Skip to main content

Application of magnetic nanoparticles in cell therapy

Abstract

Fe3O4 magnetic nanoparticles (MNPs) are biomedical materials that have been approved by the FDA. To date, MNPs have been developed rapidly in nanomedicine and are of great significance. Stem cells and secretory vesicles can be used for tissue regeneration and repair. In cell therapy, MNPs which interact with external magnetic field are introduced to achieve the purpose of cell directional enrichment, while MRI to monitor cell distribution and drug delivery. This paper reviews the size optimization, response in external magnetic field and biomedical application of MNPs in cell therapy and provides a comprehensive view.

Introduction

Stem cells have the function of tissue regeneration and repair. In addition, EVs and exosomes from cells also have the function of repairing injury. In particular, they have the attributes of low immunogenicity and drug delivery potential, and they carry a variety of signaling biomolecules (proteins, mRNAs and miRNAs). At present, stem cells and their secretory vesicles have been widely used in disease treatment. Researchers intravenously inject repair cells or vesicles into organisms to treat diseases, hoping that they can accumulate at the injured site via the blood circulation [1]. The key is the successful delivery of drugs to the lesion. Although stem cells have homing ability, they still have shortcomings. The actual enrichment effect is not ideal [2, 3]. Kyrtatos et al. [4] showed data in vitro by computer simulation that a few cells were retained in the target tissue area in the presence of blood flow. By intracoronary injection, the cell residence time was only a few minutes in the study of myocardial regeneration [5]. At the same time, EVs were cleared in a short time (1.2–1.3 min) after intravenous injection into the blood, and most of them accumulated in the liver and spleen. Therefore, the poor targeting, the low retention rate and the poor therapeutic effect limit the clinical application in cell therapy [6].

SPIONs have obvious advantages of low toxicity to organisms, magnetic targeting, magnetic resonance imaging tracking, hyperthermia and drug delivery ability. More importantly, SPIONs are an iron-containing preparation approved by the FDA. Therefore, the development of SPIONs has attracted increasing attention, especially in medicine. It is important to target and transport the therapeutic cells and EVs under the joint action of MNPs and an external magnetic field. This method can not only improve precise positioning within organisms [4, 7,8,9], improve the retention rate and prolong the drug half-life but also reduce the drug dosage and enhance the efficacy, even in high-flow systems (such as the arterial vascular system). In the literature [4, 10], it was found that targeted stem cells were enriched approximately five times more than nontargeted cells in arteries. After labeling with MNPs, EVs were preserved in the blood for 7–8 h in a magnetic field. Magnetic targeting was found to enhance myocardial retention of intravascular EPCs. They could also effectively cross the BBB and deliver drugs [11, 12] under a magnetic field. Therefore, cells and EVs labeled by MNPs achieved multiple functions of treatment, targeting, drug delivery, magnetic hyperthermia and MRI, which has a great development potential in medicine.

Here, we introduce the combination of MNPs not only with cells, but also with extracellular vesicles, exosomes and artificial simulated liposomes (known as membrane system or membrane structure for convenience). In this paper, we review the requirements for the size of MNPs when used in combination with membrane systems, the response under external magnetic field and its application in medicine.

Application size of MNPs

In this paper, we have reviewed the literature from the last 20 years and further explained the importance of the appropriate size of MNPs in Table 1.

Table 1 Literature summary of medical applications of MNPs in recent 20 years

A total of 36 literature studies are counted in Table 1. There are 14 articles on loading MNPs into cells, 2 articles on loading MNPs into vesicles, 11 articles on exosomes and 6 articles on liposomes. Table 1 shows that most researchers prefer to use MNPs of approximately 10 nm in the field of nanomedicine, because there are 25 literature studies introducing MNPs of 5–15 nm, accounting for 69%. This is consistent with the results in the literature [47, 48]. MNPs (< 200 nm) are rapidly filtered by the spleen and liver and cleared by the kidney at 2 nm, while 10-nm particles easily enter the circulatory system. Moreover, in the process of preparing MNPs, ultra-small magnetic particles (< 5 nm) have very good dispersion and stability in the organic coating. However, the response to external magnetic field is poor. Therefore, it is less used for magnetic targeting in cell therapy and more used as MRI contrast agent. When the size increases to 10 nm, it not only enhances the dispersion, but also enhances the response to the external magnetic field. In addition, with the continuous expansion of the scale, its agglomeration phenomenon is becoming more and more serious. Combined with the results summarized in the literature [6], the hydrodynamic size advantages [47] and optimization [49] of MNPs, we believe that a core diameter of approximately 10 nm has great advantages. At the same time, when preparing MNPs-labeled exosomes, the size of exosomes should be considered to facilitate the loading of MNPs. In addition, there are two main types of MNPs, namely Fe3O4 and γ-Fe2O3, but γ-Fe2O3 is better and safer for cells because ferric iron causes less damage to the recipient cells.

Effect of the magnetic field on the cells labeled by MNPs

The process of labeling cells with MNPs includes simple culture, transfection agents, magnetoelectric perforation and magnetoacoustic perforation [50]. The small volume and negative surface charge of MNPs are conducive to their nonspecific adsorption on the plasma membrane, thus triggering their endocytosis pathway in cells [51]. MNPs were ingested by cells and encapsulated in endosomes inside the cells. The endosomes were located close to the cytoplasm and away from the nucleus and not affected the normal activity of the cell. Analysis of the intracellular MNP distribution in the literature [17] showed that the MNP vesicles captured by cells diffuse along the cytoplasm and respond to magnetic fields [52], as shown in Fig. 1. A review [13] indicated that endosomes were elongated and aligned in the direction of the magnetic force line after the cells had phagocytized anionic colloidal ferromagnetic nanoparticles into the endosomes. In addition, the cells movement depended on the gradient in the magnetic field instead of feeling the strength [53,54,55]. The cells migrated toward the region with the maximum gradient. The study on the survival rate and proliferation rate of BMSCs under a high-intensity static magnetic field suggested that it had the least effect on cell proliferation within 24 h [56]. The effect depended on the degree of iron load [4]. If applied for a short time, cells can withstand a higher magnetic force. Moreover, stem cells were induced to differentiate by an extremely low-frequency magnetic field to obtain an ideal phenotype [57,58,59]. A study [51] revealed that the rotational movement of SPIONs induced by a dynamic magnetic field led to changes in cell membrane permeability and even apoptosis, because applying magnetic force to SPIONs distorted the inner membrane of the cell [13]. At the same time, the vesicles encapsulating MNPs not existed in cells for a long time and were soon excreted by cells to form free labeled extracellular vesicles.

