Experimental study of endothelial progenitor cells labeled with superparamagnetic iron oxide in vitro

  • Authors:
    • Meng‑Qi Wei
    • Di‑Di Wen
    • Xiao‑Ying Wang
    • Yi Huan
    • Yong  Yang
    • Jian Xu
    • Kang Cheng
    • Min‑Wen Zheng
  • View Affiliations

  • Published online on: December 22, 2014     https://doi.org/10.3892/mmr.2014.3122
  • Pages: 3814-3819
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endothelial progenitor cells (EPCs) have an essential role in counteracting risk factor‑induced endothelial injury and protecting against the development of vascular injury, such as myocardial infarction. Magnetic resonance imaging (MRI) was reported to be effective in tracking transplanted stem cells following cell‑labeling with superparamagnetic iron oxide (SPIO) nanoparticles. SPIO has previously been used to label and track EPCs; however, the safest concentration of SPIO for labeling EPCs on a cellular level has remained to be elucidated. In addition, the optimum number of SPIO‑labeled cells required to produce the highest quality magnetic resonance images has not yet been determined. In the present study, EPCs were isolated from the bone marrow of minipigs using density gradient centrifugation. Their biological activity was then studied using flow cytometric analysis. Cells were incubated at different concentrations of SPIO for different durations and then the growth curve, apoptosis, morphology and labeling efficiency of the EPCs were detected using optical and electron microscopy. T2‑weighted fast spin‑echo (T2WITSE) MRI of the different numbers of SPIO‑labeled EPCs (35 µg/ml) were then obtained in axial and sagittal planes. The results of the present study demonstrated that EPCs were efficiently labeled with SPIO, with a labeling efficiency in each group of ~100% following incubation for 24 h. SPIO was found to be localized in the endosomal vesicles of EPCs, which was confirmed by electron microscopy. When the concentration of SPIO was <70 µg/ml, no significant differences were observed in cell viability, proliferative capability (P>0.05) and morphology between labeled and unlabeled EPCs. Furthermore, the T2WITSE signal intensity was significantly decreased in the groups of 5.0x105/ml and 1.0x105/ml compared with that of the control (P<0.05). In conclusion, the results of the present study indicated that 35 µg/ml was the most effective concentration of SPIO to label EPCs in vitro and acquire a high quality MRI. These findings may therefore contribute to the development of a promising novel therapeutic method for the treatment of myocardial infarction following autograft with SPIO‑labeled EPCs in vivo.

Introduction

Endothelial progenitor cells (EPCs) have the ability to differentiate ex vivo into endothelial-phenotyped cells and were initially detected in the peripheral circulation in 1997 via the isolation of cells expressing the CD34 antigen (1,2). EPCs develop into endothelial cells (ECs) during embryogenesis and human hematopoietic stem cytogenesis (3,4). It has also been reported that hemangioblasts were the multipotent precursor cells of EPCs and hematopoietic stem cells (HSCs). EPCs are known to contribute to the growth of vessels and it was reported that they may induce prolonged vascular recovery from ischemia (5,6). In addition, the transplantation of healthy EPCs to compensate for the role of their dysfunctional counterparts demonstrated promising improvements in numerous animal models of ischemic disease (79). EPCs may enter into the circulation by detaching from activated or damaged vessels. Studies have reported the number of circulating EPCs was significantly increased under several pathological conditions that involved vascular injury or instability, including myocardial infarction and cancer (10,11). This therefore suggested that circulating EPCs may have provided an endogenous repair mechanism that counteracted the ongoing risk factor-induced endothelial injury and therefore protected against the development of vascular injuries, such as myocardial infarction (1013). Therefore, the present study hypothesized that changes in circulating levels of EPCs may act as a predictor for the incidence of acute myocardial infarction.

