Optimal labeling dose, labeling time, and magnetic resonance imaging detection limits of ultrasmall superparamagnetic iron-oxide nanoparticle labeled mesenchymal stromal cells

Anders Bruun Mathiasen, Louise Hansen, Tina Friis, Carsten Thomsen, Kishore Bhakoo, Jens Kastrup, Anders Bruun Mathiasen, Louise Hansen, Tina Friis, Carsten Thomsen, Kishore Bhakoo, Jens Kastrup

Abstract

Background. Regenerative therapy is an emerging treatment modality. To determine migration and retention of implanted cells, it is crucial to develop noninvasive tracking methods. The aim was to determine ex vivo magnetic resonance imaging (MRI) detection limits of ultrasmall superparamagnetic iron-oxide (USPIO) labeled mesenchymal stromal cells (MSCs). Materials and Methods. 248 gel-phantoms were constructed and scanned on a 1.5T MRI-scanner. Phantoms contained human MSCs preincubated with USPIO nanoparticles for 2, 6, or 21 hours using 5 or 10 μ g USPIO/10(5) MSCs. In addition, porcine hearts were scanned after injection of USPIO labeled MSCs. Results. Using 21 h incubation time and 10 μ g USPIO/10(5) MSCs, labeled cells were clearly separated from unlabeled cells on MRI using 250.000 (P < 0.001), 500.000 (P = 0.007), and 1.000.000 MSCs (P = 0.008). At lower incubation times and doses, neither labeled nor unlabeled cells could be separated. In porcine hearts labeled, but not unlabeled, MSCs were identified on MRI. Conclusions. As few as 250.000 MSCs can be detected on MRI using 21 h incubation time and 10 μ g USPIO/10(5) MSCs. At lower incubation times and doses, several million cells are needed for MRI detection. USPIO labeled cells can be visualized by MRI in porcine myocardial tissue.

Figures

Figure 1
Figure 1
MRI phantoms. (a) Two phantoms containing USPIO labeled cells. (b) MRI image of 2 phantoms with an ellipsoid region of interest placed in the upper phantom. USPIO: ultrasmall superparamagnetic iron-oxide.
Figure 2
Figure 2
Cellular iron content. The iron content per cell was determined in unlabeled MSC and MSC incubated with half or full dose USPIO for 2, 6, and 21 hours. MSC: mesenchymal stromal cells. USPIO: ultrasmall superparamagnetic iron-oxide. USPIO dose—full: 10 μg per 105 cells; half: 5 μg per 105 cells.
Figure 3
Figure 3
Absolute phantom MRI intensities after 21 hour USPIO incubation. MRI intensities are absolute mean values. USPIO: ultrasmall superparamagnetic iron-oxide. USPIO dose: full = 10 μg per 105 cells; half = 5 μg per 105 cells.
Figure 4
Figure 4
Phantoms intensity differences after 21-hour USPIO incubation. MRI intensity differences are the mean numeric difference between absolute MRI intensities of phantoms and reference gels. USPIO = ultrasmall superparamagnetic iron-oxide. USPIO dose: full = 10 μg per 105 cells; half = 5 μg per 105 cells.
Figure 5
Figure 5
MRI images of porcine myocardium before and after USPIO labeled MSC injection. T2*-images of porcine myocardium before injection (a1–a5) and after injection (b1–b5) of USPIO-labeled MSCs. USPIO labeled MSCs are identified as hypointense areas (arrows). MSCs: mesenchymal stromal cells. USPIO: ultrasmall superparamagnetic iron-oxide.
Figure 6
Figure 6
MRI images of porcine myocardium before and after unlabeled MSC injection. T2*-images of porcine myocardium before injection (a1–a5) and after injection (b1–b5) of unlabeled MSCs. Unlabeled cells cannot be identified. MSCs: mesenchymal stromal cells. USPIO: ultrasmall superparamagnetic iron-oxide.