Fig. 1
figure 1

Changes of MNPs phagocytized by cells in magnetic field

In summary, the key is to pay attention to the load of MNPs, the strength and time in the magnetic field and the preparation time of cells' sample. Because of the long time, the vesicles encapsulating MNPs were produced in the test, as shown in Fig. 2.

Fig. 2
figure 2

Cellular changes after phagocytosis of MNPs

Application of MNP-labeled membrane systems in biomedicine

Application of MNP-labeled cells in biomedicine

Stem cells and secreted vesicles have multidirectional differentiation potential, migrate to inflammatory sites and repair tissues and regenerate [60]. Therefore, they are widely used in cell therapy and have great prospects in the field of regenerative medicine. Stem cells have homing ability. However, they still have limitations and deficiencies. When stem cells are injected intravenously into the body, they are easily dispersed by blood flow and cleared by organisms, resulting in difficult localization, a short half-life and a low retention rate. To enhance targeting ability, prolong the retention rate and enhance the therapeutic effect of stem cells, MNPs are introduced. In addition, it was also important to minimize the injection dose, the toxicity and the side effects on healthy tissues and to cause the labeled cells or EVs to accumulate in the target tissue to reduce the in vitro administration concentration and frequency. As shown in Table 1, stem cells were labeled with MNPs and then injected into organisms under the magnetic field to treat lung injury [39], atrial fibrillation [61], coronary artery embolism [62], vascular diseases [63], systemic osteoporosis [36], retinitis [64], high intraocular pressure [30], Parkinson’s disease [40, 65], myocardial infarction [42] and spinal cord injury [43] and to regenerate articular cartilage and bone [66,67,68,69,70,71].

As shown in Fig. 2, MNP is engulfed by stem cells to form endosomes, which are then transformed into MVEs. The MVEs have two fates. One is to combine with lysosomes (low pH) in cells, be further digested and decomposed into Fe3+. The free metal ions released into the cytoplasm promote pathways of JNK activation and c-Jun phosphorylation, cause the upregulation of a variety of cytokines in cells, and induce angiogenesis, anti-apoptosis and anti-inflammatory [44]. Moreover, in the treatment of myocardial infarction, it was found to induce the increased expression of connexin 43 (Cx43) in H9c2 [42]. However, the other is excreted outside the cell in the form of extracellular vesicles. At the same time, exosomes prepared with MSC-IONP also contained a large number of therapeutic growth factors [44].

Application of MNP-labeled EVs in biomedicine

In the past decade, there have been an increasing number of studies on EVs, especially exosomes (50–150 nm). Due to the biological characteristics such as low immunogenicity and drug delivery, they have better application prospects in the field of tissue regeneration [6]. EVs are effectors of intercellular communication and act as natural endogenous carriers. To improve their low separation rate and insufficient targeting ability, MNPs and magnetic localization were used to enhance the directional distribution ability of EVs. To internalize SPIONs directly into EVs, electroporation, natural incubation and other methods were used. However, the membrane is incomplete due to electroporation. MNP-labeled cells' secretory vesicles are the primary choice for natural processes. Under the external magnetic field, MNP-labeled EVs were concentrated and enriched at the target position for tissue repair and regeneration. Many studies have used this method to improve and treat scar formation in acute and chronic porcine myocardial infarction [72], ischemic stroke [44], skin trauma [27], spinal cord injury [43, 73], infarcted heart [22], wound healing [23], bone and angiogenesis [74], and myocardial infarction [45].

Application of magnetic liposomes in biomedicine

Liposomes are similar to EVs released by cells. Synthetic liposomes are usually spherical closed structures composed of lipid bilayers enclosing the internal hydrophilic chamber [75]. The size of liposomes ranges from 20 nm to several microns [76]. Like vesicles released by cells, liposomes can encapsulate molecules with different solubilities. Liposomes have the advantage of encapsulating MNPs without being affected by enzyme degradation, but there are no proteins or other biomolecules from precursor cells [16]. At the same time, loading hydrophilic USPIO into the hydrophilic chamber of liposome can also achieve the magnetic targeting [77], drug delivery and MRI imaging [21, 78, 79] and generate multifunctional liposomes [77, 80]. Moreover, the preparation of liposomes is simple and controllable, so researchers have given increasing attention to the use of liposomes. Under a permanent magnetic field, magnetic liposomes deform into slender ellipsoids [81]. A study [82] first reported the fusion of dry magnetic liposomes and cell membrane models, evaluated their interactions (giant monolayer vesicles, GUVs) and considered the future application in drug delivery of such magnetic systems. Ideally, the target release system allows external control of the time and dose of products released at the target location [83].

In summary, the application of the MNP-labeled membrane systems has treated many diseases successfully. In addition, the method can also be extended to the cell localization of other organs and provide a useful tool for systematic cell therapy. At the same time, it can also treat acute organ failure with high mortality. For example, acute lung injury and acute kidney injury are caused by COVID-19 or other diseases. However, there are few studies on these diseases. Combined with the rapid targeting enrichment and the therapeutic advantages of stem cells and exosomes, it will be possible to treat acute organ failure.

Tumor treatment with magnetic hyperthermia

In addition to tissue repair and regeneration, the combination of MNPs and membrane systems is also used to treat tumors. By changing the frequency of the external magnetic field, the high-speed movement of MNPs can be controlled. The magnetic fields mentioned here include the high-frequency and the low-frequency magnetic field. The high-frequency AMF makes MNPs in organism rotate at high speed, increase membrane permeability [11, 16, 21], enhance drug release, and even tear cell membrane [83]. It can also convert magnetic energy into heat to kill tumor cells. Protect the surrounding environment while the local temperature far exceeds the body temperature [12]. The damaging effect of magnetic hyperthermia on microglial BV2 cells [17] and increasing drug release [84] were studied. However, under a low-frequency magnetic field, the mechanism of destroying the membrane was mechanical and not dependent on heat. Therefore, nonspecific heating of surrounding tissues caused by AMF is avoided [83].

