings indicated that miR-138 suppresses hypoxia-induced HIF-1α upregulation in EPCs.

miR-138 inhibits hypoxia-induced upregulation of VEGF expression as well as MAPK and AKT signaling in EPCs

It has been demonstrated that VEGF, MAPK and AKT signaling are associated with EPC proliferation (15). Therefore, the effect of miR-138 overexpression on the expression of VEGF, as well as the activity of p38 MAPK and AKT signaling in EPCs cultured under hypoxia, was investigated. Hypoxia treatment significantly enhanced the expression of VEGF protein in EPCs compared with EPCs cultured under normoxic conditions; however, transfection with miR-138 mimics significantly reduced the expression of VEGF protein in EPCs cultured under hypoxia compared with untransfected cells cultured under hypoxia (P<0.01; Fig. 7A). Hypoxia treatment significantly enhanced the levels of phosphorylated p38 MAPK and AKT protein in EPCs, when compared with those in EPCs cultured under normoxia (P<0.01; Fig. 7B and C), indicating that the activity of p38 MAPK and AKT was upregulated. However, overexpression of miR-138 inhibited the hypoxia-induced upregulation of phosphorylated protein levels of p38 MAPK and AKT in EPCs (P<0.01; Fig. 7B and C). Taken together, these results suggested that miR-138 inhibits the hypoxia-induced upregulation of VEGF expression as well as MAPK and AKT signaling.

Discussion

It has been demonstrated that ischemia/reperfusion injury is strongly associated with fatal diseases, including stroke and coronary atherosclerosis caused by myocardial infarction (16,17). In addition, oxidative stress-induced endothelial dysfunction is involved in ischemia/reperfusion injury (18). EPCs are crucial in the angiogenesis of ECs and it has been suggested that they serve an important role in angiogenesis occurring under hypoxia/ischemia (19,20). Hypoxia/ischemia stimulates EPCs to migrate from the bone marrow into the peripheral blood, adhere to the endothelium at sites of hypoxia/ischemia and participate in new vessel formation by differentiating into ECs (21). miR-138 may be involved in the regulation of hypoxia-induced EC dysfunction (22); however, to the best of our knowledge, the exact role of miR-138 in mediating EPC proliferation under hypoxia has not been studied. The present study demonstrated that miR-138 significantly suppressed the hypoxia-induced proliferation of EPCs, possibly by inducing cell cycle arrest at the G1 stage. A mechanistic investigation revealed that miR-138 suppressed the hypoxia-induced upregulation of HIF-1α and VEGF expression, as well as the activity of MAPK and AKT signaling in EPCs.

Previous studies have demonstrated that hypoxia inhibits the senescence of EPCs in elderly individuals (3) and stimulates ECs to secrete macrophage migration inhibitory factor, which enhances the recruitment and migration of EPCs to hypoxic tissues (2). The present study revealed that hypoxia induced EPC proliferation. Nishimura et al (23) reported that hypoxia induced the proliferation of tissue-resident EPCs in the lung. Furthermore, Sen et al (22) indicated that exposure to pro-inflammatory cytokines, including angiotensin II, endothelin-1 and tumor necrosis factor, may result in EC dysfunction and the downregulation of S100A1 expression by inducing miR-138 in a manner dependent on the stabilization of HIF-1α. This suggests that miR-138 is involved in EC dysfunction (22). The results of the present study showed that miR-138 overexpression significantly suppressed hypoxia-induced EPC proliferation.

HIF-1α is a transcriptional factor that acts as a determinant of oxygen-dependent gene regulation in angiogenesis and participates in the regulation of biological processes in EPCs (11,24). Jiang et al (11) demonstrated that HIF-1α knockdown suppressed the differentiation of EPCs into ECs, and another study showed that HIF-1α overexpression promoted the differentiation of EPCs into ECs (25). Furthermore, knockdown of HIF-1α expression was shown to inhibit the expression of VEGF, VEGF receptor 2, endothelial nitric oxide synthase, as well as nitric oxide production (11,24). The present study showed that HIF-1α expression was significantly upregulated in EPCs cultured under hypoxia. Furthermore, it was observed that miR-138 negatively mediates levels of HIF-1α protein in EPCs, and overexpression of miR-138 was found to decrease hypoxia-induced HIF-1α expression in EPCs. Jiang et al (26) reported that HIF-1α overexpression promoted hypoxia-induced EPC differentiation, proliferation and migration. Therefore, the inhibitory effect of miR-138 overexpression on hypoxia-induced EPCs proliferation may occur by inhibiting the hypoxia-induced upregulation of HIF-1α expression.

