Transplantation of bone marrow‑derived endothelial progenitor cells overexpressing Delta‑like‑4 enhances functional neovascularization in ischemic myocardium

  • Authors:
    • Hong Huang
    • Feng Huang
    • Jian‑Ping Huang
  • View Affiliations

  • Published online on: August 28, 2013     https://doi.org/10.3892/mmr.2013.1657
  • Pages: 1556-1562
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Delta‑like‑4 (Dll‑4) prevents excess angiogenic sprouting and promotes the formation of a well‑differentiated vascular network. Therefore, transplantation of Dll‑4‑overexpressing endothelial progenitor cells (EPCs) was hypothesized to be superior to transplantation of EPCs in the treatment of ischemic heart disease. In the current study, EPCs harvested from C57BL/6 mouse bone marrow were infected in vitro with Dll‑4 (EPCDll‑4+) or Dll‑4 knockdown (EPCDll‑4‑) with recombinant lentiviral vectors and the control cells were non‑transfected or transduced with mock vectors (EPCnull). Eight‑week‑old C57BL/6 mice underwent ligation of the left anterior descending artery to establish a myocardial infarction (MI) model. The ligated animals were randomly divided into 5 groups, which, following one week, were intravenously injected with EPCs, EPCnull, EPCDll‑4+, EPCDll‑4‑ or medium. Two weeks later, echocardiographic assessment, western blotting, fluorescent microsphere and histological studies were performed. The results demonstrated that the number of mature vessels and blood flow in ischemic myocardium were increased in the EPCDll‑4+ group, but were markedly decreased in the EPCDll‑4‑ group compared with the control groups. The expression levels of Dll‑4, hairy/enhancer of split (Hes)‑related protein 1 (Hey‑1), phosphorylation of mammalian target of rapamycin (mTOR) and p70S6 kinase (p70S6K) were significantly increased in the EPCDll‑4+ group, while they were markedly decreased in the EPCDll‑4‑ group. Furthermore, for EPCDll‑4+‑treated animals, an enhanced cardiac function was observed as assessed by echocardiography. Thus, the transplantation of Dll‑4‑overexpressing EPCs stimulates neovascularization effectively, increases the blood flow to the ischemic zone and improves cardiac function. These effects may be due to the activation of Notch/Hey‑1/mTOR/p70S6K signaling pathways, which are initiated by Dll‑4.

Introduction

The formation of new blood vessels is an important process required for healing wounds and for restoring blood flow to tissue following injury or insult (1). A number of stem/progenitor cells, including bone marrow-derived endothelial progenitor cells (EPCs) and mononuclear cell, have been identified for their pro-angiogenic potential to improve perfusion as an experimental or clinical option (2,3). As precursors of mature endothelial cells (ECs), EPCs are capable of increasing the neovascularization of ischemic tissue and delaying the initiation and progression of coronary artery disease (4,5). Circulating EPCs were found to exhibit similar features of ECs, possess the ability to home to sites of ischemia and contribute to the formation of new blood vessels (6). Thus, EPCs have been used as seeding cells in tissue engineering and stem cell therapy.

Increasing evidence has demonstrated that the Notch signaling pathway is one of the most important mechanisms involved in the regulation of neovascularization. In mammals, the Notch signaling pathway includes five transmembrane ligands, i.e., Delta-like (Dll)-1, Dll-3, Dll-4, Jagged (Jag)-1 and Jag-2 and four transmembrane receptors, i.e., Notch-1, -2, -3 and -4. The interaction between the Notch receptor and ligand leads to cleavage of the Notch intracellular domain (NICD) and translocation to the nucleus, thereby activating downstream target genes, including basic helix-loop-helix (bHLH) proteins which include hairy/enhancer of split (Hes) and Hes-related protein (Hey) (7,8). The Notch ligand Dll-4 has been identified as a promising new target for angiogenesis in preclinical studies. For example, the balance between sprout and tube formation is established as being important for the generation of a new functional vessel, which is hypothesized to be modulated by Dll-4 from tip ECs to neighboring stalk ECs in order to restrict the emergence of excessive sprout through the repression of vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) transcription and consequently the reduction of responsiveness to VEGF (9,10). By contrast, the inhibition of Dll-4-mediated signaling is hypothesized to retard tumor growth, despite an increase of tumor vasculature density since the established vascular network is functionally inefficient (1113), suggesting that Dll-4 attenuates the formation of ineffective vascular branch and promotes the remainder to form functional vessels. Thus, Dll-4-overexpressing EPCs were hypothesized to be superior to untreated EPCs in the formation of productive blood vessels in ischemic myocardium.