According to the literature for magnetic hyperthermia, in this paper, we mainly reviewed the combination of MNPs and membrane systems for magnetic hyperthermia to treat tumors. Therefore, the use of only heated MNP to kill tumors is not described here. Cell communication promotes tumor development through vesicles. It is believed that the vesicles are used as Trojan horses to provide a therapeutic payload for cancer cells [76]. Therefore, exosomes in blood were combined with MNPs to target cancer [26]. Recently, it was reported that SPION-modified exosomes transferred TNF-α to cancer cells through magnetic targeting and significantly inhibited tumor growth [28].

With regard to the study of magnetic liposomes, the literature studies [81, 85, 86] have reported the possibility of using magnetic liposomes and in vitro magnets to treat solid tumors. Some reports [84, 87] prepared multifunctional magnetoliposomes loaded with the anticancer drug DOX to inhibit cancer. In high-frequency magnetic field, multifunctional liposomes cause perforation effect or change the permeability of vesicle membrane through local heating to increase drug release [88]. This method opens up a new prospect for the development of intelligent drug delivery system.

MRI application of MNP-labeled membrane systems

MNP-labeled membrane systems were used for three-dimensional noninvasive imaging positioning to achieve real-time monitoring of biological distribution in vivo [47, 89,90,91,92], as well as MRI of embryonic tissue [93, 94], adipose [95, 96], human adipose-derived stem cells [97, 98] and extracellular vesicles [99,100,101]. The dual exogenous substances in which cells are incubated with photosensitizers and MNPs [76] provide magnetic and optical responsiveness to vesicles for treatment and monitoring distribution. In addition to the situation described above, MNPs were encapsulated in liposomes together with fluorescent nanoparticles [102], rhodamine labeling [103] and quantum dots [104] for tracking. In conclusion, the application of MNP-labeled membrane systems can simultaneously have the advantages of treatment and visualization, so as to timely monitor the embolism caused by foreign substances and evaluate the metabolism.

Conclusions and expectations

This paper reviews the research progress of the application of MNP-labeled membrane systems in biomedicine. They have a wide range of applications in medicine and open up a potential application prospect for the directional positioning of biological entities. Through the perfect combination and utilization of magnetic targeting, MRI and magnetic hyperthermia, many diseases are expected to be successfully treated. In particular, through the continuous progress of magnetic field design, noninvasive treatment of deep diseases can be realized, not just on the surface of the body. The development of MNPs in precision medicine needs more exploration. In the future, it is expected to achieve more mature, comprehensive (systematic) and accurate (positioning) disease treatment in medicine.

Availability of data and materials

The data used to support the findings of this study are available from the corresponding author upon request.

Abbreviations

SPIONs:

Superparamagnetic iron oxide nanoparticles

MNPs:

Magnetic nanoparticles

USPIO:

Ultra-small superparamagnetic iron oxide

MT:

Magnetic targeting

ADSCs:

Human primary adipose-derived stem cells

hMSCs:

Human mesenchymal stem cells

MSCs:

Mesenchymal stromal cells

NSCs:

Neural stem cells

ESCs:

Embryonic stem cells

EVs:

Extracellular vesicles

BBB:

Blood–brain barrier

EPCs:

Endothelial progenitor cells

DOX:

Doxorubicin

MRI:

Magnetic resonance imaging

TNF-α:

Tumor necrosis factor

IONP:

Iron oxide nanoparticle

H9C2:

Cardiomyoblasts

Cx43:

Connexin 43

MVEs:

Mature multivesicular endosomes  Alternating magnetic field(AMF)

AMF:

Alternating magnetic field

References

  1. Them K, Salamon J, Szwargulski P, Sequeira S, Kaul MG, Lange C, et al. Increasing the sensitivity for stem cell monitoring in system-function based magnetic particle imaging. Phys Med Biol. 2016;61:3279–90. https://doi.org/10.1088/0031-9155/61/9/3279.

    Article  CAS  PubMed  Google Scholar 

  2. Hristov M, Weber C. The therapeutic potential of progenitor cells in ischemic heart disease. Basic Res Cardiol. 2006;101:1–7. https://doi.org/10.1007/s00395-005-0573-0.

    Article  PubMed  Google Scholar 

  3. Charwat S, Gyöngyösi M, Lang I, Graf S, Beran G, Hemetsberger R, et al. Role of adult bone marrow stem cells in the repair of ischemic myocardium: current state of the art. Exp Hematol. 2008;36:672–80.

    Article  CAS  PubMed  Google Scholar 

  4. Kyrtatos PG, Lehtolainen P, Junemann-Ramirez M, Garcia-Prieto A, Price AN, Martin JF, et al. Magnetic tagging increases delivery of circulating progenitors in vascular injury. JACC Cardiovasc Interv. 2009;2:794–802. https://doi.org/10.1016/j.jcin.2009.05.014.

    Article  PubMed  Google Scholar 

  5. Roncalli JG, Tongers J, Renault M-A, Losordo DW. Endothelial progenitor cells in regenerative medicine and cancer: a decade of research. Trends Biotechnol. 2008;26:276–83. https://doi.org/10.1016/j.tibtech.2008.01.005.

    Article  CAS  PubMed  Google Scholar 

  6. Zhuo Z, Wang J, Luo Y, Zeng R, Zhang C, Zhou W, et al. Targeted extracellular vesicle delivery systems employing superparamagnetic iron oxide nanoparticles. Acta Biomater. 2021. https://doi.org/10.1016/j.actbio.2021.07.027.

    Article  PubMed  Google Scholar 

  7. Consigny PM, Silverberg DA, Vitali NJ. Use of endothelial cells containing superparamagnetic microspheres to improve endothelial cell delivery to arterial surfaces after angioplasty. J Vasc Interv Radiol JVIR. 1999;10:155.

    Article  CAS  PubMed  Google Scholar 

  8. Pislaru SV, Harbuzariu A, Gulati R, Witt T, Sandhu NP, Simari RD, et al. Magnetically targeted endothelial cell localization in stented vessels. J Am Coll Cardiol. 2006;48:1839–45.

