D-mannose-Coating of Maghemite Nanoparticles Improved Labeling of Neural Stem Cells and Allowed Their Visualization by ex vivo MRI after Transplantation in the Mouse Brain

. 2019 May ; 28 (5) : 553-567. [epub] 20190711

Jazyk angličtina Země Spojené státy americké Médium print-electronic

Typ dokumentu časopisecké články, práce podpořená grantem

Perzistentní odkaz   https://www.medvik.cz/link/pmid31293167

Magnetic resonance imaging (MRI) of superparamagnetic iron oxide-labeled cells can be used as a non-invasive technique to track stem cells after transplantation. The aim of this study was to (1) evaluate labeling efficiency of D-mannose-coated maghemite nanoparticles (D-mannose(γ-Fe2O3)) in neural stem cells (NSCs) in comparison to the uncoated nanoparticles, (2) assess nanoparticle utilization as MRI contrast agent to visualize NSCs transplanted into the mouse brain, and (3) test nanoparticle biocompatibility. D-mannose(γ-Fe2O3) labeled the NSCs better than the uncoated nanoparticles. The labeled cells were visualized by ex vivo MRI and their localization subsequently confirmed on histological sections. Although the progenitor properties and differentiation of the NSCs were not affected by labeling, subtle effects on stem cells could be detected depending on dose increase, including changes in cell proliferation, viability, and neurosphere diameter. D-mannose coating of maghemite nanoparticles improved NSC labeling and allowed for NSC tracking by ex vivo MRI in the mouse brain, but further analysis of the eventual side effects might be necessary before translation to the clinic.

Zobrazit více v PubMed

Park KI, Teng YD, Snyder EY. The injured brain interacts reciprocally with neural stem cells supported by scaffolds to reconstitute lost tissue. Nat Biotechnol. 2002;20(11):1111–1117. PubMed

Daadi MM, Steinberg GK. Manufacturing neurons from human embryonic stem cells: biological and regulatory aspects to develop a safe cellular product for stroke cell therapy. Regen Med. 2009;4(2):251–263. PubMed PMC

Bliss T, Guzman R, Daadi M, Steinberg G. Cell transplantation therapy for stroke. Stroke. 2007;38(2 Suppl):817–826. PubMed

Lee J-P, Jeyakumar M, Gonzalez R, Takahashi H, Lee P-J, Baek RC, Clark D, Rose H, Fu G, Clarke J, McKercher S. et al. Stem cells act through multiple mechanisms to benefit mice with neurodegenerative metabolic disease. Nat Med. 2007;13(4):439–447. PubMed

Michelsen KA, Acosta-Verdugo S, Benoit-Marand M, Espuny-Camacho I, Gaspard N, Saha B, Gaillard A, Vanderhaeghen P. Area-specific reestablishment of damaged circuits in the adult cerebral cortex by cortical neurons derived from mouse embryonic stem cells. Neuron. 2015;85(5):982–997. PubMed

Lu P, Woodruff G, Wang Y, Graham L, Hunt M, Wu D, Boehle E, Ahmad R, Poplawski G, Brock J, Goldstein LSB. et al. Long-distance axonal growth from human induced pluripotent stem cells after spinal cord injury. Neuron. 2014;83(4):789–796. PubMed PMC

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. J Neurosci Methods. 2009;183(2):141–148. PubMed

Laver CRJ, Metcalfe AL, Szczygiel L, Yanai A, Sarunic M V., Gregory-Evans K. Bimodal in vivo imaging provides early assessment of stem-cell-based photoreceptor engraftment. Eye. 2015;29(5):681–690. PubMed PMC

Guzman R, Uchida N, Bliss TM, He D, Christopherson KK, Stellwagen D, Capela A, Greve J, Malenka RC, Moseley ME, Palmer TD. et al. Long-term monitoring of transplanted human neural stem cells in developmental and pathological contexts with MRI. Proc Natl Acad Sci U S A. 2007;104(24):10211–10216. PubMed PMC