Materials and methods

Animal ethics

All procedures were performed in compliance with the guidelines for the Care and Use of Laboratory Animals published by the National Institutes of Health (NIH Publication No. 85-23, revised 1996) and approved by the Animal Care and Use Committee of the First People’s Hospital of Nanning, China.

Isolation, cultivation and identification of EPCs

Two 6-week-old C57BL/6 mice were sacrificed by cervical dislocation. Following removal of the tips of the hind legs and vertebrae, bone marrow was collected by flushing out the content of femurs and tibias with PBS. Mononuclear cells were collected from the bone marrow by density gradient centrifugation using Histopaque 1077 (Sigma-Aldich, St. Louis, MO, USA) according to the manufacturer’s instructions. The isolated cells were cultivated in dishes coated with fibronectin (R&D Systems, Minneapolis, MN, USA) and induced by EBM-2 Single Quots (Lonza, Basel, Switzerland) with supplements at 37°C with 5% CO2 in humidified air at a density of 5×106 cells/cm2. Following 3 days in culture, non-adherent cells were removed by washing with PBS, new medium was applied and the cultivation was maintained for 7 days. Immunofluorescence staining and flow cytometry were used to identify EPCs, which was performed as previously described by Liu et al(14).

Recombinant lentiviral vector construction and cell infection

The procedures for recombinant lentiviral vector construction, cell infection and selection of the stable cell line were performed as previously described by Chen and Zhou (15). In brief, to produce Dll-4 recombinant lentiviral vectors, the plasmids pAJ-Ubi-eGFP-3Flag, psPAX2 (gag/pol element) and pMD2.G (VSVG element) (Auragene Bioscience Inc., Changsha, China) were used according to the manufacturer’s instructions. Following lentiviral vector infection, the stable cell line overexpressing Dll-4 was cultured in a 5% CO2-humidified incubator at 37°C. EPCs infected with Dll-4 recombinant lentiviral vectors were labeled EPCDll-4+ and EPCs infected with mock vectors were labeled EPCnull.

To determine whether transplantation of Dll-4-inhibited EPCs had an opposed effect on neovascularization, EPCs were infected with lentiviral constructs encoding short hairpin RNA (shRNA) against Dll-4. The plasmids pAJ-U6-shRNA-CMV-Puro/eGFP, psPAX2 and pMD2.G (Auragene Bioscience) were transfected into 293T cells according to the instructions for Lipofectamine 2000 (Invitrogen Life Technologies, Carlsbad, CA, USA). Following a 48-h transduction, infected EPCs were selected to generate a stable Dll-4-shRNA line. EPCs transfected with recombinant lentiviral vectors encoding Dll-4-shRNA were labled EPCDll-4−.

Quantitative PCR (qPCR) was used to identify the efficiency of gene transfection

Total RNA was extracted from each sample with TRIzol reagent (Invitrogen Life Technologies) and reversed transcribed into first-strand cDNA by RevertAid™ First Strand cDNA Synthesis kit (Fermentas, Vilnius, Lithuania) according to the manufacturer’s instructions. The synthesized cDNA was used for qPCR analysis of Dll-4 mRNA expression with SYBR® Premix Ex Taq™ (Takara Bio, Inc., Shiga, Japan). The sense sequence of Dll-4 primers was 5′-CGAGGGAACAGAGTTGAGGAGT-3′ and the antisense sequence was 5′-AATACAGATGCCCACAGGAGC-3′. Fluorescence qPCR was performed on the ABI PRISM 7300 SDS apparatus (Applied Biosystems, Foster City, CA, USA).