    Article  CAS  PubMed  Google Scholar 

  9. Pislaru SV. Magnetic forces enable rapid endothelialization of synthetic vascular grafts. Circulation. 2006;114:314–8.

    Article  Google Scholar 

  10. Zhang B, Mo X, Yu F, Ma Y, Yan F. Ultrasound monitoring of magnet-guided delivery of mesenchymal stem cells labeled with magnetic lipid-polymer hybrid nanobubbles. Biomater Sci. 2020;8:3628–39. https://doi.org/10.1039/d0bm00473a.

    Article  CAS  PubMed  Google Scholar 

  11. Guardia P, Di Corato R, Lartigue L, Wilhelm C, Espinosa A, Garcia-Hernandez M, et al. Water-soluble iron oxide nanocubes with high values of specific absorption rate for cancer cell hyperthermia treatment. ACS Nano. 2012;6:3080–91. https://doi.org/10.1021/nn2048137.

    Article  CAS  PubMed  Google Scholar 

  12. Amstad E, Kohlbrecher J, Müller E, Schweizer T, Textor M, Reimhult E. Triggered release from liposomes through magnetic actuation of iron oxide nanoparticle containing membranes. Nano Lett. 2011;11:1664–70. https://doi.org/10.1021/nl2001499.

    Article  CAS  PubMed  Google Scholar 

  13. Wilhelm C, Cebers A, Bacri JC, Gazeau F. Deformation of intracellular endosomes under a magnetic field. Eur Biophys J. 2003;32:655–60.

    Article  CAS  PubMed  Google Scholar 

  14. Béalle G, Di Corato R, Kolosnjaj-Tabi J, Dupuis V, Clément O, Gazeau F, et al. Ultra magnetic liposomes for MR imaging, targeting, and hyperthermia. Langmuir. 2012;28:11834–42. https://doi.org/10.1021/la3024716.

    Article  CAS  PubMed  Google Scholar 

  15. Mart RJ, Liem KP, Webb SJ. Creating functional vesicle assemblies from vesicles and nanoparticles. Pharm Res. 2009;26:1701–10. https://doi.org/10.1007/s11095-009-9880-8.

    Article  CAS  PubMed  Google Scholar 

  16. Dwivedi P, Kiran S, Han S, Dwivedi M, Khatik R, Fan R, et al. Magnetic targeting and ultrasound activation of liposome-microbubble conjugate for enhanced delivery of anticancer therapies. ACS Appl Mater Interfaces. 2020;12:23737–51. https://doi.org/10.1021/acsami.0c05308.

    Article  CAS  PubMed  Google Scholar 

  17. Calatayud MP, Soler E, Torres TE, Campos-Gonzalez E, Junquera C, Ibarra MR, et al. Cell damage produced by magnetic fluid hyperthermia on microglial BV2 cells. Sci Rep. 2017;7:8627. https://doi.org/10.1038/s41598-017-09059-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mart RJ, Liem KP, Webb SJ. Magnetically-controlled release from hydrogel-supported vesicle assemblies. Chem Commun (Camb). 2009. https://doi.org/10.1039/b901472a.

    Article  Google Scholar 

  19. Martina M-S, Wilhelm C, Lesieur S. The effect of magnetic targeting on the uptake of magnetic-fluid-loaded liposomes by human prostatic adenocarcinoma cells. Biomaterials. 2008;29:4137–45. https://doi.org/10.1016/j.biomaterials.2008.07.011.

    Article  CAS  PubMed  Google Scholar 

  20. Wilhelm C, Lavialle F, Péchoux C, Tatischeff I, Gazeau F. Intracellular trafficking of magnetic nanoparticles to design multifunctional biovesicles. Small. 2008;4:577–82. https://doi.org/10.1002/smll.200700523.

    Article  CAS  PubMed  Google Scholar 

  21. Bulte J, Cuyper M, Despres D, Frank JA. Preparation, relaxometry, and biokinetics of PEGylated magnetoliposomes as MR contrast agent. J Magn Magn Mater. 1999;194:204–9.

    Article  CAS  Google Scholar 

  22. Liu S, Chen X, Bao L, Liu T, Yuan P, Yang X, et al. Treatment of infarcted heart tissue via the capture and local delivery of circulating exosomes through antibody-conjugated magnetic nanoparticles. Nat Biomed Eng. 2020;4:1063–75. https://doi.org/10.1038/s41551-020-00637-1.

    Article  CAS  PubMed  Google Scholar 

  23. Wu D, Kang L, Tian J, Wu Y, Liu J, Li Z, et al. Exosomes derived from bone mesenchymal stem cells with the stimulation of FeO nanoparticles and static magnetic field enhance wound healing through upregulated miR-21-5p. Int J Nanomed. 2020;15:7979–93. https://doi.org/10.2147/IJN.S275650.

    Article  CAS  Google Scholar 

  24. Jung KO, Jo H, Yu JH, Gambhir SS, Pratx G. Development and MPI tracking of novel hypoxia-targeted theranostic exosomes. Biomaterials. 2018;177:139–48. https://doi.org/10.1016/j.biomaterials.2018.05.048.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Qi H, Liu C, Long L, Ren Y, Zhang S, Chang X, et al. Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano. 2016;10:3323–33. https://doi.org/10.1021/acsnano.5b06939.

    Article  CAS  PubMed  Google Scholar 

  26. Qi H, Yang L, Li X, Zhan Q, Han D, Zhao J, et al. Exosomes separated based on the “STOP” criteria for tumor-targeted drug delivery. J Mater Chem B. 2018;6:2758–68. https://doi.org/10.1039/c8tb00355f.

    Article  CAS  PubMed  Google Scholar 

  27. Li X, Wang Y, Shi L, Li B, Li J, Wei Z, et al. Magnetic targeting enhances the cutaneous wound healing effects of human mesenchymal stem cell-derived iron oxide exosomes. J Nanobiotechnol. 2020;18:113. https://doi.org/10.1186/s12951-020-00670-x.

    Article  CAS  Google Scholar 

  28. Zhuang M, Chen X, Du D, Shi J, Deng M, Long Q, et al. SPION decorated exosome delivery of TNF-α to cancer cell membranes through magnetism. Nanoscale. 2020;12:173–88. https://doi.org/10.1039/c9nr05865f.