Due to its low toxicity, superparamagnetic iron oxide (SPIO) nanoparticles coated with bioprobes were developed for highly specific labeling of targeted tumors in tumor examination and treatment (1417). Magnetic resonance imaging (MRI) was demonstrated to be effective in tracking transplanted stem cells by labeling cells with superparamagnetic iron oxide (SPIO) nanoparticles (18). The present study investigated the intracellular iron content, labeling efficiency and cell viability of SPIO-labeled EPCs as well as analyzed the MRI results in order to set up a theoretical foundation for the application of autograft EPCs in vivo.

Materials and methods

Cells

The present study was approved by the ethics committee of Xijing Hospital, Fourth Military Medical University (Xi’an, China). EPCs were derived and cultured as previously described (19). In brief, mononuclear cells (MNC) from minipig (the minipig was purchased from the Experimental Animal Center, Forth Military Medical University, Xi’an, China) bone marrow were first isolated using density gradient centrifugation. MNCs were plated on un-coated tissue culture flasks at a density of 1×106/ml in Dulbecco’s modified Eagle’s medium (DMEM; HyClone, Thermo Fisher Scientific, Waltham, MA, USA) containing 10% fetal bovine serum (FBS; HyClone). Non-adherent cells were then collected and plated on culture flasks following four days of culture and the medium was replaced. Following a further 24 h of culture, non-adherent cells were collected and seeded into culture flasks at a density of 1×106/ml in DMEM supplemented with 10% FBS, vascular endothelial growth factor (VEGF; 10 ng/ml; PeproTech EC Ltd, London, UK) and basic fibroblast growth factor (bFGF; 10 ng/ml; PeproTech EC Ltd). Cells were then maintained at 37°C and 5% CO2. Media was observed daily and changed every two to three days.

Fluorescence-activated cell sorting (FACS) analysis

Flow cytometric staining and analyses were performed as previously described (20). In brief, EPCs were resuspended in 100 ml rinsing buffer and incubated with the following monoclonal antibodies: Phycoerythrin (PE)-conjugated mouse anti-human CD31 (1:50), fetal liver kinase (Flk)-1 (1:100) and factor VIII (1:16,000) (Santa Cruz Biotechnology, Inc, Dallas, TX, USA) for 30 min at room temperature. Following washing with phosphate-buffered saline (PBS; pH 7.35), the expression of membranous antigen on the cells was detected using a FACSCaliburTM flow cytometer (BD Biosciences, San Jose, CA, USA) equipped with the Cell Quest software (BD Biosciences). Flow cytometric data were analyzed using appropriate controls (cat no.: 1-001-A; R&D Systems, Inc., Minneapolis, MN, USA) with isotype-matched immunoglobulin G and unstained controls.

SPIO labeling in vitro

Concentrations of SPIO (Bayer Healthcare Pharmaceuticals, Montville, NJ, USA) in cell culture medium were as follows: Group 1, 17.5 μg/ml; group 2, 35 μg/ml; group 3, 70 μg/ml; and group 4, 140 μg/ml, the control group consisted of DMEM without SPIOs. EPCs labeled with SPIO were incubated at 37°C and 5% CO2 atmosphere, EPCs were then washed with culture DMEM and subsequently used for in vitro studies. Following incubation for one day, the growth and morphology of EPCs in culture were observed daily.

Cell viability

Following incubation for one week, all EPCs were washed three times with DMEM, trypsinized, counted and then resuspended. Cells from each group (18 wells/group) were initially seeded in un-coated 96-well plates and cultured in 37°C and 5% CO2. Following incubation for a further 24 h, cells from each group were added to 500 μl MTT (Beyotime Institue of Biotechnology, Haimen, China) and assessed using a standard MTT assay for 4 h. Supernatant fluid was then discarded and 150 μl dimethylsulfoxide (DMSO) was added to each well for 10 min with agitation. The light absorption of cells was measured using an ELISA reader (iMark; Bio-Rad Laboratories, Inc., Hercules, CA, USA). Finally a MTT bar graph was drawn.