Western blot analysis

Each sample was lysed in 0.2 ml lysis buffer (Calbiochem, La Jolla, CA, USA). The protein concentrations were determined by the Bradford method (Bio-Rad, Hercules, CA, USA). Total protein (20 μg) was separated on 10% SDS-PAGE gels and transferred to PVDF membranes (Millipore, Billerica, MA, USA) using the semi-dry transfer method. Membranes were blocked for 1 h in Tris-buffered saline containing 0.01% Tween-20 with 10% non-fat dried milk and incubated overnight at 4°C with the relevant antibodies: Dll-4 and Hey-1 antibody (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), mammalian target of rapamycin (mTOR) and phospho-mTOR (Ser2448) antibody (Abcam, Cambridge, UK), p70S6 kinase (p70S6K) and phospho-p70S6K (Thr421/Ser424) antibody (Cell Signaling Technology, Inc., Danvers, MA, USA). Membranes were rinsed and incubated for 1 h with the corresponding peroxidase-conjugated secondary antibodies. Chemiluminescent detection was performed using the ECL kit (Pierce Biotechnology, Inc., Rockford, IL, USA). All bands were analysed using Image J software (version 1.6 NIH).

In vitro angiogenesis assay

Tubulogenesis was induced using an in vitro Angiogenesis Assay kit (#ECM625, Millipore) following the manufacturer’s instructions (16). Briefly, ECMatrix™ solution was thawed on ice overnight, mixed with 10X ECMatrix™ diluents and placed in a 96-well tissue culture plate at 37°C for 1 h to allow the matrix solution to solidify. EPCs (1×104 cells/well) in 50 μl of medium were cultured on the top of the solidified matrix solution. Following 18 h of incubation at 37°C, the tubule formation was inspected under an inverted light microscope at ×200 magnification. Tubule formation was defined as a structure exhibiting a length four times its width. Five random microscopic fields were assessed for each well and the average number and the total length of tubules/200× field was determined and compared with the control cells.

Assessment of cell death induced by H2O2

The viability of EPCs was determined by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) (Sigma-Aldrich) assay. A total of 1×104 cells were equally seeded into each well in 96-well microplates. Following incubation with H2O2 for 1 h, the medium was replaced with MTT solution (0.5 mg/ml in PBS). Incubation was continued for 4 h and then the supernatant was gently removed. Dimethyl sulfoxide (DMSO) (Sigma-Aldrich) was added and the absorbance was read at 490 nm on a spectrophotometer (Bio-Rad) and the percentage of cell viability was obtained.

Creation of myocardial infarction (MI) model and cell transplantation

Eight-week-old C57BL/6 mice (weighing 20±2 g) underwent ligation of the left coronary artery to produce MI. Following anesthetization, the animals were orally intubated with a 1.0-mm OD intubation cannula and connected to a small animal volume-control ventilator (HES-HA MiniVent 845, Harvard Apparatus, Holliston, MA, USA). The left anterior descending artery was ligated 2–3 mm from its origin between the pulmonary artery conus and the left atrium using 8-0 sutures. The ligated animals were monitered by electrocardiograph using the RM6240BD system (Chengdu Instrument Company, Chengdu, China) to determine MI models. One week following coronary ligation, the surviving mice were randomly subdivided into 5 groups (PBS, EPCs, EPCnull, EPCDll-4+ and EPCDll-4−; 20 animals in each) and administered with intravenous injection of PBS, EPCs, EPCnull, EPCDll-4− and EPCDll-4+ in the tail vein. Each animal received an injection of 5×106 cells/100 μl in PBS or PBS alone with a total volume of 50 μl.

Assessment of cardiac function by echocardiography

A transthoracic echocardiographic study was performed by an experienced blinded cardiologist at 14 days post-transplantation using an echocardiographic system (SONOS 5500, Hewlett-Packard, Andover, MA, USA) equipped with a 15.0 MHz transducer. For analysis of left ventricular (LV) function, left ventricular internal dimensions (LVID) were measured at diastole (LVIDD) and systole (LVIDS). After two-dimensional images were obtained, the M-mode cursor was positioned to the parasternal long axis view at the papillary muscle level. LV ejection fraction (EF) and fractional shortening (FS) were calculated as follows: EF (%) = (LVIDD3 - LVIDS3)/LVIDD3 × 100% and FS (%) = [(LVIDD - LVIDS)/LVIDD] × 100%, respectively. Each parameter was measured from a minimum of 3 consecutive beat cycles in each image.

Myocardial blood flow

Measurement of blood flow to the peri-infarcted and infarcted area of mouse hearts was performed as previously described (17).