References

    1. Friis T, Haack-Sørensen M, Mathiasen AB, et al. Mesenchymal stromal cell derived endothelial progenitor treatment in patients with refractory angina. Scandinavian Cardiovascular Journal. 2011;45(3):161–168.
    1. Mathiasen AB, Haack-Sørensen M, Kastrup J. Mesenchymal stromal cells for cardiovascular repair: current status and future challenges. Future Cardiology. 2009;5(6):605–617.
    1. Kastrup J. Stem cells therapy for cardiovascular repair in ischemic heart disease: how to predict and secure optimal outcome? EPMA Journal. 2011;2(1):107–117.
    1. Zhang WY, Ebert AD, Narula J, Wu JC. Imaging cardiac stem cell therapy: translations to human clinical studies. Journal of Cardiovascular Translational Research. 2011;4(4):514–522.
    1. Bulte JWM. In vivo MRI cell tracking: clinical studies. American Journal of Roentgenology. 2009;193(2):314–325.
    1. Amsalem Y, Mardor Y, Feinberg MS, et al. Iron-oxide labeling and outcome of transplanted mesenchymal stem cells in the infarcted myocardium. Circulation. 2007;116(11):I38–I45.
    1. Bulte JWM, Kostura L, Mackay A, et al. Feridex-labeled mesenchymal stem cells: cellular differentiation and MR assessment in a canine myocardial infarction model. Academic Radiology. 2005;12(5):S2–S6.
    1. Kraitchman DL, Heldman AW, Atalar E, et al. In vivo magnetic resonance imaging of mesenchymal stem cells in myocardial infarction. Circulation. 2003;107(18):2290–2293.
    1. Ma GS, Qi CM, Liu NF, et al. Efficiently tracking of stem cells in vivo using different kinds of uperparamagnetic iron oxide in swine with myocardial infarction. Chinese Medical Journal. 2011;124(8):1199–1204.
    1. Yang CY, Hsiao JK, Tai MF, et al. Direct labeling of hMSC with SPIO: the long-term influence on toxicity, chondrogenic differentiation capacity, and intracellular distribution. Molecular Imaging and Biology. 2011;13(3):443–451.
    1. Graham JJ, Foltz WD, Vaags AK, et al. Long-term tracking of bone marrow progenitor cells following intracoronary injection post-myocardial infarction in swine using MRI. American Journal of Physiology. 2010;299(1):H125–H133.
    1. Peng C, Yang K, Xiang P, et al. Effect of transplantation with autologous bone marrow stem cells on acute myocardial infarction. International Journal of Cardiology. 2013;162(3):158–165.
    1. Chapon C, Jackson JS, Aboagye EO, Herlihy AH, Jones WA, Bhakoo KK. An in vivo multimodal imaging study using MRI and pet of stem cell transplantation after myocardial infarction in rats. Molecular Imaging and Biology. 2009;11(1):31–38.
    1. Callera F, De Melo CMTP. Magnetic resonance tracking of magnetically labeled autologous bone marrow CD34+ cells transplanted into the spinal cord via lumbar puncture technique in patients with chronic spinal cord injury: CD34+ cells’ migration into the injured site. Stem Cells and Development. 2007;16(3):461–466.
    1. De Vries IJM, Lesterhuis WJ, Barentsz JO, et al. Magnetic resonance tracking of dendritic cells in melanoma patients for monitoring of cellular therapy. Nature Biotechnology. 2005;23(11):1407–1413.
    1. Karussis D, Karageorgiou C, Vaknin-Dembinsky A, et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Archives of Neurology. 2010;67(10):1187–1194.
    1. Nighoghossian N, Wiart M, Cakmak S, et al. Inflammatory response after ischemic stroke: a USPIO-enhanced MRI study in patients. Stroke. 2007;38(2):303–307.
    1. Toso C, Vallee JP, Morel P, et al. Clinical magnetic resonance imaging of pancreatic islet grafts after iron nanoparticle labeling. American Journal of Transplantation. 2008;8(3):701–706.
    1. Zhu J, Zhou L, XingWu F. Tracking neural stem cells in patients with brain trauma. The New England Journal of Medicine. 2006;355(22):2376–2378.