Guzman R, Bliss T, De Los Angeles A, Moseley M, Palmer T, Steinberg G. Neural progenitor cells transplanted into the uninjured brain undergo targeted migration after stroke onset. J Neurosci Res. 2008;86(4):873–882. PubMed

Hoehn M. How do we assess regenerative success after stem cell implantation? An experimental approach. Regen Med. 2011;6(4):417–419. PubMed

Modo M, Mellodew K, Cash D, Fraser SE, Meade TJ, Price J, Williams SCR. Mapping transplanted stem cell migration after a stroke: a serial, in vivo magnetic resonance imaging study. Neuroimage. 2004;21(1):311–317. PubMed

Zhang ZG, Jiang Q, Zhang R, Zhang L, Wang L, Zhang L, Arniego P, Ho KL, Chopp M. Magnetic resonance imaging and neurosphere therapy of stroke in rat. Ann Neurol. 2003;53(2):259–263. PubMed

Song M, Kim Y, Kim Y, Ryu S, Song I, Kim SU, Yoon BW. MRI tracking of intravenously transplanted human neural stem cells in rat focal ischemia model. Neurosci Res. 2009;64(2):235–239. PubMed

Mishra SK, Khushu S, Singh AK, Gangenahalli G. Homing and tracking of iron oxide labelled mesenchymal stem cells after infusion in traumatic brain injury mice: a longitudinal in vivo MRI study. Stem Cell Rev. 2018;14(6):888–900. PubMed

Berman SC, Galpoththawela C, Gilad AA, Bulte JWM, Walczak P. Long-term MR cell tracking of neural stem cells grafted in immunocompetent versus immunodeficient mice reveals distinct differences in contrast between live and dead cells. Magn Reson Med. 2011;65(2):564–574. PubMed PMC

Modo M, Beech JS, Meade TJ, Williams SCR, Price J. A chronic 1 year assessment of MRI contrast agent-labelled neural stem cell transplants in stroke. Neuroimage. 2009;47(Suppl. 2):133–142. PubMed PMC

Barrow M, Taylor A, Murray P, Rosseinsky MJ, Adams DJ. Design considerations for the synthesis of polymer coated iron oxide nanoparticles for stem cell labelling and tracking using MRI. Chem Soc Rev. 2015;44(19):6733–6748. PubMed

Wang Y-XJ. Superparamagnetic iron oxide based MRI contrast agents: current status of clinical application. Quant Imaging Med Surg. 2011;1(1):35–40. PubMed PMC

Wang Y, Xu C, Ow H. Commercial nanoparticles for stem cell labeling and tracking. Theranostics. 2013;3(8):544–560. PubMed PMC

Delikatny EJ, Poptani H. MR techniques for in vivo molecular and cellular imaging. Radiol Clin North Am. 2005;43(1):205–220. PubMed

Wang EC, Wang AZ. Nanoparticles and their applications in cell and molecular biology. Integr Biol (Camb). 2014;6(1):9–26. PubMed PMC

Lodhia J, Mandarano G, Ferris NJ, Eu P, Cowell SF. Development and use of iron oxide nanoparticles (Part 1): synthesis of iron oxide nanoparticles for MRI. Biomed Imaging Interv J. 2010;6(2):e12. PubMed PMC

Muhammad G, Xu J, Bulte JWM, Jablonska A, Walczak P, Janowski M. Transplanted adipose-derived stem cells can be short-lived yet accelerate healing of acid-burn skin wounds: a multimodal imaging study. Sci Rep. 2017;7(1):4644. PubMed PMC

Bulte JWM. In vivo MRI cell tracking: clinical studies. Am Roentgenol. 2009;193(2):314–325. PubMed PMC

Walczak P, Wojtkiewicz J, Nowakowski A, Habich A, Holak P, Xu J, Adamiak Z, Chehade M, Pearl MS, Gailloud P, Lukomska B. et al. Real-time MRI for precise and predictable intra-arterial stem cell delivery to the central nervous system. J Cereb Blood Flow Metab. 2017;37(7):2346–2358. PubMed PMC

Bulte JWM, Krishnan KM. Quantitative “hot spot” imaging of transplanted stem cells using superparamagnetic tracers and magnetic particle imaging (MPI). Tomography. 2015;1(2):91–97. PubMed PMC