    Article  CAS  PubMed  Google Scholar 

  29. Zhuang M, Du D, Pu L, Song H, Deng M, Long Q, et al. SPION-decorated exosome delivered BAY55-9837 targeting the pancreas through magnetism to improve the blood GLC response. Small. 2019;15: e1903135. https://doi.org/10.1002/smll.201903135.

    Article  CAS  PubMed  Google Scholar 

  30. Snider EJ, Kubelick KP, Tweed K, Kim RK, Li Y, Gao K, et al. Improving stem cell delivery to the trabecular meshwork using magnetic nanoparticles. Sci Rep. 2018;8:12251. https://doi.org/10.1038/s41598-018-30834-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tukmachev D, Lunov O, Zablotskii V, Dejneka A, Babic M, Syková E, et al. An effective strategy of magnetic stem cell delivery for spinal cord injury therapy. Nanoscale. 2015;7:3954–8. https://doi.org/10.1039/c4nr05791k.

    Article  CAS  PubMed  Google Scholar 

  32. Vaněček V, Zablotskii V, Forostyak S, Růžička J, Herynek V, Babič M, et al. Highly efficient magnetic targeting of mesenchymal stem cells in spinal cord injury. Int J Nanomed. 2012;7:3719–30. https://doi.org/10.2147/IJN.S32824.

    Article  CAS  Google Scholar 

  33. Yin PT, Shah S, Pasquale NJ, Garbuzenko OB, Minko T, Lee K-B. Stem cell-based gene therapy activated using magnetic hyperthermia to enhance the treatment of cancer. Biomaterials. 2016;81:46–57. https://doi.org/10.1016/j.biomaterials.2015.11.023.

    Article  CAS  PubMed  Google Scholar 

  34. Andreas K, Georgieva R, Ladwig M, Mueller S, Notter M, Sittinger M, et al. Highly efficient magnetic stem cell labeling with citrate-coated superparamagnetic iron oxide nanoparticles for MRI tracking. Biomaterials. 2012;33:4515–25. https://doi.org/10.1016/j.biomaterials.2012.02.064.

    Article  CAS  PubMed  Google Scholar 

  35. Du V, Luciani N, Richard S, Mary G, Gay C, Mazuel F, et al. A 3D magnetic tissue stretcher for remote mechanical control of embryonic stem cell differentiation. Nat Commun. 2017;8:400. https://doi.org/10.1038/s41467-017-00543-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Labusca L, Herea D-D, Danceanu C-M, Minuti AE, Stavila C, Grigoras M, et al. The effect of magnetic field exposure on differentiation of magnetite nanoparticle-loaded adipose-derived stem cells. Mater Sci Eng C Mater Biol Appl. 2020;109: 110652. https://doi.org/10.1016/j.msec.2020.110652.

    Article  CAS  PubMed  Google Scholar 

  37. Ishii M, Shibata R, Numaguchi Y, Kito T, Suzuki H, Shimizu K, et al. Enhanced angiogenesis by transplantation of mesenchymal stem cell sheet created by a novel magnetic tissue engineering method. Arterioscler Thromb Vasc Biol. 2011;31:2210–5. https://doi.org/10.1161/ATVBAHA.111.231100.

    Article  CAS  PubMed  Google Scholar 

  38. Lu C-W, Hung Y, Hsiao J-K, Yao M, Chung T-H, Lin Y-S, et al. Bifunctional magnetic silica nanoparticles for highly efficient human stem cell labeling. Nano Lett. 2007;7:149–54.

    Article  CAS  PubMed  Google Scholar 

  39. Silva LHA, Silva MC, Vieira JB, Lima ECD, Silva RC, Weiss DJ, et al. Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis. Stem Cells Transl Med. 2020;9:1244–56. https://doi.org/10.1002/sctm.20-0004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Moayeri A, Darvishi M, Amraei M. Homing of super paramagnetic iron oxide nanoparticles (SPIONs) labeled adipose-derived stem cells by magnetic attraction in a rat model of Parkinson’s disease. Int J Nanomed. 2020;15:1297–308. https://doi.org/10.2147/IJN.S238266.

    Article  CAS  Google Scholar 

  41. Yun S, Shin T-H, Lee J-H, Cho MH, Kim I-S, Kim J-W, et al. Design of magnetically labeled cells (Mag-Cells) for in vivo control of stem cell migration and differentiation. Nano Lett. 2018;18:838–45. https://doi.org/10.1021/acs.nanolett.7b04089.

    Article  CAS  PubMed  Google Scholar 

  42. Han J, Kim B, Shin J-Y, Ryu S, Noh M, Woo J, et al. Iron oxide nanoparticle-mediated development of cellular gap junction crosstalk to improve mesenchymal stem cells’ therapeutic efficacy for myocardial infarction. ACS Nano. 2015;9:2805–19. https://doi.org/10.1021/nn506732n.

    Article  CAS  PubMed  Google Scholar 

  43. Kim HY, Kumar H, Jo M-J, Kim J, Yoon J-K, Lee J-R, et al. Therapeutic efficacy-potentiated and diseased organ-targeting nanovesicles derived from mesenchymal stem cells for spinal cord injury treatment. Nano Lett. 2018;18:4965–75. https://doi.org/10.1021/acs.nanolett.8b01816.

    Article  CAS  PubMed  Google Scholar 

  44. Kim HY, Kim TJ, Kang L, Kim Y-J, Kang MK, Kim J, et al. Mesenchymal stem cell-derived magnetic extracellular nanovesicles for targeting and treatment of ischemic stroke. Biomaterials. 2020;243: 119942. https://doi.org/10.1016/j.biomaterials.2020.119942.

    Article  CAS  PubMed  Google Scholar 

  45. Lee J-R, Park B-W, Kim J, Choo YW, Kim HY, Yoon J-K, et al. Nanovesicles derived from iron oxide nanoparticles-incorporated mesenchymal stem cells for cardiac repair. Sci Adv. 2020;6:eaaz0952. https://doi.org/10.1126/sciadv.aaz0952.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wang J, Chen P, Dong Y, Xie H, Wang Y, Soto F, et al. Designer exosomes enabling tumor targeted efficient chemo/gene/photothermal therapy. Biomaterials. 2021;276: 121056. https://doi.org/10.1016/j.biomaterials.2021.121056.