It is well established that MAPK signaling is involved in regulating cellular survival, proliferation, differentiation and migration (27), and that MAPK signaling serves a key role in regulating the proliferation of EPCs (28). In addition, AKT signaling is also involved in hypoxia-induced EPC proliferation (28,29). Qiu et al (28) demonstrated that granulocyte-macrophage colony-stimulating factor induces cyclin D1 expression and the proliferation of EPCs via AKT and MAPK signaling. Dai et al (30) showed that hypoxia may protect against serum withdrawal-induced EPC apoptosis, at least in part, via AKT pathway activation (30). Yu et al (29) demonstrated that activation of the liver X receptor enhanced the proliferation and migration of EPCs and promoted vascular repair by activating the AKT signaling pathway. In the present study, it was demonstrated that miR-138 overexpression suppressed the hypoxia-induced upregulation of AKT and MAPK in EPCs. Therefore, the inhibition of AKT and MAPK signaling may be involved in the suppressive effect of miR-138 overexpression on hypoxia-induced EPC proliferation.

In conclusion, miR-138 may serve an inhibitory role in the regulation of hypoxia-induced EPC proliferation, possibly by directly suppressing the expression of HIF-1α protein. In addition, MAPK and AKT signaling may be involved in the miR-138-mediated inhibition of hypoxia-induced EPC proliferation. Therefore, miR-138 may become a potential target for the treatment of ischemia/reperfusion injury.

References

1 

Endemann DH and Schiffrin EL: Endothelial dysfunction. J Am Soc Nephrol. 15:1983–1992. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Simons D, Grieb G, Hristov M, Pallua N, Weber C, Bernhagen J and Steffens G: Hypoxia-induced endothelial secretion of macrophage migration inhibitory factor and role in endothelial progenitor cell recruitment. J Cell Mol Med. 15:668–678. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Lee SH, Lee JH, Yoo SY, Hur J, Kim HS and Kwon SM: Hypoxia inhibits cellular senescence to restore the therapeutic potential of old human endothelial progenitor cells via the hypoxia-inducible factor-1α-TWIST-p21 axis. Arterioscler Thromb Vasc Biol. 33:2407–2414. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Bartoszewska S, Kochan K, Piotrowski A, Kamysz W, Ochocka RJ, Collawn JF and Bartoszewski R: The hypoxia-inducible miR-429 regulates hypoxia-inducible factor-1α expression in human endothelial cells through a negative feedback loop. FASEB J. 29:1467–1479. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Zhou S, Zhang P, Liang P and Huang X: The expression of miR-125b regulates angiogenesis during the recovery of heat-denatured HUVECs. Burns. 41:803–811. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Feng N, Wang Z, Zhang Z, He X, Wang C and Zhang L: miR-487b promotes human umbilical vein endothelial cell proliferation, migration, invasion and tube formation through regulating THBS1. Neurosci Lett. 591:1–7. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Duan Q, Yang L, Gong W, Chaugai S, Wang F, Chen C, Wang P, Zou MH and Wang DW: MicroRNA-214 is up-regulated in heart failure patients and suppresses XBP1-mediated endothelial cells angiogenesis. J Cell Physiol. 230:1964–1973. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Sen A, Ren S, Lerchenmüller C, Sun J, Weiss N, Most P and Peppel K: MicroRNA-138 regulates hypoxia-induced endothelial cell dysfunction by targeting S100A1. PLoS One. 8:e786842013. View Article : Google Scholar : PubMed/NCBI

10 

Li S, Ran Y, Zhang D, Chen J, Li S and Zhu D: MicroRNA-138 plays a role in hypoxic pulmonary vascular remodelling by targeting Mst1. Biochem J. 452:281–291. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Jiang M, Wang CQ, Wang BY and Huang DJ: Inhibitory effect of siRNA targeting HIF-1alpha on differentiation of peripheral blood endothelial progenitor cells. Ai Zheng. 24:1293–1300. 2005.(In Chinese). PubMed/NCBI

12 

Yeh YM, Chuang CM, Chao KC and Wang LH: MicroRNA-138 suppresses ovarian cancer cell invasion and metastasis by targeting SOX4 and HIF-1α. Int J Cancer. 133:867–878. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Song T, Zhang X, Wang C, Wu Y, Cai W, Gao J and Hong B: MiR-138 suppresses expression of hypoxia-inducible factor 1α (HIF-1α) in clear cell renal cell carcinoma 786-O cells. Asian Pac J Cancer Prev. 12:1307–1311. 2011.PubMed/NCBI