Them K, Salamon J, Szwargulski P, Sequeira S, Kaul MG, Lange C, Ittrich H, Knopp T. Increasing the sensitivity for stem cell monitoring in system-function based magnetic particle imaging. Phys Med Biol. 2016;61(9):3279–3290. PubMed

Zheng Y, Huang J, Zhu T, Li R, Wang Z, Ma F, Zhu J. Stem cell tracking technologies for neurological regenerative medicine purposes. Stem Cells Int. 2017;2017:2934149. PubMed PMC

Wahajuddin Arora S. Superparamagnetic iron oxide nanoparticles: magnetic nanoplatforms as drug carriers. Int J Nanomedicine. 2012;7:3445–3471. PubMed PMC

Wilkinson KE, Palmberg L, Witasp E, Kupczyk M, Feliu N, Gerde P, Seisenbaeva GA, Fadeel B, Dahlén SE, Kessler VG. Solution-engineered palladium nanoparticles: model for health effect studies of automotive particulate pollution. ACS Nano. 2011;5(7):5312–5324. PubMed

Ekstrand-Hammarstrom B, Akfur CM, Andersson PO, Lejon C, Osterlund L, Bucht A. Human primary bronchial epithelial cells respond differently to titanium dioxide nanoparticles than the lung epithelial cell lines A549 and BEAS-2B. Nanotoxicology. 2012;6(6):623–634. PubMed

Kermanizadeh A, Gaiser BK, Ward MB, Stone V. Primary human hepatocytes versus hepatic cell line: assessing their suitability for in vitro nanotoxicology. Nanotoxicology. 2013;7(7):1255–1271. PubMed

Joris F, Valdepérez D, Pelaz B, Wang T, Doak SH, Manshian BB, Soenen SJ, Parak WJ, De Smedt SC, Raemdonck K. Choose your cell model wisely: the in vitro nanoneurotoxicity of differentially coated iron oxide nanoparticles for neural cell labeling. Acta Biomater. 2017;55:204–213. PubMed

Iwasaki A, Medzhitov R. Control of adaptive immunity by the innate immune system. Nat Immunol. 2015;16(4):343–353. PubMed PMC

Turner MW. The role of mannose-binding lectin in health and disease. Mol Immunol. 2003;40(7):423–429. PubMed

Muller S, Schaffer T, Flogerzi B, Seibold-Schmid B, Schnider J, Takahashi K, Darfeuille-Michaud A, Vazeille E, Schoepfer AM, Seibold F. Mannan-binding lectin deficiency results in unusual antibody production and excessive experimental colitis in response to mannose-expressing mild gut pathogens. Gut. 2010;59(11):1493–1500. PubMed

Sharma AA, Jen R, Butler A, Lavoie PM. The developing human preterm neonatal immune system: a case for more research in this area. Clin Immunol. 2012;145(1):61–68. PubMed PMC

Hu X, Shi Y, Zhang P, Miao M, Zhang T, Jiang B. d-Mannose: properties, production, and applications: an overview. Compr Rev Food Sci Food Saf. 2016;15(4):773–785. PubMed

Stutman O, Dien P, Wisun RE, Lattime EC. Natural cytotoxic cells against solid tumors in mice: blocking of cytotoxicity by D-mannose. Proc Natl Acad Sci U S A. 1980;77(5):2895–2898. PubMed PMC

Ranta K, Nieminen K, Ekholm FS, Polakova M, Roslund MU, Saloranta T, Leino R, Savolainen J. Evaluation of immunostimulatory activities of synthetic mannose-containing structures mimicking the -(1->2)-linked cell wall mannans of Candida albicans. Clin Vaccine Immunol. 2012;19(11):1889–1893. PubMed PMC

Kamel MM, Ali HI, Anwar MM, Mohamed NA, Soliman AM. Synthesis, antitumor activity and molecular docking study of novel sulfonamide-schiff’s bases, thiazolidinones, benzothiazinones and their C-nucleoside derivatives. Eur J Med Chem. 2010;45(2):572–580. PubMed

