Open Access

Transcription factor E2-2 inhibits the proliferation of endothelial progenitor cells by suppressing autophagy

  • Authors:
    • Li Zhang
    • Yang Yu
    • Xi Xia
    • Yang Ma
    • Xie-Wan Chen
    • Zhen-Hong Ni
    • Hong Wang
  • View Affiliations

  • Published online on: March 9, 2016     https://doi.org/10.3892/ijmm.2016.2521
  • Pages: 1254-1262
  • Copyright: © Zhang 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

Endothelial progenitor cells (EPCs) play a key role in repairing the injured vascular endothelium by differentiating into mature endothelial cells (ECs) or secreting cytokines in a paracrine manner to promote proliferation of existing ECs. However, the mechanisms underlying the proliferation of EPCs were not fully understood. In order to investigate the mechanisms of EPC proliferation, we isolated EPCs from mononuclear cells of mouse spleens. By manipulating E2-2 expression in vitro, we observed that E2-2 negatively regulated the proliferation of EPCs. Moreover, we noted that E2-2 negatively regulated the autophagy of EPCs by studying the expression of LC3II and p62. We also demonstrated that an autophagy inhibitor chloroquine (CQ) decreased the proliferation of EPCs in a concentration-dependent manner. Interestingly, CQ reversed the increase in cell proliferation and autophagy in the E2-2 knockdown group. Furthermore, we detected the expression of autophagy‑related protein ATG7 in EPCs which had been transfected with small interfering (siRNA)‑E2-2 and siRNA‑autophagy related 7 (ATG7) or were untransfected. Our study revealed that E2-2 regulated EPC autophagy via mediating ATG7 expression. We conclude that E2-2 inhibited EPC proliferation via suppressing their autophagy, and E2-2 regulated EPC autophagy by mediating the expression of ATG7.

Introduction

Since the successful isolation of endothelial progenitor cells (EPCs) by Asahara et al in 1997 (1), EPCs have received increasing attention in the field of research on endothelial injury and repair. As the origins of vascular endothelial cells (ECs), EPCs repair damaged endothelium by directly differentiating into mature ECs and indirectly secreting paracrine cytokines that promote the proliferation of ECs (2,3). To date, EPC transplantation has been used to treat atherosclerosis and coronary heart disease caused by endothelial injuries (46). However, due to the limited proliferative capacity of EPCs, this strategy often leads to insufficient efficacy (7,8). Therefore, enhancing the proliferative ability of EPCs is a key step in improving the effects of EPC transplantation therapy.

The transcription factor E2-2, also referred to as transcription factor 4 (TCF4) or class A basic helix-loop-helix (bHLH) transcription factor, is a member of the E-protein family that is universally expressed in mammalian cells. E2-2 regulates transcription by binding to the E-box binding site in the promoter and enhancer regions of various genes, including nerve-, pancreas- and tumor-specific genes (9,10). It also promotes the proliferation of human liver cancer cells and dermal papilla cells through activating the Wnt/β-catenin signaling pathway (11,12). By contrast, another study has shown that knockout of E2-2 increased the proliferative and vessel-forming capacities of mature ECs in mice (13). However, the role and mechanism by which E2-2 regulates the proliferation of EPCs remains unclear.

Autophagy is a highly conserved metabolic process in eukaryotes, and one which is essential for maintaining cellular homeostasis. It has been previously reported that regulation of autophagy affects the proliferation of tumor cells (14), nerve cells (15) and vascular smooth muscle cells (16). A recent study has revealed that inhibition of basal autophagy by 3-methyl-adenine (3-MA) decreases the growth ability of human EPCs and prevents their differentiation into mature ECs, while hypoxia-induced autophagy increased the growth and differentiation of human EPCs, which led to an increased survival rate of EPCs under hypoxic conditions (17). In recent years, E2-2 has been shown to suppress autophagy by activating the Wnt/β-catenin signaling pathway in tumor cells (18). However, whether E2-2 regulates levels of autophagy in EPCs and whether levels of autophagy affect the proliferation of EPCs have not been previously reported.