Electron microscopy

In order to detect the iron concentration within EPCs and observe the morphology of EPCs in culture, SPIO-labeled EPCs were grown in a six-well tissue culture plate until they reached 80% confluence. Following incubation, the medium was removed and the plate was gently washed twice with sterile PBS. Samples were then digested using trypsin, transferred into a tube and centrifuged at 1,500 × g for 15 min. The supernatant was removed and cells were fixed with 4% paraformaldehyde and visualized using an electron microscope (IX83; Olympus Corp., Tokyo, Japan).

Magnetic resonance imaging of SPIO-labeled EPCs in vitro

Different concentrations of SPIO-labeled EPC solution (SPIO, 35 μg/ml) were collected by removing the free SPIO and washed three times with PBS. The EPCs were suspended in 1% agarose prior to being transferred into 1.5-ml microcentrifuge tubes. Concentrations of SPIO-labeled EPC cells were as follows: Group a, control; group b, 1.0×104/ml; group c, 5.0×104/ml; group d, 1.0×105/ml; and group e, 5.0×105/ml. In vitro MRI of the tubes was then conducted.

MRI scans were performed on a clinical 3.0T whole-body MRI system (MAGNETOMTrioTim; Siemens Healthcare, Erlangen, Germany) following 24 h culture. T2-weighted fast spin-echo (T2WITSE) MRI measurements were initially obtained in axial and sagittal planes. Imaging parameters for these images were: Repetition time/echo time, 2652110 ms; flip angle, 90°; field of view, 210 mm; slice thickness, 3 mm; matrix size, 205×256.

Statistical analysis

Statistical analysis was performed using SPSS (version 13.0; SPSS Inc., Chicago, IL, USA). Values are expressed as the mean ± standard deviation and analyzed using the one-way analysis of variance. P<0.05 was considered to indicate a statistically significant difference between values.

Results

FACS analysis confirms the identity of EPCs

A large number of molecular markers were reported to be associated with EPCs (2123); in the present study, several of these molecular markers were detected using flow cytometry in order to identify EPCs. The results of the FACS analysis revealed EPCs were positive for the markers CD31, Flk-1 and factor VIII in 83.52, 85.34 and 83.86% of cells, respectively (Fig. 1). Thus these cells were identified as EPCs.

Optical microscopy confirms the labeling efficiency of SPIO

Endocytosis of SPIO particles was observed in EPCs using optical microscopy. Following 6 h of incubation of EPCs with 35 μg/ml SPIO, characteristic granular SPIO particles were detected within the cytoplasm (Fig. 2). Following 24 h, the SPIO labeling efficiency increased to 100% in all groups and the endocytotic rate of SPIO particles positively correlated with the concentration of SPIO. However, no significant difference was found among the labeling efficiencies of the groups (Fig. 3). Furthermore, characteristic intracytoplasmic granular SPIO particles were observed following 28 days in culture (data not shown).

Cell growth and viability of EPCs labeled with different concentrations of SPIO

Fig. 4 shows the growth curve of EPC labeling with different concentration of SPIO (17.5, 35, 70 and 140 μg/ml, respectively) following one week in culture. In groups 1, 2 and 3, the trends of cell growth were not significantly different to those of the control group (P>0.05); in addition, MTT analysis of cell viability revealed no significant difference between the absorbance (490 nm) of groups 1, 2 and 3 compared with that of the control (P>0.05) (Fig. 5). However, the cell growth and viability were markedly suppressed in the fourth group (140 μg/ml) as well as cell growth for group 3 (70 μl/mg).

Morphological changes of EPCs labeled with SPIO

Following one week in culture, electron microscopy revealed micorvilli on the surface of control cells as well as numerous intracellular organelles and prominent nuclear euchromatin with sharp nucleoli (Fig. 6A). EPCs labeled with SPIO at concentrations of 17.5 μg/ml and 35 μg/ml revealed identical characteristics, including figure, shape and nucleolus structure, compared to those of the control group (Fig. 6B and C). All concentrations of SPIO-labeled EPCs contained endolysosomal iron particles, whereas these were not observed in EPCs of the control group (Fig. 6B–D); in addition, EPCs labeled with 70 μg/ml SIPO exhibited an increased number of endolysosomes (Fig. 6E). However, EPCs labeled with 140 μg/ml SIPO underwent apoptotic cell death; morphologically, EPCs in this group demonstrated a reduced quantity of microvilli, a highly concentrated cytoplasm and the nucleus was located at the edge of the nuclear membrane (Fig. 6F). These results indicated that 35 μg/ml SPIO was a safe concentration for EPC-labeling, without affecting the biological characteristics of cells.