Singh N, Hopkins SJ, Hulme S, Galea JP, Hoadley M, Vail A, Hutchinson PJ, Grainger S, Rothwell NJ, King AT, Tyrrell PJ. The effect of intravenous interleukin-1 receptor antagonist on inflammatory mediators in cerebrospinal fluid after subarachnoid haemorrhage: a phase II randomised controlled trial. J Neuroinflammation. 2014;11(1):1. PubMed PMC

Chen F-E, Zhao J-F, Xiong F-J, Xie B, Zhang P. An improved synthesis of a key intermediate for (+)-biotin from d-mannose. Carbohydr Res. 2007;342(16):2461–2464. PubMed

Horák D, Babič M, Jendelová P, Herynek V, Trchová M, Pientka Z, Pollert E, Hájek M, Syková E. D-mannose-modified iron oxide nanoparticles for stem cell labeling. Bioconjug Chem. 2007;18(3):635–644. PubMed

Borisova T, Krisanova N, Borysov A, Sivko R, Ostapchenko L, Babic M, Horak D. Manipulation of isolated brain nerve terminals by an external magnetic field using D-mannose-coated γ-Fe2O3 nano-sized particles and assessment of their effects on glutamate transport. Beilstein J Nanotechnol. 2014;5(1):778–788. PubMed PMC

Horák D, Babič M, Jendelová P, Herynek V, Trchová M, Likavčanová K, Kapcalová M, Hájek M, Syková E. Effect of different magnetic nanoparticle coatings on the efficiency of stem cell labeling. J Magn Magn Mater. 2009;321(10):1539–1547.

Vinković Vrček I, Pavičić I, Crnković T, Jurašin D, Babič M, Horák D, Lovrić M, Ferhatović L, Ćurlin M, Gajović S. Does surface coating of metallic nanoparticles modulate their interference with in vitro assays? RSC Adv. 2015;5(87):70787–70807.

Pongrac IM, Pavičić I, Milić M, Brkić Ahmed L, Babič M, Horák D, Vinković Vrček I, Gajović S. Oxidative stress response in neural stem cells exposed to different superparamagnetic iron oxide nanoparticles. Int J Nanomedicine. 2016;11:1701–1715. PubMed PMC

Azari H, Sharififar S, Rahman M, Ansari S, Reynolds BA. Establishing embryonic mouse neural stem cell culture using the neurosphere assay. J Vis Exp. 2011;11(47):1–4. PubMed PMC

Kosi N, Alić I, Kolačević M, Vrsaljko N, Jovanov Milošević N, Sobol M, Philimonenko A, Hozák P, Gajović S, Pochet R, Mitrečić D. Nop2 is expressed during proliferation of neural stem cells and in adult mouse and human brain. Brain Res. 2015;1597:65–76. PubMed

Pongrac IM, Dobrivojević M, Ahmed LB, Babič M, Šlouf M, Horák D, Gajović S. Improved biocompatibility and efficient labeling of neural stem cells with poly(L-lysine)-coated maghemite nanoparticles. Beilstein J Nanotechnol. 2016;7(1):926–936. PubMed PMC

Yang CY, Tai MF, Lin CP, Lu CW, Wang JL, Hsiao JK, Liu HM. Mechanism of cellular uptake and impact of ferucarbotran on macrophage physiology. PLoS One. 2011;6(9):1–7. PubMed PMC

Zucker RM, Massaro EJ, Sanders KM, Degn LL, Boyes WK. Detection of TiO2 nanoparticles in cells by flow cytometry. Cytometry A. 2010;77(7):677–685. PubMed

Overton WR. Modified histogram subtraction technique for analysis of flow cytometry data. Cytometry. 1988;9:619–626. PubMed

Alić I, Kosi N, Kapuralin K, Gorup D, Gajović S, Pochet R, Mitrečić D. Neural stem cells from mouse strain Thy1 YFP-16 are a valuable tool to monitor and evaluate neuronal differentiation and morphology. Neurosci Lett. 2016;634:32–41. PubMed