In the present study, we firstly demonstrated that inhibition of E2-2 expression significantly increased the proliferative ability of EPCs through its effects on autophagy. Moreover, we found that E2-2 downregulated the autophagy level by decreasing the expression of AGT7. These findings provide a new insight into EPC proliferation, as we have targeted the 'E2-2/autophagy related 7 (ATG7)/autophagy' pathway, which will benefit EPC transplantation therapy used to treat atherosclerosis and coronary heart disease caused by endothelial injury.

Materials and methods

Isolation and characterization of EPCs

All animal experiments were approved by the Center of Experimental Animals Committee of Xinqiao Hospital (Chongqing, China). The male C57BL/6J mice (6–8 weeks, 18–20 g, from Xinqiao Hospital Experimental Animal Center, Chongqing, China) were sacrificed by cervical dislocation, and spleens were explanted and thoroughly minced. Spleen-derived mononuclear cells were isolated by density gradient centrifugation (Histopaque 1083; Sigma-Aldrich, St. Louis, MO, USA) at 400 × g for 20 min in 4°C. The cells were collected, washed in phosphate-buffered saline (PBS) three times, and then resuspended in Dulbecco's modified Eagle's medium/nutrient mixture F12 (DMEM/F12; Gibco BRL, Gaithersburg, MD, USA) supplemented with 20% fetal calf serum (FCS; HyClone, Los Angeles, CA, USA), 20 ng/ml vascular endothelial growth factor (VEGF; R&D Systems, Inc., Minneapolis, MN, USA), 100 U/ml penicillin and 100 g/ml streptomycin. Cells were seeded into gelatin-coated cell culture flasks and incubated at 37°C under an atmosphere with 5% CO2. Forty-eight hours later, non-attached cells were removed and adherent cells were cultured continuously. Only adherent cells were used in further experiments. The medium was refreshed with complete medium every 2 or 3 days.

To verify the phenotype of EPCs, cells were incubated with Dil-ac-LDL (Biomedical Technologies, Inc., Stoughton, MA, USA) for 3 h, fixed with 4% paraformaldehyde and then incubated with fluorescein isothiocyanate (FITC)-labeled lectin (UEA-I; Sigma-Aldrich) for 1 h, then washed with PBS three times. The cells were then observed under an immunofluorescence laser scanning confocal microscope (Leica TCS; Leica, Mannheim, Germany). Dual-stained cells positive for Dil-ac-LDL and UEA-I were identified as EPCs. Additionally, fluorescence-activated cell sorting (FACS) analysis was performed using FITC-conjugated antibodies against mouse Sca-1 (ab25031) and vascular endothelial growth factor receptor 2 (VEGFR2; ab11939), and the corresponding isotype control antibodies (ab18446, ab171870; all from Abcam, Cambridge, UK).

Small interfering RNA (siRNA)-mediated silencing of genes

Transient silencing of E2-2 and ATG7 was induced by transfection with siRNAs (siRNA-E2-2 and siRNA-ATG7, respectively) (both from GenePharma, Shanghai, China). The selected siRNA duplex sequences specifically targeted mouse E2-2 and ATG7, and showed no homology to any other sequences during a BLAST search. The siRNA-E2-2 efficient sequence was: sense, 5′-CAAGAAGGAUAUCAAAUCA-3′ and antisense, 5′-UGAUUUGAUAUCCUUCUUG-3′. The siRNA-ATG7 sequence was: sense, 5′-GGAGUCACAGCUCUUCCUUTT-3′ and antisense, 5′-AAGGAAGAGCUGUGACUCCTT-3′. The corresponding non-silencing control (siRNA-con) sequences (GenePharma) were designed according to the sequence of a negative control. Transfection with siRNA-E2-2 or siRNA-ATG7 was carried out using Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA, USA) with a molar ratio between DNA and lipid of approximately 1:3. Forty-eight hours after transfection, cells were collected and used for subsequent assays.