14 

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

15 

Tang Y, Huang B, Sun L, Peng X, Chen X and Zou X: Ginkgolide B promotes proliferation and functional activities of bone marrow-derived endothelial progenitor cells: Involvement of Akt/eNOS and MAPK/p38 signaling pathways. Eur Cell Mater. 21:459–469. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Ganguly R, Lytwyn MS and Pierce GN: Differential effects of trans and polyunsaturated fatty acids on ischemia/reperfusion injury and its associated cardiovascular disease states. Curr Pharm Des. 19:6858–6863. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Kalogeris T, Baines CP, Krenz M and Korthuis RJ: Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 298:229–317. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Yang D, Zhang P, Wang T, Gao L, Qiao Z, Liang Y and Yu B: SalA attenuates ischemia/reperfusion-induced endothelial barrier dysfunction via down-regulation of VLDL receptor expression. Cell Physiol Biochem. 33:747–757. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Xu BY, Xiang MX and Wang JA: Endothelial progenitor cells and in-stent restenosis. Curr Stem Cell Res Ther. 10:364–371. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Yoder MC: Human endothelial progenitor cells. Cold Spring Harb Perspect Med. 2:a0066922012. View Article : Google Scholar : PubMed/NCBI

21 

Machalinska A: The role of circulating endothelial progenitor cells in the development of vascular retinal diseases. Klin Oczna. 115:158–162. 2013.(In Polish). PubMed/NCBI

22 

Sen A, Most P and Peppel K: Induction of microRNA-138 by pro-inflammatory cytokines causes endothelial cell dysfunction. FEBS Lett. 588:906–914. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Nishimura R, Nishiwaki T, Kawasaki T, Sekine A, Suda R, Urushibara T, Suzuki T, Takayanagi S, Terada J, Sakao S and Tatsumi K: Hypoxia-induced proliferation of tissue-resident endothelial progenitor cells in the lung. Am J Physiol Lung Cell Mol Physiol. 308:L746–L758. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Jiang M, Wang B, Wang C, He B, Fan H, Guo TB, Shao Q, Gao L and Liu Y: Inhibition of hypoxia-inducible factor-1alpha and endothelial progenitor cell differentiation by adenoviral transfer of small interfering RNA in vitro. J Vasc Res. 43:511–521. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Jiang M, Wang CQ, Wang BY, He B, Shao Q and Huang DJ: Overexpression of hypoxia inducible factor-1alpha (HIF-1alpha) promotes the differentiation of endothelial progenitor cell ex vivo. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 14:565–570. 2006.(In Chinese). PubMed/NCBI

26 

Jiang M, Wang B, Wang C, He B, Fan H, Guo TB, Shao Q, Gao L and Liu Y: Angiogenesis by transplantation of HIF-1 alpha modified EPCs into ischemic limbs. J Cell Biochem. 103:321–334. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Kyriakis JM and Avruch J: Mammalian MAPK signal transduction pathways activated by stress and inflammation: A 10-year update. Physiol Rev. 92:689–737. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Qiu C, Xie Q, Zhang D, Chen Q, Hu J and Xu L: GM-CSF induces cyclin D1 expression and proliferation of endothelial progenitor cells via PI3K and MAPK signaling. Cell Physiol Biochem. 33:784–795. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Yu J, Wang Q, Wang H, Lu W, Li W, Qin Z and Huang L: Activation of liver X receptor enhances the proliferation and migration of endothelial progenitor cells and promotes vascular repair through PI3K/Akt/eNOS signaling pathway activation. Vascul Pharmacol. 62:150–161. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Dai T, Zheng H and Fu GS: Hypoxia confers protection against apoptosis via the PI3K/Akt pathway in endothelial progenitor cells. Acta Pharmacol Sin. 29:1425–1431. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2017
Volume 13 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
Zhou W, Zhou W, Zeng Q and Xiong J: MicroRNA-138 inhibits hypoxia-induced proliferation of endothelial progenitor cells via inhibition of HIF-1α-mediated MAPK and AKT signaling Retraction in /10.3892/etm.2021.10428. Exp Ther Med 13: 1017-1024, 2017
APA
Zhou, W., Zhou, W., Zeng, Q., & Xiong, J. (2017). MicroRNA-138 inhibits hypoxia-induced proliferation of endothelial progenitor cells via inhibition of HIF-1α-mediated MAPK and AKT signaling Retraction in /10.3892/etm.2021.10428. Experimental and Therapeutic Medicine, 13, 1017-1024. https://doi.org/10.3892/etm.2017.4091
MLA
Zhou, W., Zhou, W., Zeng, Q., Xiong, J."MicroRNA-138 inhibits hypoxia-induced proliferation of endothelial progenitor cells via inhibition of HIF-1α-mediated MAPK and AKT signaling Retraction in /10.3892/etm.2021.10428". Experimental and Therapeutic Medicine 13.3 (2017): 1017-1024.
Chicago
Zhou, W., Zhou, W., Zeng, Q., Xiong, J."MicroRNA-138 inhibits hypoxia-induced proliferation of endothelial progenitor cells via inhibition of HIF-1α-mediated MAPK and AKT signaling Retraction in /10.3892/etm.2021.10428". Experimental and Therapeutic Medicine 13, no. 3 (2017): 1017-1024. https://doi.org/10.3892/etm.2017.4091