Capillary density

To identify mature capillary density, the tissue sections (5 μm) of the infarcted zone were stained with anti-VIII factor antibody (Santa Cruz Biotechnology, Inc.). Immunohistochemical staining was performed as previously described (17).

Statistical analysis

Data are presented as the mean ± SD. A method of ANOVA (analysis of variance) with Scheffe’s post-hoc test was used to identify differences among the groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Efficiency of gene transfection and alteration of relative signals

qPCR data indicated that Dll-4 gene expression in EPCDll-4+ was 17-fold higher compared with the expression in EPCs. The Dll-4 mRNA level was ~65.4% lower in EPCDll-4− compared with EPCs. Western blot analysis showed that the expression of Dll-4, Hey-1, phospho-mTOR and phospho-p70S6K were markedly increased in EPCDll-4+ and markedly decreased in EPCDll-4− (Fig. 1). Furthermore, a similar alteration was also detected in the EPCs-, EPCnull-, EPCDll-4+- and EPCDll-4−-treated animals when the peri-infarcted and infarcted heart tissue was extracted for western blot analysis. These observations suggested that the overexpression of Dll-4 results in the increase of its downstream target molecule, Hey-1 and thus, the activation of mTOR signaling pathway.

Protective effect of Dll-4 on the H2O2-injured EPCs

To determine the effect of Dll-4 on the viability of H2O2-treated EPCs, the cells of each group were exposed to increasing concentrations of H2O2 from 0 to 1,000 μmol/l (data from the pilot study were not shown). The previous study showed that 150 μmol/l H2O2 is the optimal concentration. Dll-4-overexpressing EPCs were more resistant to H2O2 compared with EPCs or EPCnull following incubation with the apoptotic stimulus (P<0.01). By contrast, Dll-4-inhibited EPCs were more readily damaged by H2O2 (Fig. 2).

Capillary-like tube formation

EPCs were seeded on a solidified matrix and incubated with collected medium for 18 h. Quantification of branch points/200× microscopic field demonstrated that the number of tubular structures in the EPCDll-4+ group was higher compared with the EPC and EPCnull groups. Tubules in the EPCDll-4+ group were qualitatively different and more complex compared with those in the control wells. Despite the number of emerging sprouts being greater in the EPCDll-4− group, they had an irregular and disorganized shape, suggesting that knockdown of the Dll-4 gene impacts on EPCs ability to form functional tubules (Fig. 3).

Improvement of LV contractile function

To assess cardiac function, echocardiographic studies were performed 2 weeks post-transplantation. EF and FS were improved in the EPCs, EPCnull and EPCDll-4+ groups when compared with the PBS group (P<0.01 or P<0.001). Furthermore, EF and FS were higher in the EPCDll-4+ group compared with the EPCs and EPCnull groups (P<0.05; Fig. 4). However, there was no statistical significance in enhancing cardiac function between EPCDll-4− and PBS therapy (P>0.05). These results indicate that transplantation of the Dll-4-overexpressing EPCs had an improved therapeutic effect for improving the LV function of MI animals compared with transplantation of untreated EPCs. By contrast, pre-inhibition of Dll-4 in the transplanted cells resulted in a poor effect.

Transplantation of Dll-4-overexpressing EPCs increases to form mature micovessels in ischemic myocardium

Semiquantitative analysis showed that, 2 weeks following cell transplantation, the number of blood vessels in peri-infarcted and infarcted tissue was significantly increased in the EPC (23.1±4.8) and EPCnull (22.6±5.2) groups when compared with the PBS group (13.2±2.4) (P<0.01). The microvessel number was further increased in the EPCDll-4+ group and an increase in immature vessel proliferation was observed in the peri-infarcted and infarcted area of mouse heart 2 weeks following treatment with EPCDll-4−, however, mature vessels with a lumen were barely detected (Fig. 5). This indicated that Dll-4 may contribute to angiogenesis and tubulogenesis in ischemic myocardium.