Adenovirus transfection

Adenovirus vector expressing E2-2 (Ad-E2-2) and adenovirus-carrying blank vector (Ad-vector) were both constructed by HanBio Technology Co., Ltd. (Shanghai, China). Before transfection, EPCs were incubated in DMEM/F12 medium without FCS and antibiotics for 6 h. Adenovirus vector was then added to the cells at a multiplicity of infection (MOI) of 100. The transfection medium was removed 2 h later, and the cells were maintained in the complete medium for 48 h. Ad-vector was used as a control. The mRFP-GFP-LC3 adenovirus vector for detecting autophagy was also purchased from HanBio Technology Co., Ltd. The process of mRFP-GFP-LC3 adenovirus transfection was similar to that of Ad-E2-2. Autophagy was observed under an immunofluorescence laser scanning confocal microscope (Leica TCS). Autophagy was detected by counting the number of green fluorescent protein (GFP) and red fluorescent protein (RFP) dots (dots/cell).

Cell proliferation assay

EPCs were collected from the cultures and replated into a 96-well plate (1×105 cells/ml) and underwent different treatments. Cell proliferation was measured using a Cell Counting Kit-8 (CCK-8; Beyotime Biotech, Jiangsu, China) according to the manufacturer's instructions. Before reading the absorbance at 450 nm, 10 µl CCK-8 solution and 100 µl complete medium were sequentially added to each well.

In addition, the cell cycle was analyzed by flow cytometric analysis. EPCs were harvested and resuspended in PBS solution at 1×106 cells/ml. The cells were then stored in 70% ethanol at 4°C overnight. Before being analyzed, the cells were mixed with 1 ml propidium iodide (PI; 1 mg/ml) (BD Biosciences, Bedford, MA, USA) and then incubated at 4°C for 30 min. After centrifugation, the cells were resuspended and the cell cycle was analyzed immediately using a flow cytometer (MoFloTMXDP; Beckman Coulter, Co. Fullerton, CA, USA). The proliferation index was defined as the total percentage of cells in S and G2/M phases. All groups of experiments were undertaken in triplicate.

Western blot analysis

EPCs were lysed in ice-cold lysis buffer (RIPA) containing a protease inhibitor (0.5 mM PMSF) (both from Beyotime Biotech). The cell lysates were further centrifuged at 15,000 × g for 15 min at 4°C. Proteins from EPC lysates were measured using the Bradford method. The same amount of protein was loaded in each lane, separated by 10–15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and transferred to 0.22 µm polyvinylidene difluoride (Millipore Corp., Boston, MA, USA) membranes. Membranes were blocked with 5% non-fat milk 2 h followed by incubated with primary antibodies overnight at 4°C. The primary antibodies included: rabbit anti-mouse LC3 polyclonal antibody (1:1,000; Cat. no. 2775), rabbit anti-mouse p62 monoclonal antibody (1:1,000; Cat. no. 5114), rabbit anti-mouse ATG7 polyclonal antibody (1:1,000; Cat. no. 8558) (all from Cell Signaling Technology, Inc., Boston, MA, USA) and rat anti-mouse β-actin monoclonal antibody (1:1,000; Cat. no. AA128; Beyotime Biotech). The membranes were then incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (1:5,000; A0208, A0216; Beyotime Biotech) for 1 h at 37°C. Protein bands were visualized by enhanced chemiluminescence (ECL) (Thermo Fisher Scientific, Waltham, MA, USA), and densitometric signals were quantified using ImageQuant TL software (General Electric, Co., Fairfield, CT, USA).

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from EPCs using RNAiso Plus (Takara Bio, Dalian, China) according to the manufacturer's instructions. Single cDNAs was synthesized from RNA using a PrimeScript™ RT reagent kit (Takara Bio). The following primer sequences were used for ATG7 (sense, 5′-ACCCAGAAGAAGCTGAACGA-3′, and antisense, 5′-CTCATTTGCTGCTTGTTCCA-3′); β-actin (sense, 5′-TTCTACAATGAGCTGCGTGTGG-3′ and anti-sense, 5′-GTGTTGAAGGTCTCAAACATGAT-3′). SYBR® Premix Ex Taq™ II (Takara Bio) was used according to the manufacturer's instructions, and RT-qPCR was performed using a StepOnePlus™ Real-Time PCR system (Applied Biosystems Life Technologies, Waltham, MA, USA). The reaction conditions were as follows: 95°C for 30 sec, 40 cycles of 95°C for 5 sec, and 60°C for 30 sec. Relative expression of target mRNA was normalized to β-actin using the ΔΔCt method, as previously described (19).