    Article  CAS  PubMed  Google Scholar 

  47. Daldrup-Link HE, Golovko D, Ruffell B, Denardo DG, Castaneda R, Ansari C, et al. MRI of tumor-associated macrophages with clinically applicable iron oxide nanoparticles. Clin Cancer Res. 2011;17:5695–704. https://doi.org/10.1158/1078-0432.CCR-10-3420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen J, Huang N, Ma B, Maitz MF, Wang J, Li J, et al. Guidance of stem cells to a target destination in vivo by magnetic nanoparticles in a magnetic field. ACS Appl Mater Interfaces. 2013;5:5976–85. https://doi.org/10.1021/am400249n.

    Article  CAS  PubMed  Google Scholar 

  49. Trekker J, Leten C, Struys T, Lazenka VV, Argibay B, Micholt L, et al. Sensitive in vivo cell detection using size-optimized superparamagnetic nanoparticles. Biomaterials. 2014;35:1627–35. https://doi.org/10.1016/j.biomaterials.2013.11.006.

    Article  CAS  PubMed  Google Scholar 

  50. Cromer Berman SM, Walczak P, Bulte JWM. Tracking stem cells using magnetic nanoparticles. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2011;3:343–55. https://doi.org/10.1002/wnan.140.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang L, Wang X. Coarse-grained modeling of vesicle responses to active rotational nanoparticles. Nanoscale. 2015;7:13458–67. https://doi.org/10.1039/c5nr01652e.

    Article  CAS  PubMed  Google Scholar 

  52. Zablotskii V, Polyakova T, Lunov O, Dejneka A. How a high-gradient magnetic field could affect cell life. Sci Rep. 2016;6:37407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Miyakoshi J. Effects of static magnetic fields at the cellular level. Prog Biophys Mol Biol. 2005;87:213–23.

    Article  CAS  PubMed  Google Scholar 

  54. Romeo S, Sannino A, Scarfì MR, Massa R, d’Angelo R, Zeni O. Lack of effects on key cellular parameters of MRC-5 human lung fibroblasts exposed to 370 mT static magnetic field. Sci Rep. 2016;6:19398. https://doi.org/10.1038/srep19398.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hore PJ. Are biochemical reactions affected by weak magnetic fields? Proc Natl Acad Sci USA. 2012;109:1357–8. https://doi.org/10.1073/pnas.1120531109.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Javani Jouni F, Abdolmaleki P, Movahedin M. Investigation on the effect of static magnetic field up to 15 mT on the viability and proliferation rate of rat bone marrow stem cells. In Vitro Cell Dev Biol Anim. 2013;49:212–9. https://doi.org/10.1007/s11626-013-9580-x.

    Article  PubMed  Google Scholar 

  57. Zablotskii V, Polyakova T, Dejneka A. Cells in the non-uniform magnetic world: how cells respond to high-gradient magnetic fields. BioEssays. 2018;40: e1800017. https://doi.org/10.1002/bies.201800017.

    Article  PubMed  Google Scholar 

  58. Shah B, Yin PT, Ghoshal S, Lee K-B. Multimodal magnetic core-shell nanoparticles for effective stem-cell differentiation and imaging. Angew Chem Int Ed Engl. 2013;52:6190–5. https://doi.org/10.1002/anie.201302245.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yan J, Dong L, Zhang B, Qi N. Effects of extremely low-frequency magnetic field on growth and differentiation of human mesenchymal stem cells. Electromagn Biol Med. 2010;29:165–76. https://doi.org/10.3109/01676830.2010.505490.

    Article  CAS  PubMed  Google Scholar 

  60. Lepik KV, Muslimov AR, Timin AS, Sergeev VS, Romanyuk DS, Moiseev IS, et al. Mesenchymal stem cell magnetization: magnetic multilayer microcapsule uptake, toxicity, impact on functional properties, and perspectives for magnetic delivery. Adv Healthc Mater. 2016;5:3182–90. https://doi.org/10.1002/adhm.201600843.

    Article  CAS  PubMed  Google Scholar 

  61. Szili-Torok T, Akca F. Remote magnetic navigation in atrial fibrillation. Expert Rev Med Devices. 2012;9:249–55. https://doi.org/10.1586/erd.12.11.

    Article  CAS  PubMed  Google Scholar 

  62. Huang Z, Shen Y, Pei N, Sun A, Xu J, Song Y, et al. The effect of nonuniform magnetic targeting of intracoronary-delivering mesenchymal stem cells on coronary embolisation. Biomaterials. 2013;34:9905–16. https://doi.org/10.1016/j.biomaterials.2013.08.092.

    Article  CAS  PubMed  Google Scholar 

  63. Combined targeting of lentiviral vectors and positioning of transduced cells by magnetic nanoparticles. In: Proceedings of the National Academy of Sciences of the United States of America; 2009.

  64. Yanai A, Häfeli UO, Metcalfe AL, Soema P, Addo L, Gregory-Evans CY, et al. Focused magnetic stem cell targeting to the retina using superparamagnetic iron oxide nanoparticles. Cell Transplant. 2012;21:1137–48. https://doi.org/10.3727/096368911X627435.

    Article  PubMed  Google Scholar 

  65. Stroh A, Boltze J, Sieland K, Hild K, Gutzeit C, Jung T, et al. Impact of magnetic labeling on human and mouse stem cells and their long-term magnetic resonance tracking in a rat model of Parkinson disease. Mol Imaging. 2009;8:166–78.

    Article  CAS  PubMed  Google Scholar 

  66. Ochi M. Mesenchymal stromal cell transplantation in the regeneration of articular cartilage and bone using a magnetic cell delivery system. J Am Acad Orthop Surg. 2013;21:61–2. https://doi.org/10.5435/JAAOS-21-01-61.

    Article  PubMed  Google Scholar 

  67. Sugioka T, Ochi M, Yasunaga Y, Adachi N, Yanada S. Accumulation of magnetically labeled rat mesenchymal stem cells using an external magnetic force, and their potential for bone regeneration. J Biomed Mater Res A. 2008;85:597–604.