    1. Terrovitis J, Stuber M, Youssef A, et al. Magnetic resonance imaging overestimates ferumoxide-labeled stem cell survival after transplantation in the heart. Circulation. 2008;117(12):1555–1562.
    1. Stuckey DJ, Carr CA, Martin-Rendon E, et al. Iron particles for noninvasive monitoring of bone marrow stromal cell engraftment into, and isolation of viable engrafted donor cells from, the heart. Stem Cells. 2006;24(8):1968–1975.
    1. Yang K, Xiang P, Zhang C, et al. Magnetic resonance evaluation of transplanted mesenchymal stem cells after myocardial infarction in swine. Canadian Journal of Cardiology. 2011;27(6):818–825.
    1. Tran N, Li Y, Maskali F, et al. Short-term heart retention and distribution of intramyocardial delivered mesenchymal cells within necrotic or intact myocardium. Cell Transplantation. 2006;15(4):351–358.
    1. Song M, Woo KM, Kim Y, Lim D, Song IC, Yoon BW. Labeling efficacy of superparamagnetic iron oxide nanoparticles to human neural stem cells: comparison of ferumoxides, monocrystalline iron oxide, cross-linked iron oxide (CLIO)-NH2 and tat-CLIO. Korean Journal of Radiology. 2007;8(5):365–371.
    1. Bonnemain B. Nanoparticles: the industrial viewpoint. Applications in diagnostic imaging. Annales Pharmaceutiques Francaises. 2008;66(5-6):263–267.
    1. Wunderbaldinger P, Josephson L, Weissleder R. Crosslinked iron oxides (CLIO): a new platform for the development of targeted MR contrast agents. Academic Radiology. 2002;9(2):S304–S306.
    1. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–317.
    1. Josephson L, Tung CH, Moore A, Weissleder R. High-efficiency intracellular magnetic labeling with novel superparamagnetic-tat peptide conjugates. Bioconjugate Chemistry. 1999;10(2):186–191.
    1. Jackson J, Chapon C, Jones W, Hirani E, Qassim A, Bhakoo K. In vivo multimodal imaging of stem cell transplantation in a rodent model of Parkinson’s disease. Journal of Neuroscience Methods. 2009;183(2):141–148.
    1. Freyman T, Polin G, Osman H, et al. A quantitative, randomized study evaluating three methods of mesenchymal stem cell delivery following myocardial infarction. European Heart Journal. 2006;27(9):1114–1122.
    1. Hou D, Youssef EAS, Brinton TJ, et al. Radiolabeled cell distribution after intramyocardial, intracoronary, and interstitial retrograde coronary venous delivery: implications for current clinical trials. Circulation. 2005;112(9):I150–I156.
    1. Müller-Ehmsen J, Krausgrill B, Burst V, et al. Effective engraftment but poor mid-term persistence of mononuclear and mesenchymal bone marrow cells in acute and chronic rat myocardial infarction. Journal of Molecular and Cellular Cardiology. 2006;41(5):876–884.
    1. Wang J, Zhang S, Rabinovich B, et al. Human CD34+ cells in experimental myocardial infarction: long-term survival, sustained functional improvement, and mechanism of action. Circulation Research. 2010;106(12):1904–1911.
    1. Dinniwell R, Chan P, Czarnota G, et al. Pelvic lymph node topography for radiotherapy treatment planning from ferumoxtran-10 contrast-enhanced magnetic resonance imaging. International Journal of Radiation Oncology Biology Physics. 2009;74(3):844–851.
    1. Froehlich JM, Triantafyllou M, Fleischmann A, Vermathen P, Thalmann GN, Thoeny HC. Does quantification of USPIO uptake-related signal loss allow differentiation of benign and malignant normal-sized pelvic lymph nodes? Contrast Media and Molecular Imaging. 2012;7(3):346–355.
    1. Thorek DLJ, Tsourkas A. Size, charge and concentration dependent uptake of iron oxide particles by non-phagocytic cells. Biomaterials. 2008;29(26):3583–3590.
    1. Oude Engberink RD, Van Der Pol SMA, Döpp EA, De Vries HE, Blezer ELA. Comparison of SPIO and USPIO for in vitro labeling of human monocytes: MR detection and cell function. Radiology. 2007;243(2):467–474.

Source: PubMed

3
Suscribir