Statistical analysis

Data from at least three independent experiments are expressed as the means ± SD. SPSS 16.0 software was used for statistical analysis. Student's t-tests were used for comparisons between groups. A P-value <0.01, or P<0.05 was considered to indicate a statistically significant difference.

Results

Biological properties of EPCs

After 7 days of being cultured, round, oval and spindle shapes of attached spleen-derived mono-nuclear cells were noted under the microscope (Fig. 1A), similar to morphologies which have been previously described (20). Immunocytochemical (ICC) staining demonstrated that 92.33±2.63% (n=3) of attached cells were stained positively for FITC-UEA-I and Dil-ac-LDL (Fig. 1B), validating the theory that the FITC-UEA-I/Dil-ac-LDL double-positive cells possessed the characteristics of ECs. Furthermore, the attached cells were identified by analyzing the expression of the mouse stem-cell marker Sca-1 and EC marker VEGFR2 using flow cytometry. The percentage of positive cells was 88.93±1.31 and 85.13±2.83% (n=3), respectively (Fig. 1C). Taking these results into account, the attached cells were considered EPCs.

E2-2 inhibits the proliferation of EPCs

To determine whether E2-2 regulated the proliferation of EPCs, we overexpressed or silenced E2-2 by transfecting Ad-E2-2 or siRNA-E2-2 into the cells, and proliferation was then detected by CCK-8 and cell cycle assays in vitro. The CCK-8 assay demonstrated that the proliferation of EPCs significantly decreased in the E2-2 overexpression (Ad-E2-2) group compared with the adenoviral control vector (Ad-vector) group (Fig. 2A). Cell cycle analysis showed that the proliferation index (S + G2/M) of the EPCs was lower in the Ad-E2-2 group than in the Ad-vector group (Fig. 2B and C). The trend of the cell cycle was similar to the results of the CCK-8 assay. However, in the knockdown E2-2 (siRNA-E2-2) group, the proliferative capacity of EPCs significantly increased compared with that in the negative control (siRNA-con) group (Fig. 2D). The EPC proliferation index (S+G2/M) of the siRNA-E2-2 group was higher than that of the siRNA-con group (Fig. 2E and F). These results suggest that E2-2 suppressed the proliferation of EPCs in vitro.

E2-2 negatively regulates the autophagy of EPCs

To investigate whether E2-2 regulates autophagy in EPCs, we firstly studied the expression of LC3II and p62 while overexpressing or knocking down E2-2. We noted a significant decrease in LC3II expression, but a marked increase in the bands of p62, in the Ad-E2-2 group compared with the Ad-vector group (Fig. 3A), suggesting that overexpression of E2-2 induced suppression of autophagy in EPCs. By contrast, LC3II expression increased and the protein level of p62 decreased in the knockdown E2-2 group (Fig. 3B). Thus, knockdown of E2-2 upregulated autophagy of EPCs. In addition, we used the mRFP-GFP-LC3 adenovirus to confirm the regulatory effects of E2-2 on autophagy. This assay is used to monitor progression from autophagosome to autolysosome based on the pH difference between the acidic autolysosome and the neutral autophagosome and the pH sensitivity differences displayed by GFP and RFP. When autolysosomes are formed by fusing autophagosome with a lysosome, the GFP moiety degrades from the tandem protein, but RFP-LC3 maintains the dots (21,22). The red dots indicate autophagosomes, and yellow dots (merge of RFP and GFP) represent autolysosomes. When we co-transfected mRFP-GFP-LC3 adenovirus with Ad-E2-2 or siRNA-E2-2 in EPCs for 48 h, we observed that overexpression of E2-2 reduced the numbers of autophagosomes and autolysosomes (Fig. 3C). Conversely, knockdown of E2-2 increased not only the number of autophagosomes but also that of autolysosomes (Fig. 3D). The results further confirmed that E2-2 negatively regulated autophagy of EPCs.