Hillegass JM, Shukla A, Lathrop SA, MacPherson MB, Fukagawa NK, Mossman BT. Assessing nanotoxicity in cells in vitro. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2010;2(3):219–231. PubMed PMC

Brandenberger C, Mühlfeld C, Ali Z, Lenz AG, Schmid O, Parak WJ, Gehr P, Rothen-Rutishauser B. Quantitative evaluation of cellular uptake and trafficking of plain and polyethylene glycol-coated gold nanoparticles. Small. 2010;6(15):1669–1678. PubMed

Pritz CO, Bitsche M, Salvenmoser W, Dudás J, Schrott-Fischer A, Glueckert R. Endocytic trafficking of silica nanoparticles in a cell line derived from the organ of Corti. Nanomedicine (Lond). 2013;8(2):239–252. PubMed

Bulte JWM, Kraitchman DL. Iron oxide MR contrast agents for molecular and cellular imaging. NMR Biomed. 2004;17(7):484–499. PubMed

Hsiao JK, Chu HH, Wang YH, Lai CW, Chou PT, Hsieh ST, Wang JL, Liu HM. Macrophage physiological function after superparamagnetic iron oxide labeling. NMR Biomed. 2008;21(8):820–829. PubMed

Arbab AS, Bashaw LA, Miller BR, Jordan EK, Lewis BK, Kalish H, Frank JA. Characterization of biophysical and metabolic properties of cells labeled with superparamagnetic iron oxide nanoparticles and transfection agent for cellular MR imaging. Radiology. 2003;229(3):838–846. PubMed

Arbab AS, Wilson LB, Ashari P, Jordan EK, Lewis BK, Frank JA. A model of lysosomal metabolism of dextran coated superparamagnetic iron oxide (SPIO) nanoparticles: implications for cellular magnetic resonance imaging. NMR Biomed. 2005;18(6):383–389. PubMed

Küstermann E, Himmelreich U, Kandal K, Geelen T, Ketkar A, Wiedermann D, Strecker C, Esser J, Arnhold S, Hoehn M. Efficient stem cell labeling for MRI studies. Contrast Media Mol Imaging. 2008;3(1):27–37. PubMed

Novotna B, Jendelova P, Kapcalova M, Rossner P, Turnovcova K, Bagryantseva Y, Babic M, Horak D, Sykova E. Oxidative damage to biological macromolecules in human bone marrow mesenchymal stromal cells labeled with various types of iron oxide nanoparticles. Toxicol Lett. 2012;210(1):53–63. PubMed

Umashankar A, Corenblum MJ, Ray S, Valdez M, Yoshimaru ES, Trouard TP, Madhavan L. Effects of the iron oxide nanoparticle Molday ION Rhodamine B on the viability and regenerative function of neural stem cells: relevance to clinical translation. Int J Nanomedicine. 2016;11:1731–1748. PubMed PMC

Lu CW, Hsiao JK, Liu HM, Wu CH. Characterization of an iron oxide nanoparticle labelling and MRI-based protocol for inducing human mesenchymal stem cells into neural-like cells. Sci Rep. 2017;7(1):3587. PubMed PMC

Addicott B, Willman M, Rodriguez J, Padgett K, Han D, Berman D, Hare JM, Kenyon NS. Mesenchymal stem cell labeling and in vitro MR characterization at 1.5 T of new SPIO contrast agent: Molday ION Rhodamine-BTM . Contrast Media Mol Imaging. 2011;6(1):7–18. PubMed PMC

Linehan SA, Martínez-Pomares L, Stahl PD, Gordon S. Mannose receptor and its putative ligands in normal murine lymphoid and nonlymphoid organs: in situ expression of mannose receptor by selected macrophages, endothelial cells, perivascular microglia, and mesangial cells, but not dendritic cells. J Exp Med. 1999;189(12):1961–1972. PubMed PMC

Régnier-Vigouroux A. The mannose receptor in the brain. Int Rev Cytol. 2003;226:321–342. PubMed

Macedo-Ramos H, Campos FSO, Carvalho LA, Ramos IB, Teixeira LM, De Souza W, Cavalcante LA, Baetas-da-Cruz W. Olfactory ensheathing cells as putative host cells for Streptococcus pneumoniae: evidence of bacterial invasion via mannose receptor-mediated endocytosis. Neurosci. Res. 2011;69(4):308–313. PubMed