Treatment of Dll-4-overexpressing EPCs increases the blood flow in ischemic myocardium

To determine whether new blood vessels translate to increased coronary blood flow to the infarcted myocardium, functional microvessels were identified in the infarcted heart using the fluorescent microsphere method for regional blood flow assessment 2 weeks following transplantation. There was a significant decrease in blood flow to the infarcted and peri-infarcted zone and an increase to ~48% of the normal level was observed when EPCs and EPCnull treatment was induced. Notably, transplantation of EPCDll-4+ further increased the blood flow to ~65.1% that of the normal level. However, no evident improvement was detected when treated with PBS or EPCDll-4− (Fig. 6).

Discussion

In terms of EPCs, abundant evidence supports their involvement in capillary growth and the formation of collateral vessels. These consequent effects have led to improved perfusion and functional recovery in animal models of myocardial and peripheral ischemia (4,18). In early clinical trials, the therapeutic administration of EPCs to patients with MI or chronic angina has been associated with positive trends in perfusion (2). However, low survival and the angiogenic potential of treated cells in ischemic myocardium affects the outcome of EPC transplantation for the treatment of ischemic disease. Cell sheet grafts with genetically engineered properties to prolong stem cell survival and promote blood vessel networks integrated with pre-existing coronary may provide a potential approach to repair dead or injured myocardium (19). The present study suggests that EPCs may be efficiently transfected with lentiviral vectors encoding Dll-4 without any adverse effect on the cell viability and that EPCDll-4+ exhibit a higher resistance against oxidative stress and the transfected EPCs survive for a longer period of time in the ischemic area.

Vascular network formation is coordinated by VEGF and Dll-4. Dll-4 may act downstream of VEGF as a ‘brake’ on VEGF-mediated angiogenic sprouting (20) and it may act to prevent overexuberant angiogenic sprouting, promoting the timely formation of a well-differentiated vascular network (21). Inhibition of Dll-4 induces the proliferation of immature vascular networks and results in poor tissue perfusion (1113). The current in vitro study demonstrated that the number of tubular structures in the EPCDll-4+ group was higher compared with the EPC and EPCnull groups. In in vivo experiments, a significantly greater capacity was observed when productive vessels were formed and further increased blood flow to the infarcted and peri-infarcted zone in the groups receiving EPCDll-4+, with the exception of the group transplanted with untreated EPCs. Two weeks following cell transplantation, the cardiac function in the Dll-4-overexpressing EPCs group gradually recovered, whereas the PBS and untreated EPCs groups did not exhibit such effects, suggesting that tubulogenesis and enhancement of tissue perfusion played a key role in the improvement of cardiac function in MI animals. EPCDll-4+ also promotes the formation of mature vessels in the ischemic myocardium and enhances cardiac function. Thus, treated EPCs are hypothesized to be superior to the untreated EPCs. By contrast, specific knockdown of Dll-4 attenuates the ability of EPCs to form mature vascular structures in the in vitro angiogenesis assay. Injection of Dll-4-inhibited EPCs into MI animals, resulted in the formation of a dense capillary network, which appeared irregular and disorganized, thus, unable to supply ischemic myocardium with adequate perfusion. These observations were consistent with a previous study showing that Dll-4 blockade causes functional defects in angiogenesis following ischemia in mouse limp (22).

With the exception of Notch, the signaling pathways of mTOR are also closely linked to stem cell development and neovascularization. In ECs, mTOR may be necessary for EPC development since inhibition of mTOR pathways with rapamycin may lead to EPC death, which may result from inhibiting growth factor signaling (23). As an important component of cardiac tissue protection and regeneration, angiogenesis may be regulated by mTOR (24). Inhibition of mTOR may lead to a sequence of events, including elevated matrix metalloproteinase-1 and the blockade of tissue inhibitor of metalloprotease-3 resulting in impaired angiogenesis (25). Moreover, loss of mTOR activity may also lead to a blockage of endothelial proliferation and angiogenesis (26) as well as proliferation of EPCs (23). In the current study, the expression of Notch downstream target molecule, Hey-1 and the phosphorylated status of mTOR and p70S6K (one of mTOR downstream effectors) were significantly increased under a higher expression of Dll-4 in EPCs. EPCDll-4+transplantation also led to a similar variation as the mouse ischemic myocardium was extracted for western blot analysis. Notably, an opposite effect was observed in in vitro cultered EPCDll-4− and the animals treated with EPCnull. A previous study by Chan et al observed that Notch signals may positively regulate the activity of the mTOR pathway in T-cell acute lymphoblastic leukemia (27). Overexpression of Dll-4 was thus hypothesized to increase Hey-1 expression via interaction with the Notch-1 or -4 receptor (28,29) and in turn activate the mTOR/p70S6K-mediated angiogenesis signaling pathways.