E2-2 inhibits the proliferation of EPCs via suppressing autophagy

To validate the role autophagy plays in EPC proliferation, we treated EPCs with chloroquine (CQ) at different concentrations and different periods of time. It was observed that CQ inhibited the proliferation of EPCs in a concentration-dependent manner. After treatment with CQ for 24 h at 20 µM, the proliferation of EPCs as measured by CCK-8 decreased markedly compared with the group which was not treated with CQ, and the degree of decrease changed slightly with the increasing periods of time (Fig. 4B). Subsequently, to determine whether autophagy was involved in E2-2-mediated EPC proliferation, we added 20 µM CQ to EPCs after transfection with siRNA-E2-2 for 24 h. After transfection for 48 h, the CCK-8 assay showed that the increase in proliferation in the siRNA-E2-2 group was partially reversed by the administration of CQ (Fig. 4C). The alteration of the proliferation index, as measured by flow cytometry, was similar to the results of the CCK-8 assay (Fig. 4D). Furthermore, we transfected the different groups of EPCs with siRNA-con, siRNA-E2-2 or siRNA-E2-2 and CQ and found that CQ inhibited the increase in autophagosomes and autolysosomes (Fig. 4E), suggesting that EPC autophagy induced by E2-2 knockdown was reversed by CQ. These results demonstrated that inhibition of basal autophagy reduced the proliferation of EPCs, and E2-2 inhibited the proliferation of EPCs through its effects on autophagy.

E2-2 regulates autophagy of EPCs via mediating the expression of ATG7

To further explore the mechanism by which E2-2 regulates autophagy in EPCs, we detected the expression of the autophagy-related gene ATG7 after E2-2 overexpression or knockdown. We found that overexpression of E2-2 decreased the expression level of ATG7 mRNA (Fig. 5A), and knockdown of E2-2 upregulated the expression of ATG7 mRNA (Fig. 5B). Subsequently, we noted that overexpression of E2-2 downregulated ATG7 protein expression, and knockdown of E2-2 upregulated ATG7 protein expression (Fig. 5C and D). Furthermore, to demonstrate whether ATG7 was involved in E2-2-mediated autophagy, we transfected the cells with siRNA-E2-2 and siRNA-ATG7 simultaneously or respectively. The transfection revealed that autophagy of EPCs was downregulated by ATG7 knockdown alone, and the increased autophagy induced by siRNA-E2-2 was reversed by ATG7 knockdown. Interestingly, the level of autophagy was still higher in the co-transfected siRNA-E2-2 and siRNA-ATG7 group than in siRNA-ATG7 group (Fig. 5E). We demonstrated that E2-2 regulated autophagy of EPCs via partially mediating the expression of ATG7.

Discussion

As a transcription factor, E2-2 binds to the promoter regions and affects the expression of target proteins, ultimately influencing several cellular biological functions, including proliferation. In ECs, Bach1 binds to E2-2, which represses the activation of Wnt/β-catenin signaling and decreases the proliferative and vessel-forming abilities of the cells (23). Tanaka et al revealed that E2-2 suppressed the proliferation of vascular ECs via binding to the promoter of VEGFR2. Knockdown of the E2-2 gene significantly improved the proliferative and vessel-forming abilities of ECs (13). Consistent with these reports, our findings showed that E2-2 negatively regulated the proliferation of EPCs. However, another study has shown that downregulation of E2-2 suppressed the proliferation and survival of corneal epithelial stem cells (24). Xiong et al demonstrated that upregulation of E2-2 activated the Wnt/β-catenin signaling pathway, initiating the proliferation of dermal papilla cells (12). Therefore, we consider that the mechanisms of E2-2 in the regulation of cell proliferation are complicated, and may be associated with the different cellular contents and microenvironment.

Autophagy is a highly conserved cell activity that involves degradation of dysfunctional organelles and proteins. The breakdown of cellular components promotes cellular survival by maintaining cellular energy levels and homeostasis (25,26). Previously it has been confirmed that autophagy plays an indispensable role in maintaining stem-cell characteristics, self-renewal, and the capacity for differentiation in various stem cells (27). Moreover, previous research has demonstrated that upregulation of autophagy promotes the proliferation of cancer cells (28), while downregulation of autophagy decreases cell proliferation (29). In nerve cells, autophagy induced by high mobility group box 1 protein (HMGB1) increased the growth of neuroblasts (30). In immune cells, inhibition of autophagy decreased cell growth and differentiation and promoted apoptosis (31). By contrast, another study has suggested that induction of autophagy suppressed cancer cell proliferation (32). In the present study, inhibition of autophagy at basal levels by the inhibitor CQ decreased the proliferative ability of EPCs. Therefore, we suggest that distinct cell types have different levels of autophagy, thus generating various effects on cell growth and survival. The underlying mechanism of action remains to be explored.