In vitro MRI

EPCs were labeled with 35 μg/ml SPIO and MRI was performed on different concentrations of SPIO-labeled cells (control, 1.0×104, 5.0×104, 1.0×105 and 5.0×105/ml) in vitro (Fig. 7A). As shown in Fig. 7B, the T2WI signal intensity of SPIO-labeled EPCs decreased with increasing concentration of EPCs; in addition, 5×104/ml SPIO-labeled EPCs was the lowest concentration of cells observed within the imaging parameters. Furthermore, the T2WI signal intensity significantly decreased at labeled cell concentrations of 5.0×105/ml and 1.0×105/ml compared with that of the control group (P<0.05).

Discussion

EPCs are widely considered to be an effective therapeutic agent for the treatment of certain vascular diseases (24). Numerous studies have confirmed that EPCs may be used as an alternative cell-based approach for the enhancement of angio- and vasculogenic responses (25). Therefore, it was hypothesized that autograft EPCs may yield promising improvements in various animal models of ischemic disease. The MRI technique has numerous advantages, including a wide variety of imaging sequences, high resolution and improved soft-tissue contrast without radiation damage, which suggested its potential use for monitoring transplanted cells (26). However, the traditional MRI was not able to differentiate the transplanted stem cells from the histiocytes; therefore, in order improve the contrast of the cells using MRI, the transplanted cells required modification. SPIO has been widely used as a negative marker to label cells (27,28). In order to accommodate the requirements of preoperative and intraoperative examinations using simple SPIO without additional indicators, the superior magnetic characteristics of SPIO required investigation to ensure its safety in vivo. In the present study, the reaction time, ratio and appropriate concentrations of SPIO and SPIO-labeled EPCs were calculated and analyzed. Previous studies confirmed that the absorption of iron particles had a positive correlation with cell number, SPIO concentration and the time of incubation (21). The results of the present study demonstrated that following 6 h of incubation with SPIO iron particles were phagocytized in all groups and labeling efficiency reached 100% in the group labeled with 140 μg/ml SPIO, while lower concentrations of SPIO reached 100% efficiency following 12, 18–24 and 30–36 h, respectively. This therefore indicated that the number of iron particles absorbed by EPCS increased with incubation time.

Arbab et al (29) and Himes et al (30) previously confirmed that the SPIO at concentrations <50 μg/ml produced no side effects on cell activity. The results of the present study indicated that cell growth was unaffected by concentrations of SPIO <70 μg/ml; however, in EPCs labeled with 70 μg/ml SPIO, lysosome enhancement was observed and concentrations of SPIO >70 μg/ml suppressed the biological activity of the cells. These results therefore indicated that it was safe to label EPCs with SPIO at concentrations of 20–70 μg/ml. In addition, the reaction time of cells was elongated at concentrations of SPIO <20 μg/ml; therefore, 35 μg/ml SPIO was selected to label target EPCs for subsequent experiments.

Due to the paramagnetism of SPIO, the T2WI signal significantly decreased during MRI, which was consistent with the effects of the negative contrast agent. As the cell number increased, the contrast effect was enhanced, which was thought to be due to the increased number of cells available to absorb the iron particles. In vitro MRI demonstrated that 5×104/ml was the lowest observable concentration of SPIO-labeled EPCs.

In conclusion, the results of the present study indicated that MRI was able to reflect changes in concentrations of intracellular iron and therefore has the potential for use in studying changes in SPIO-labeled EPCs in vivo for the treatment of myocardial infarction. However, further studies are required in order to determine the effect of SPIO-labeling of EPCs in vivo.