Giraldi-Guimarães A, De Freitas HT, Coelho BDP, MacEdo-Ramos H, Mendez-Otero R, Cavalcante LA, Baetas-Da-Cruz W. Bone marrow mononuclear cells and mannose receptor expression in focal cortical ischemia. Brain Res. 2012;1452:173–184. PubMed

Macedo-Ramos H, Batista AF, Carrier-Ruiz A, Alves L, Allodi S, Ribeiro-Resende VT, Teixeira LM, Baetas-Da-Cruz W. Evidence of involvement of the mannose receptor in the internalization of Streptococcus pneumoniae by Schwann cells. BMC Microbiol. 2014;14:211. PubMed PMC

Apostolopoulos V, Pietersz GA, Gordon S, Martinez-Pomares L, McKenzie LFC. Aldehyde-mannan antigen complexes target the MHC class I antigen-presentation pathway. Eur J Immunol. 2000;30(6):1714–1723. PubMed

Nel A. Toxic potential of materials at the nanolevel. Science. 2006;311(5761):622–627. PubMed

Yang H, Liu C, Yang D, Zhang H, Xi Z. Comparative study of cytotoxicity, oxidative stress and genotoxicity induced by four typical nanomaterials: the role of particle size, shape and composition. J Appl Toxicol. 2009;29(1):69–78. PubMed

Sheridan C. Proof of concept for next-generation nanoparticle drugs in humans. Nat Biotechnol. 2012;30(6):471–473. PubMed

Jurašin DD, Ćurlin M, Capjak I, Crnković T, Lovrić M, Babič M, Horák D, Vrček IV, Gajović S. Surface coating affects behavior of metallic nanoparticles in a biological environment. Beilstein J Nanotechnol. 2016;7(1):246–262. PubMed PMC

Kallur T, Farr TD, Böhm-Sturm P, Kokaia Z, Hoehn M. Spatio-temporal dynamics, differentiation and viability of human neural stem cells after implantation into neonatal rat brain. Eur J Neurosci. 2011;34(3):382–393. PubMed

Tennstaedt A, Mastropietro A, Nelles M, Beyrau A, Hoehn M. In vivo fate imaging of intracerebral stem cell grafts in mouse brain. PLoS One. 2015;10(12):e0144262. PubMed PMC

Tennstaedt A, Aswendt M, Adamczak J, Collienne U, Selt M, Schneider G, Henn N, Schaefer C, Lagouge M, Wiedermann D, Kloppenburg P. et al. Human neural stem cell intracerebral grafts show spontaneous early neuronal differentiation after several weeks. Biomaterials. 2015;44:143–154. PubMed

Boehm-Sturm P, Aswendt M, Minassian A, Michalk S, Mengler L, Adamczak J, Mezzanotte L, Löwik C, Hoehn M. A multi-modality platform to image stem cell graft survival in the naïve and stroke-damaged mouse brain. Biomaterials. 2014;35(7):2218–2226. PubMed

Janowski M, Engels C, Gorelik M, Lyczek A, Bernard S, Bulte JWM, Walczak P. Survival of neural progenitors allografted into the CNS of immunocompetent recipients is highly dependent on transplantation site. Cell Transplant. 2014;23(2):253–262. PubMed PMC

Focke A, Schwarz S, Foerschler A, Scheibe J, Milosevic J, Zimmer C, Schwarz J. Labeling of human neural precursor cells using ferromagnetic nanoparticles. Magn Reson Med. 2008;60(6):1321–1328. PubMed

Joris F, Valdepérez D, Pelaz B, Soenen SJ, Manshian BB, Parak WJ, De Smedt SC, Raemdonck K. The impact of species and cell type on the nanosafety profile of iron oxide nanoparticles in neural cells. J Nanobiotechnology. 2016;14(1):69. PubMed PMC

Najít záznam

Citační ukazatele

Nahrávání dat ...

Možnosti archivace

Nahrávání dat ...