In the present study, to the best of our knowledge, we were the first to find that E2-2 inhibited the autophagy of EPCs via decreasing the expression of ATG7. ATG7 is a gene that plays a key role in autophagy and normal cell physiology (33,34). Previous studies showed that ATG7 is required for normal functioning of ECs via maintaining the basal levels of autophagy (35). In HeLa cells, inhibition of ATG7 suppressed the level of autophagy, which decreased cell proliferation (36). Moreover, Lee et al discovered that depletion of ATG7 suppressed autophagy and inhibited the activation of p53 in mouse embryonic fibroblasts, thus inducing cell cycle arrest and apoptosis (37). Our present study is thus the first to elucidate that overexpression of E2-2 in mouse EPCs downregulated ATG7 mRNA and protein levels, whereas knockdown of E2-2 led to upregulation of ATG7 mRNA and protein levels. After simultaneous or respective knockout of E2-2 and ATG7 with siRNAs, we found that knockdown of ATG7 caused a decrease in autophagy levels. Moreover, increased autophagy levels after knockdown of E2-2 waspartially reversed by ATG7 knockdown. Our findings suggest that in EPCs ATG7 mRNA and protein levels are regulated by E2-2. Increased levels of autophagy after depletion of E2-2 may be partially attributed to the enhanced transcription of ATG7.

Therefore, we noted the existence of the E2-2/ATG7/autophagy signaling pathway and its regulatory effects on EPC proliferation. However, the binding site of E2-2 with ATG7 and the mechanism by which ATG7 transcription is regulated remain to be explored in future research.

The present study attempted to clarify the mechanism underlying EPC proliferation by investigating autophagy, and provided new insights into research on EPCs and potential novel therapies for repairing endothelial injury and treating atherosclerosis and coronary heart disease.

Acknowledgments

The present study was supported by grants from the National Natural and Science Foundation of China (NSFC) (no. 81270224). We thank Ms. Huali Kang, Ms. Chunxing Xu and Mr. Jie Yang for their kind help (Department of Cardiology, Institute of Cardiovascular Science of PLA, Xinqiao Hospital, Third Military Medical University). Thanks to Professor Jihang Zhang (Department of Cardiology, Institute of Cardiovascular Science of PLA, Xinqiao Hospital, Third Military Medical University) for a critical reading of the manuscript.

References

1 

Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G and Isner JM: Isolation of putative progenitor endothelial cells for angiogenesis. Science. 275:964–967. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Ii M, Nishimura H, Iwakura A, Wecker A, Eaton E, Asahara T and Losordo DW: Endothelial progenitor cells are rapidly recruited to myocardium and mediate protective effect of ischemic preconditioning via 'imported' nitric oxide synthase activity. Circulation. 111:1114–1120. 2005. View Article : Google Scholar : PubMed/NCBI

3 

George AL, Bangalore-Prakash P, Rajoria S, Suriano R, Shanmugam A, Mittelman A and Tiwari RK: Endothelial progenitor cell biology in disease and tissue regeneration. J Hematol Oncol. 4(24)2011. View Article : Google Scholar : PubMed/NCBI

4 

Wassmann S, Werner N, Czech T and Nickenig G: Improvement of endothelial function by systemic transfusion of vascular progenitor cells. Circ Res. 99:e74–e83. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Lee PS and Poh KK: Endothelial progenitor cells in cardiovascular diseases. World J Stem Cells. 6:355–366. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Xu JY, Lee YK, Wang Y and Tse HF: Therapeutic application of endothelial progenitor cells for treatment of cardiovascular diseases. Curr Stem Cell Res Ther. 9:401–414. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Wöhrle J, Birkemeyer R, Markovic S, Nguyen TV, Sinha A, Miljak T, Spiess J, Rottbauer W and Rittger H: Prospective randomised trial evaluating a paclitaxel-coated balloon in patients treated with endothelial progenitor cell capturing stents for de novo coronary artery disease. Heart. 97:1338–1342. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Raval Z and Losordo DW: Cell therapy of peripheral arterial disease: from experimental findings to clinical trials. Circ Res. 112:1288–1302. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Massari ME and Murre C: Helix-loop-helix proteins: regulators of transcription in eucaryotic organisms. Mol Cell Biol. 20:429–440. 2000. View Article : Google Scholar