Acknowledgements

The present study was supported by grants from the National Science Foundation (nos. 81201135 and 30370821).

References

1 

Asahara T, Murohara T, Sullivan A, et al: Isolation of putative progenitor endothelial cells for angiogenesis. Science. 275:964–967. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Ribatti D: The involvement of endothelial progenitor cells in tumor angiogenesis. J Cell Mol Med. 8:294–300. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Murasawa S and Asahara T: Endothelial progenitor cells for vasculogenesis. Physiology (Bethesda). 20:36–42. 2005. View Article : Google Scholar

4 

Schmidt-Lucke C, Rössing L, Fichtlscherer S, et al: Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair. Circulation. 111:2981–2987. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Losordo DW and Dimmeler S: Therapeutic angiogenesis and vasculogenesis for ischemic disease: part II: cell-based therapies. Circulation. 109:2692–2697. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Tongers J, Roncalli JG and Losordo DW: Role of endothelial progenitor cells during ischemia-induced vasculogenesis and collateral formation. Microvasc Res. 79:200–206. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Kawamoto A, Katayama M, Handa N, et al: Intramuscular transplantation of G-CSF-mobilized CD34(+) cells in patients with critical limb ischemia: a phase IIIa, multicenter, single-blinded, dose-escalation clinical trial. Stem Cells. 27:2857–2864. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Cho SW, Moon SH, Lee SH, et al: Improvement of postnatal neovascularization by human embryonic stem cell derived endothelial-like cell transplantation in a mouse model of hindlimb ischemia. Circulation. 116:2409–2419. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Losordo DW, Schatz RA, White CJ, et al: Intramyocardial transplantation of autologous CD34+ stem cells for intractable angina: a phase IIIa double-blind, randomized controlled trial. Circulation. 115:3165–3172. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Thal MA, Krishnamurthy P, Mackie AR, et al: Enhanced angiogenic and cardiomyocyte differentiation capacity of epigenetically reprogrammed mouse and human endothelial progenitor cells augments their efficacy for ischemic myocardial repair. Circ Res. 111:180–190. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Giannoni E, Taddei ML, Parri M, et al: EphA2-mediated mesenchymal-amoeboid transition induced by endothelial progenitor cells enhances metastatic spread due to cancer-associated fibroblasts. J Mol Med (Berl). 91:103–115. 2013. View Article : Google Scholar

12 

Yin M, Liao Z, Yuan X, et al: Polymorphisms of the vascular endothelial growth factor gene and severe radiation pneumonitis in non-small cell lung cancer patients treated with definitive radiotherapy. Cancer Sci. 103:945–950. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Liu Y, Xia T, Zhang W, et al: Variations of circulating endothelial progenitor cells and transforming growth factor-beta-1 (TGF-β1) during thoracic radiotherapy are predictive for radiation pneumonitis. Radiat Oncol. 8:1892013. View Article : Google Scholar

14 

Yang SY, Sun JS, Liu CH, et al: Ex vivo magnetofection with magnetic nanoparticles: a novel platform for nonviral tissue engineering. Artif Organs. 32:195–204. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Wu CC, Lin LY, Lin LC, et al: Bio-functionalized magnetic nanoparticles for in vitro labeling and in vivo locating specific biomolecules. Appl Phys Lett. 92:1425042008. View Article : Google Scholar

16 

Oghabian MA, Gharehaghaji N, Amirmohseni S, et al: Detection sensitivity of lymph nodes of various sizes using USPIO nanoparticles in magnetic resonance imaging. Nanomedicine. 6:496–499. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Müller S: Magnetic fluid hyperthermia therapy for malignant brain tumors - an ethical discussion. Nanomedicine. 5:387–393. 2009. View Article : Google Scholar

18 

Gazeau F and Wilhelm C: Magentic labeling, imaging and manipulation of endothelial progenitor cells using iron oxide nanoparticles. Future Med Chem. 2:397–408. 2010. View Article : Google Scholar