In conclusion, the present study suggests that the intravenous injection of Dll-4-overexpressing EPCs stimulates angiogenesis and tubulogenesis effectively and increases blood flow to the ischemic zone through the activation of Notch/Hey-1/mTOR/p70S6K signaling pathways. The combined strategy of EPCs transplantation with Dll-4 therapy may be proposed as a promising approach for the treatment of ischemic heart disease.

Abbreviations:

EPCs

endothelial progenitor cells

Dll

Delta-like

Jag

Jagged

NICD

Notch intracellular domain

bHLH

basic helix-loop-helix

Hes

hairy/enhancer of split

Hey

Hes-related protein

ECs

endothelial cells

PBS

phosphate-buffered saline

VEGF

vascular endothelial growth factor

shRNAs

short hairpin RNAs

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

MI

myocardial infarction

LV

left ventricular

LVID

left ventricular internal dimensions

LVIDD

LVID in diastole

LVIDS

LVID in systole

EF

ejection fraction

FS

fractional shortening

ANOVA

analysis of variance

mTOR

mammalian target of rapamycin

References

1 

Karamysheva AF: Mechanisms of angiogenesis. Biochemistry (Mosc). 73:751–762. 2008. View Article : Google Scholar

2 

Lawall H, Bramlage P and Amann B: Treatment of peripheral arterial disease using stem and progenitor cell therapy. J Vasc Surg. 53:445–453. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Gimble JM, Bunnell BA and Guilak F: Human adipose-derived cells: an update on the transition to clinical translation. Regen Med. 7:225–235. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Kalka C, Masuda H, Takahashi T, Kalka-Moll WM, Silver M, Kearney M, Li T, Isner JM and Asahara T: Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proc Natl Acad Sci USA. 97:3422–3427. 2000. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Carmeliet P: Mechanisms of angiogenesis and arteriogenesis. Nat Med. 6:389–395. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Artavanis-Tsakonas S, Rand MD and Lake RJ: Notch signaling: cell fate control and signal integration in development. Science. 284:770–776. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Roca C and Adams RH: Regulation of vascular morphogenesis by Notch signaling. Genes Dev. 21:2511–2524. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Leslie JD, Ariza-McNaughton L, Bermange AL, McAdow R, Johnson SL and Lewis J: Endothelial signalling by the Notch ligand Delta-like 4 restricts angiogenesis. Development. 134:839–844. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Phng LK and Gerhardt H: Angiogenesis: a team effort coordinated by notch. Dev Cell. 16:196–208. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Scehnet JS, Jiang W, Kumar SR, et al: Inhibition of Dll4-mediated signaling induces proliferation of immature vessels and results in poor tissue perfusion. Blood. 109:4753–4760. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Noguera-Troise I, Daly C, Papadopoulos NJ, Coetzee S, Boland P, Gale NW, Lin HC, Yancopoulos GD and Thurston G: Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis. Nature. 444:1032–1037. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Ridgway J, Zhang G, Wu Y, et al: Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature. 444:1083–1087. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Liu L, Wen T, Zheng XY, Yang DG, Zhao SP, Xu DY and Lü GH: Remnant-like particles accelerate endothelial progenitor cells senescence and induce cellular dysfunction via an oxidative mechanism. Atherosclerosis. 202:405–414. 2009. View Article : Google Scholar

15 

Chen JJ and Zhou SH: Mesenchymal stem cells overexpressing MiR-126 enhance ischemic angiogenesis via the AKT/ERK-related pathway. Cardiol J. 18:675–681. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Urbich C, Aicher A, Heeschen C, Dernbach E, Hofmann WK, Zeiher AM and Dimmeler S: Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells. J Mol Cell Cardiol. 39:733–742. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Huang F, Zhu X, Hu XQ, et al: Mesenchymal stem cells modified with miR-126 release angiogenic factors and activate Notch ligand Delta-like-4, enhancing ischemic angiogenesis and cell survival. Int J Mol Med. 31:484–492. 2013.PubMed/NCBI