10 

Sobrado VR, Moreno-Bueno G, Cubillo E, Holt LJ, Nieto MA, Portillo F and Cano A: The class I bHLH factors E2-2A and E2-2B regulate EMT. J Cell Sci. 122:1014–1024. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Zhao DH, Hong JJ, Guo SY, Yang RL, Yuan J, Wen CY, Zhou KY and Li CJ: Aberrant expression and function of TCF4 in the proliferation of hepatocellular carcinoma cell line BEL-7402. Cell Res. 14:74–80. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Xiong Y, Liu Y, Song Z, Hao F and Yang X: Identification of Wnt/β-catenin signaling pathway in dermal papilla cells of human scalp hair follicles: TCF4 regulates the proliferation and secretory activity of dermal papilla cell. J Dermatol. 41:84–91. 2014. View Article : Google Scholar

13 

Tanaka A, Itoh F, Nishiyama K, Takezawa T, Kurihara H, Itoh S and Kato M: Inhibition of endothelial cell activation by bHLH protein E2-2 and its impairment of angiogenesis. Blood. 115:4138–4147. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Guo S, Bai R, Liu W, Zhao A, Zhao Z, Wang Y, Wang Y, Zhao W and Wang W: miR-22 inhibits osteosarcoma cell proliferation and migration by targeting HMGB1 and inhibiting HMGB1-mediated autophagy. Tumour Biol. 35:7025–7034. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Rubinsztein DC, Mariño G and Kroemer G: Autophagy and aging. Cell. 146:682–695. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Dong N, Zhu Q, Zhang P, Zhu C, Wang M, Li W, Liu J, Liu Y, Ma B and Wu K: Autophagy downregulates thrombin-induced VSMCs proliferation through lysosomal pathway. Int J Cardiol. 159:156–158. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Wang HJ, Zhang D, Tan YZ and Li T: Autophagy in endothelial progenitor cells is cytoprotective in hypoxic conditions. Am J Physiol Cell Physiol. 304:C617–C626. 2013. View Article : Google Scholar

18 

Petherick KJ, Williams AC, Lane JD, Ordóñez-Morán P, Huelsken J, Collard TJ, Smartt HJ, Batson J, Malik K, Paraskeva C and Greenhough A: Autolysosomal β-catenin degradation regulates Wnt-autophagy-p62 crosstalk. EMBO J. 32:1903–1916. 2013. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Yin Y, Huang L, Zhao X, Fang Y, Yu S, Zhao J and Cui B: AMD3100 mobilizes endothelial progenitor cells in mice, but inhibits its biological functions by blocking an autocrine/paracrine regulatory loop of stromal cell derived factor-1 in vitro. J Cardiovasc Pharmacol. 50:61–67. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Wang X, Liu J, Zhen J, Zhang C, Wan Q, Liu G, Wei X, Zhang Y, Wang Z, Han H, et al: Histone deacetylase 4 selectively contributes to podocyte injury in diabetic nephropathy. Kidney Int. 86:712–725. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Ikeda Y, Shirakabe A, Maejima Y, Zhai P, Sciarretta S, Toli J, Nomura M, Mihara K, Egashira K, Ohishi M, et al: Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ Res. 116:264–278. 2015. View Article : Google Scholar