19 

Cheng K, Wei MQ, Jia GL, et al: Effects of metoprolol and small intestine RNA on marrow-derived endothelial progenitor cells applied for autograft transplantation in heart disease. Eur Rev Med Pharmacol Sci. 18:1666–1673. 2014.PubMed/NCBI

20 

Cho SW, Moon SH, Lee SH, et al: Improvement of postnatal neovascularization by human embryonic stem cell derived endothelial like cell transplantation in a mouse model of hind limb ischemia. Circulation. 116:2409–2419. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Gill M, Dias S, Hattori K, et al: Vascular trauma induces rapid but transient mobilization of VEGFR2(+)AC133(+) endothelial precursor cells. Circ Res. 88:167–174. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Gehling UM, Ergün S, Schumacher U, et al: In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood. 95:3106–3112. 2000.PubMed/NCBI

23 

Peichev M, Naiyer AJ, Pereira D, et al: Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood. 95:952–958. 2002.

24 

Moon SH, Kim SM, Park SJ, et al: Development of a xeno-free autologous culture system for endothelial progenitor cells derived from human umbilical cord blood. PLoS One. 8:e752242013. View Article : Google Scholar : PubMed/NCBI

25 

Rosell A, Morancho A, Navarro-Sobrino M, et al: Factors secreted by endothelial progenitor cells enhance neurorepair responses after cerebral ischemia in mice. PLoS One. 8:e732442013. View Article : Google Scholar : PubMed/NCBI

26 

Sun JH, Zhang YL, Nie CH, et al: In vitro labeling of endothelial progenitor cells isolated from peripheral blood with superparamagnetic iron oxide nanoparticles. Mol Med Rep. 6:282–286. 2012.PubMed/NCBI

27 

Zhang B, Li Q, Yin P, et al: Ultrasound-triggered BSA/SPION hybrid nanoclusters for liver-specific magnetic resonance imaging. ACS Appl Mater Interfaces. 4:6479–6486. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Yoo MK, Park IK, Lim HT, et al: Folate-PEG-superparamagnetic iron oxide nanoparticles for lung cancer imaging. Acta Biomater. 8:3005–3013. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Arbab AS, Bashaw LA, Miller BR, et al: Intracytoplasmic tagging of cells with ferumoxides and transfection agent for cellular magnetic resonance imaging after cell transplantation: methods and techniques. Transplantation. 76:1123–1130. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Himes N, Min JY, Lee R, et al: In vivo MRI of embryonic stem cells in a mouse model of myocardial infarction. Magn Reson Med. 52:1214–1219. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 11 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
Wei MQ, Wen DD, Wang XY, Huan Y, Yang Y, Xu J, Cheng K and Zheng MW: Experimental study of endothelial progenitor cells labeled with superparamagnetic iron oxide in vitro. Mol Med Rep 11: 3814-3819, 2015
APA
Wei, M., Wen, D., Wang, X., Huan, Y., Yang, Y., Xu, J. ... Zheng, M. (2015). Experimental study of endothelial progenitor cells labeled with superparamagnetic iron oxide in vitro. Molecular Medicine Reports, 11, 3814-3819. https://doi.org/10.3892/mmr.2014.3122
MLA
Wei, M., Wen, D., Wang, X., Huan, Y., Yang, Y., Xu, J., Cheng, K., Zheng, M."Experimental study of endothelial progenitor cells labeled with superparamagnetic iron oxide in vitro". Molecular Medicine Reports 11.5 (2015): 3814-3819.
Chicago
Wei, M., Wen, D., Wang, X., Huan, Y., Yang, Y., Xu, J., Cheng, K., Zheng, M."Experimental study of endothelial progenitor cells labeled with superparamagnetic iron oxide in vitro". Molecular Medicine Reports 11, no. 5 (2015): 3814-3819. https://doi.org/10.3892/mmr.2014.3122