18 

Murohara T: Angiogenesis and vasculogenesis for therapeutic neovascularization. Nagoya J Med Sci. 66:1–7. 2003.PubMed/NCBI

19 

Deuse T, Peter C, Fedak PW, et al: Hepatocyte growth factor or vascular endothelial growth factor gene transfer maximizes mesenchymal stem cell-based myocardial salvage after acute myocardial infarction. Circulation. 120(Suppl 11): S247–S254. 2009. View Article : Google Scholar

20 

Suchting S, Freitas C, le Noble F, Benedito R, Bréant C, Duarte A and Eichmann A: The Notch ligand Delta-like 4 negatively regulates endothelial tip cell formation and vessel branching. Proc Natl Acad Sci USA. 104:3225–3230. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Lobov IB, Renard RA, Papadopoulos N, Gale NW, Thurston G, Yancopoulos GD and Wiegand SJ: Delta-like ligand 4 (Dll4) is induced by VEGF as a negative regulator of angiogenic sprouting. Proc Natl Acad Sci USA. 104:3219–3224. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Al Haj Zen A, Oikawa A, Bazan-Peregrino M, Meloni M, Emanueli C and Madeddu P: Inhibition of delta-like-4-mediated signaling impairs reparative angiogenesis after ischemia. Circ Res. 107:283–293. 2010.PubMed/NCBI

23 

Miriuka SG, Rao V, Peterson M, Tumiati L, Delgado DH, Mohan R, Ramzy D, Stewart D, Ross HJ and Waddell TK: mTOR inhibition induces endothelial progenitor cell death. Am J Transplant. 6:2069–2079. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Maiese K, Chong ZZ, Shang YC and Hou J: FoxO proteins: cunning concepts and considerations for the cardiovascular system. Clin Sci (Lond). 116:191–203. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Lemaitre V, Dabo AJ and D’Armiento J: Cigarette smoke components induce matrix metalloproteinase-1 in aortic endothelial cells through inhibition of mTOR signaling. Toxicol Sci. 123:542–549. 2011. View Article : Google Scholar

26 

Humar R, Kiefer FN, Berns H, Resink TJ and Battegay EJ: Hypoxia enhances vascular cell proliferation and angiogenesis in vitro via rapamycin (mTOR)-dependent signaling. FASEB J. 16:771–780. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Chan SM, Weng AP, Tibshirani R, Aster JC and Utz PJ: Notch signals positively regulate activity of the mTOR pathway in T-cell acute lymphoblastic leukemia. Blood. 110:278–286. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Shutter JR, Scully S, Fan W, Richards WG, Kitajewski J, Deblandre GA, Kintner CR and Stark KL: Dll4, a novel Notch ligand expressed in arterial endothelium. Genes Dev. 14:1313–1318. 2000.PubMed/NCBI

29 

Hellström M, Phng LK, Hofmann JJ, et al: Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature. 445:776–780. 2007.PubMed/NCBI

Related Articles

Journal Cover

November 2013
Volume 8 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang H, Huang F and Huang JP: Transplantation of bone marrow‑derived endothelial progenitor cells overexpressing Delta‑like‑4 enhances functional neovascularization in ischemic myocardium. Mol Med Rep 8: 1556-1562, 2013
APA
Huang, H., Huang, F., & Huang, J. (2013). Transplantation of bone marrow‑derived endothelial progenitor cells overexpressing Delta‑like‑4 enhances functional neovascularization in ischemic myocardium. Molecular Medicine Reports, 8, 1556-1562. https://doi.org/10.3892/mmr.2013.1657
MLA
Huang, H., Huang, F., Huang, J."Transplantation of bone marrow‑derived endothelial progenitor cells overexpressing Delta‑like‑4 enhances functional neovascularization in ischemic myocardium". Molecular Medicine Reports 8.5 (2013): 1556-1562.
Chicago
Huang, H., Huang, F., Huang, J."Transplantation of bone marrow‑derived endothelial progenitor cells overexpressing Delta‑like‑4 enhances functional neovascularization in ischemic myocardium". Molecular Medicine Reports 8, no. 5 (2013): 1556-1562. https://doi.org/10.3892/mmr.2013.1657