    Article  PubMed  Google Scholar 

  68. Kobayashi T, Ochi M, Yanada S, Ishikawa M, Adachi N, Deie M, et al. A novel cell delivery system using magnetically labeled mesenchymal stem cells and an external magnetic device for clinical cartilage repair. Arthroscopy. 2008;24:69–76. https://doi.org/10.1016/j.arthro.2007.08.017.

    Article  PubMed  Google Scholar 

  69. Ota Y, Kamei N, Tamaura T, Adachi N, Ochi M. Magnetic resonance imaging evaluation of cartilage repair and iron particle kinetics after magnetic delivery of stem cells. Tissue Eng Part C Methods. 2018;24:679–87. https://doi.org/10.1089/ten.TEC.2018.0263.

    Article  CAS  PubMed  Google Scholar 

  70. Mahmoud EE, Kamei G, Harada Y, Shimizu R, Kamei N, Adachi N, et al. Cell magnetic targeting system for repair of severe chronic osteochondral defect in a rabbit model. Cell Transplant. 2016;25:1073–83. https://doi.org/10.3727/096368915X689613.

    Article  PubMed  Google Scholar 

  71. Kobayashi T, Ochi M, Yanada S, Ishikawa M, Adachi N, Deie M, et al. Augmentation of degenerated human cartilage in vitro using magnetically labeled mesenchymal stem cells and an external magnetic device. Arthroscopy. 2009;25:1435–41. https://doi.org/10.1016/j.arthro.2009.06.009.

    Article  PubMed  Google Scholar 

  72. Gallet R, Dawkins J, Valle J, Simsolo E, de Couto G, Middleton R, et al. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur Heart J. 2017;38:201–11. https://doi.org/10.1093/eurheartj/ehw240.

    Article  CAS  PubMed  Google Scholar 

  73. Lee J-R, Kyung JW, Kumar H, Kwon SP, Song SY, Han I-B, et al. Targeted delivery of mesenchymal stem cell-derived nanovesicles for spinal cord injury treatment. Int J Mol Sci. 2020. https://doi.org/10.3390/ijms21114185.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Wu D, Chang X, Tian J, Kang L, Wu Y, Liu J, et al. Bone mesenchymal stem cells stimulation by magnetic nanoparticles and a static magnetic field: release of exosomal miR-1260a improves osteogenesis and angiogenesis. J Nanobiotechnol. 2021;19:209. https://doi.org/10.1186/s12951-021-00958-6.

    Article  CAS  Google Scholar 

  75. Wang D, Ma B, Zhao Y, Sun Y, Luan Y, Wang J. Preparation and properties of semi-self-assembled lipopeptide vesicles. Langmuir. 2019;35:13174–81. https://doi.org/10.1021/acs.langmuir.9b02513.

    Article  CAS  PubMed  Google Scholar 

  76. Silva AKA, Kolosnjaj-Tabi J, Bonneau S, Marangon I, Boggetto N, Aubertin K, et al. Magnetic and photoresponsive theranosomes: translating cell-released vesicles into smart nanovectors for cancer therapy. ACS Nano. 2013;7:4954–66. https://doi.org/10.1021/nn400269x.

    Article  CAS  PubMed  Google Scholar 

  77. García-Jimeno S, Escribano E, Queralt J, Estelrich J. External magnetic field-induced selective biodistribution of magnetoliposomes in mice. Nanoscale Res Lett. 2012;7:452. https://doi.org/10.1186/1556-276X-7-452.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Nappini S, Bombelli FB, Bonini M, Nordèn B, Baglioni P. Magnetoliposomes for controlled drug release in the presence of low-frequency magnetic field. Soft Matter. 2009;6:154–62.

    Article  Google Scholar 

  79. Bulte JW, De Cuyper M. Magnetoliposomes as contrast agents. Methods Enzymol. 2003;373:175–98.

    Article  CAS  PubMed  Google Scholar 

  80. Fattahi H, Laurent S, Liu F, Arsalani N, Vander Elst L, Muller RN. Magnetoliposomes as multimodal contrast agents for molecular imaging and cancer nanotheragnostics. Nanomedicine (Lond). 2011;6:529–44. https://doi.org/10.2217/nnm.11.14.

    Article  CAS  Google Scholar 

  81. Sanson C, Diou O, Thévenot J, Ibarboure E, Soum A, Brûlet A, et al. Doxorubicin loaded magnetic polymersomes: theranostic nanocarriers for MR imaging and magneto-chemotherapy. ACS Nano. 2011;5:1122–40. https://doi.org/10.1021/nn102762f.

    Article  CAS  PubMed  Google Scholar 

  82. Rodrigues ARO, Gomes IT, Almeida BG, Araújo JP, Castanheira EMS, Coutinho PJG. Magnetic liposomes based on nickel ferrite nanoparticles for biomedical applications. Phys Chem Chem Phys. 2015;17:18011–21. https://doi.org/10.1039/c5cp01894c.

    Article  CAS  PubMed  Google Scholar 

  83. Reimhult E. Nanoparticle-triggered release from lipid membrane vesicles. New Biotechnol. 2015;32:665–72. https://doi.org/10.1016/j.nbt.2014.12.002.

    Article  CAS  Google Scholar 

  84. Oliveira H, Pérez-Andrés E, Thevenot J, Sandre O, Berra E, Lecommandoux S. Magnetic field triggered drug release from polymersomes for cancer therapeutics. J Control Release. 2013;169:165–70. https://doi.org/10.1016/j.jconrel.2013.01.013.

    Article  CAS  PubMed  Google Scholar 

  85. Pradhan P, Giri J, Rieken F, Koch C, Mykhaylyk O, Döblinger M, et al. Targeted temperature sensitive magnetic liposomes for thermo-chemotherapy. J Control Release. 2010;142:108–21. https://doi.org/10.1016/j.jconrel.2009.10.002.

    Article  CAS  PubMed  Google Scholar 

  86. Fortin-Ripoche JP, Martina MS, Gazeau F, Menager C, Wilhelm C, Bacri JC, et al. Magnetic targeting of magnetoliposomes to solid tumors with MR imaging monitoring in mice: feasibility. Radiology. 2006;239:415–24.