23 

Jiang L, Yin M, Wei X, Liu J, Wang X, Niu C, Kang X, Xu J, Zhou Z and Sun S: Bach1 represses Wnt/β-catenin signaling and angiogenesis. Circ Res. 117:364–375. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Lu R, Qu Y, Ge J, Zhang L, Su Z, Pflugfelder SC and Li DQ: Transcription factor TCF4 maintains the properties of human corneal epithelial stem cells. Stem Cells. 30:753–761. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Levine B and Kroemer G: Autophagy in the pathogenesis of disease. Cell. 132:27–42. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Galluzzi L, Pietrocola F, Levine B and Kroemer G: Metabolic control of autophagy. Cell. 159:1263–1276. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Guan JL, Simon AK, Prescott M, Menendez JA, Liu F, Wang F, Wang C, Wolvetang E, Vazquez-Martin A and Zhang J: Autophagy in stem cells. Autophagy. 9:830–849. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Li Y, Zhang L, Zhou J, Luo S, Huang R, Zhao C and Diao A: Nedd4 E3 ubiquitin ligase promotes cell proliferation and autophagy. Cell Prolif. 48:338–347. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Ding Y, Gao H, Zhao L, Wang X and Zheng M: Mitofusin 2-deficiency suppresses cell proliferation through disturbance of autophagy. PLoS One. 10:e01213282015. View Article : Google Scholar : PubMed/NCBI

30 

Liu Y and Song L: HMGB1-induced autophagy in Schwann cells promotes neuroblastoma proliferation. Int J Clin Exp Pathol. 8:504–510. 2015.PubMed/NCBI

31 

Pei B, Zhao M, Miller BC, Véla JL, Bruinsma MW, Virgin HW and Kronenberg M: Invariant NKT cells require autophagy to coordinate proliferation and survival signals during differentiation. J Immunol. 194:5872–5884. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Cianfanelli V, Fuoco C, Lorente M, Salazar M, Quondamatteo F, Gherardini PF, De Zio D, Nazio F, Antonioli M, D'Orazio M, et al: AMBRA1 links autophagy to cell proliferation and tumorigenesis by promoting c-Myc dephosphorylation and degradation. Nat Cell Biol. 17:20–30. 2015. View Article : Google Scholar

33 

Lee IH, Cao L, Mostoslavsky R, Lombard DB, Liu J, Bruns NE, Tsokos M, Alt FW and Finkel T: A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci USA. 105:3374–3379. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Kageyama S and Komatsu M: Impaired G1-arrest, autophagy, and apoptosis in Atg7-knockout mice. Circ Res. 111:962–964. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Torisu T, Torisu K, Lee IH, Liu J, Malide D, Combs CA, Wu XS, Rovira II, Fergusson MM, Weigert R, et al: Autophagy regulates endothelial cell processing, maturation and secretion of von Willebrand factor. Nat Med. 19:1281–1287. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Thorburn J, Andrysik Z, Staskiewicz L, Gump J, Maycotte P, Oberst A, Green DR, Espinosa JM and Thorburn A: Autophagy controls the kinetics and extent of mitochondrial apoptosis by regulating PUMA levels. Cell Reports. 7:45–52. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Lee IH, Kawai Y, Fergusson MM, Rovira II, Bishop AJ, Motoyama N, Cao L and Finkel T: Atg7 modulates p53 activity to regulate cell cycle and survival during metabolic stress. Science. 336:225–228. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 37 Issue 5

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
Zhang L, Yu Y, Xia X, Ma Y, Chen X, Ni Z and Wang H: Transcription factor E2-2 inhibits the proliferation of endothelial progenitor cells by suppressing autophagy. Int J Mol Med 37: 1254-1262, 2016
APA
Zhang, L., Yu, Y., Xia, X., Ma, Y., Chen, X., Ni, Z., & Wang, H. (2016). Transcription factor E2-2 inhibits the proliferation of endothelial progenitor cells by suppressing autophagy. International Journal of Molecular Medicine, 37, 1254-1262. https://doi.org/10.3892/ijmm.2016.2521
MLA
Zhang, L., Yu, Y., Xia, X., Ma, Y., Chen, X., Ni, Z., Wang, H."Transcription factor E2-2 inhibits the proliferation of endothelial progenitor cells by suppressing autophagy". International Journal of Molecular Medicine 37.5 (2016): 1254-1262.
Chicago
Zhang, L., Yu, Y., Xia, X., Ma, Y., Chen, X., Ni, Z., Wang, H."Transcription factor E2-2 inhibits the proliferation of endothelial progenitor cells by suppressing autophagy". International Journal of Molecular Medicine 37, no. 5 (2016): 1254-1262. https://doi.org/10.3892/ijmm.2016.2521