    Article  PubMed  Google Scholar 

  87. Skouras A, Papadia K, Mourtas S, Klepetsanis P, Antimisiaris SG. Multifunctional doxorubicin-loaded magnetoliposomes with active and magnetic targeting properties. Eur J Pharm Sci. 2018;123:162–72. https://doi.org/10.1016/j.ejps.2018.07.044.

    Article  CAS  PubMed  Google Scholar 

  88. Feng A, Yuan J. Smart nanocontainers: progress on novel stimuli-responsive polymer vesicles. Macromol Rapid Commun. 2014;35:767–79. https://doi.org/10.1002/marc.201300866.

    Article  CAS  PubMed  Google Scholar 

  89. Long CM, Bulte JWM. In vivo tracking of cellular therapeutics using magnetic resonance imaging. Expert Opin Biol Ther. 2009;9:293–306. https://doi.org/10.1517/14712590802715723.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Hinds KA, Hill JM, Shapiro EM, Laukkanen MO, Silva AC, Combs CA, et al. Highly efficient endosomal labeling of progenitor and stem cells with large magnetic particles allows magnetic resonance imaging of single cells. Blood. 2003;102:867–72.

    Article  CAS  PubMed  Google Scholar 

  91. Hill JM. Serial cardiac magnetic resonance imaging of injected mesenchymal stem cells. Circulation. 2003;108:1009–14.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Connell JJ, Patrick PS, Yu Y, Lythgoe MF, Kalber TL. Advanced cell therapies: targeting, tracking and actuation of cells with magnetic particles. Regen Med. 2015;10:757–72. https://doi.org/10.2217/rme.15.36.

    Article  CAS  PubMed  Google Scholar 

  93. Stroh A, Faber C, Neuberger T, Lorenz P, Sieland K, Jakob PM, et al. In vivo detection limits of magnetically labeled embryonic stem cells in the rat brain using high-field (17.6 T) magnetic resonance imaging. Neuroimage. 2005;24:635–45.

    Article  PubMed  Google Scholar 

  94. Crabbe A, Vandeputte C, Dresselaers T, Sacido AA, Verdugo JMG, Eyckmans J, et al. Effects of MRI contrast agents on the stem cell phenotype. Cell Transplant. 2010;19:919–36. https://doi.org/10.3727/096368910X494623.

    Article  PubMed  Google Scholar 

  95. Zhou S, Yin T, Zou Q, Zhang K, Gao G, Shapter JG, et al. Labeling adipose derived stem cell sheet by ultrasmall super-paramagnetic FeO nanoparticles and magnetic resonance tracking in vivo. Sci Rep. 2017;7:42793. https://doi.org/10.1038/srep42793.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Fan J, Tan Y, Jie L, Wu X, Yu R, Zhang M. Biological activity and magnetic resonance imaging of superparamagnetic iron oxide nanoparticles-labeled adipose-derived stem cells. Stem Cell Res Ther. 2013;4:44. https://doi.org/10.1186/scrt191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Ha S, Ahn S, Kim S, Joo Y, Chong YH, Suh Y-H, et al. In vivo imaging of human adipose-derived stem cells in Alzheimer’s disease animal model. J Biomed Opt. 2014;19: 051206. https://doi.org/10.1117/1.JBO.19.5.051206.

    Article  CAS  PubMed  Google Scholar 

  98. Elfick A, Rischitor G, Mouras R, Azfer A, Lungaro L, Uhlarz M, et al. Biosynthesis of magnetic nanoparticles by human mesenchymal stem cells following transfection with the magnetotactic bacterial gene mms6. Sci Rep. 2017;7:39755. https://doi.org/10.1038/srep39755.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Dabrowska S, Del Fattore A, Karnas E, Frontczak-Baniewicz M, Kozlowska H, Muraca M, et al. Imaging of extracellular vesicles derived from human bone marrow mesenchymal stem cells using fluorescent and magnetic labels. Int J Nanomed. 2018;13:1653–64. https://doi.org/10.2147/IJN.S159404.

    Article  CAS  Google Scholar 

  100. Chang M, Hsiao J-K, Yao M, Chien L-Y, Hsu S-C, Ko B-S, et al. Homologous RBC-derived vesicles as ultrasmall carriers of iron oxide for magnetic resonance imaging of stem cells. Nanotechnology. 2010;21: 235103. https://doi.org/10.1088/0957-4484/21/23/235103.

    Article  CAS  PubMed  Google Scholar 

  101. Liu T, Zhu Y, Zhao R, Wei X, Xin X. Visualization of exosomes from mesenchymal stem cells in vivo by magnetic resonance imaging. Magn Reson Imaging. 2020;68:75–82. https://doi.org/10.1016/j.mri.2020.02.001.

    Article  CAS  PubMed  Google Scholar 

  102. Beaune G, Ménager C, Cabuil V. Location of magnetic and fluorescent nanoparticles encapsulated inside giant liposomes. J Phys Chem B. 2008;112:7424–9. https://doi.org/10.1021/jp711811u.

    Article  CAS  PubMed  Google Scholar 

  103. Martina M-S, Fortin J-P, Fournier L, Ménager C, Gazeau F, Clément O, et al. Magnetic targeting of rhodamine-labeled superparamagnetic liposomes to solid tumors: in vivo tracking by fibered confocal fluorescence microscopy. Mol Imaging. 2007;6:140–6.

    Article  CAS  PubMed  Google Scholar 

  104. Beaune G, Dubertret B, Clément O, Vayssettes C, Cabuil V, Ménager C. Giant vesicles containing magnetic nanoparticles and quantum dots: feasibility and tracking by fiber confocal fluorescence microscopy. Angew Chem Int Ed Engl. 2007;46:5421–4.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Funding information is not applicable.

Author information

Authors and Affiliations

Authors

Contributions

Y.C. carried out data statistics and drafted the manuscript. S.H. was responsible for manuscript supervision. Both authors read and approved the final manuscript.

Corresponding author

Correspondence to Yuling Chen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Y., Hou, S. Application of magnetic nanoparticles in cell therapy. Stem Cell Res Ther 13, 135 (2022). https://doi.org/10.1186/s13287-022-02808-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-022-02808-0

Keywords