1
|
Zimina T, Sitkov N, Brusina K, Fedorov V, Mikhailova N, Testov D, Gareev K, Samochernykh K, Combs S, Shevtsov M. Magnetically Controlled Transport of Nanoparticles in Solid Tumor Tissues and Porous Media Using a Tumor-on-a-Chip Format. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:2030. [PMID: 39728566 DOI: 10.3390/nano14242030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
This study addresses issues in developing spatially controlled magnetic fields for particle guidance, synthesizing biocompatible and chemically stable MNPs and enhancing their specificity to pathological cells through chemical modifications, developing personalized adjustments, and highlighting the potential of tumor-on-a-chip systems, which can simulate tissue environments and assess drug efficacy and dosage in a controlled setting. The research focused on two MNP types, uncoated magnetite nanoparticles (mMNPs) and carboxymethyl dextran coated superparamagnetic nanoparticles (CD-SPIONs), and evaluated their transport properties in microfluidic systems and porous media. The original uncoated mMNPs of bimodal size distribution and the narrow size distribution of the fractions (23 nm and 106 nm by radii) were demonstrated to agglomerate in magnetically driven microfluidic flow, forming a stable stationary web consisting of magnetic fibers within 30 min. CD-SPIONs were demonstrated to migrate in agar gel with the mean pore size equal to or slightly higher than the particle size. The migration velocity was inversely proportional to the size of particles. No compression of the gel was observed under the magnetic field gradient of 40 T/m. In the brain tissue, particles of sizes 220, 350, 820 nm were not penetrating the tissue, while the compression of tissue was observed. The particles of 95 nm size penetrated the tissue at the edge of the sample, and no compression was observed. For all particles, movement through capillary vessels was observed.
Collapse
Affiliation(s)
- Tatiana Zimina
- Department of Micro and Nanoelectronics, St. Petersburg Electrotechnical University "LETI" (ETU "LETI"), Prof. Popova Str., 5, 197022 St. Petersburg, Russia
| | - Nikita Sitkov
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
| | - Ksenia Brusina
- Department of Micro and Nanoelectronics, St. Petersburg Electrotechnical University "LETI" (ETU "LETI"), Prof. Popova Str., 5, 197022 St. Petersburg, Russia
| | - Viacheslav Fedorov
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
| | - Natalia Mikhailova
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
| | - Dmitriy Testov
- Department of Micro and Nanoelectronics, St. Petersburg Electrotechnical University "LETI" (ETU "LETI"), Prof. Popova Str., 5, 197022 St. Petersburg, Russia
| | - Kamil Gareev
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
| | - Konstantin Samochernykh
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
| | - Stephanie Combs
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany
| | - Maxim Shevtsov
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
2
|
Chen Y, Hou S. Application of magnetic nanoparticles in cell therapy. Stem Cell Res Ther 2022; 13:135. [PMID: 35365206 PMCID: PMC8972776 DOI: 10.1186/s13287-022-02808-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
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.
Collapse
Affiliation(s)
- Yuling Chen
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China. .,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China.
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| |
Collapse
|
3
|
Gong N, Zhang Y, Teng X, Wang Y, Huo S, Qing G, Ni Q, Li X, Wang J, Ye X, Zhang T, Chen S, Wang Y, Yu J, Wang PC, Gan Y, Zhang J, Mitchell MJ, Li J, Liang XJ. Proton-driven transformable nanovaccine for cancer immunotherapy. NATURE NANOTECHNOLOGY 2020; 15:1053-1064. [PMID: 33106640 PMCID: PMC7719078 DOI: 10.1038/s41565-020-00782-3] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 09/16/2020] [Indexed: 05/05/2023]
Abstract
Cancer vaccines hold great promise for improved cancer treatment. However, endosomal trapping and low immunogenicity of tumour antigens usually limit the efficiency of vaccination strategies. Here, we present a proton-driven nanotransformer-based vaccine, comprising a polymer-peptide conjugate-based nanotransformer and loaded antigenic peptide. The nanotransformer-based vaccine induces a strong immune response without substantial systemic toxicity. In the acidic endosomal environment, the nanotransformer-based vaccine undergoes a dramatic morphological change from nanospheres (about 100 nanometres in diameter) into nanosheets (several micrometres in length or width), which mechanically disrupts the endosomal membrane and directly delivers the antigenic peptide into the cytoplasm. The re-assembled nanosheets also boost tumour immunity via activation of specific inflammation pathways. The nanotransformer-based vaccine effectively inhibits tumour growth in the B16F10-OVA and human papilloma virus-E6/E7 tumour models in mice. Moreover, combining the nanotransformer-based vaccine with anti-PD-L1 antibodies results in over 83 days of survival and in about half of the mice produces complete tumour regression in the B16F10 model. This proton-driven transformable nanovaccine offers a robust and safe strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuxuan Zhang
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xucong Teng
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China
| | - Yongchao Wang
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
| | - Shuaidong Huo
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Science, Xiamen University, Xiamen, China
| | - Guangchao Qing
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
| | - Qiankun Ni
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xianlei Li
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinjin Wang
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoxia Ye
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
| | - Tingbin Zhang
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
| | - Shizhu Chen
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry and Environmental Science, Hebei University, Baoding, China
| | - Yongji Wang
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China
| | - Jie Yu
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Paul C Wang
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
- Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington DC, USA
- Department of Electrical Engineering, Fu Jen Catholic University, Taipei, Taiwan
| | - Yaling Gan
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry and Environmental Science, Hebei University, Baoding, China
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jinghong Li
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China.
| | - Xing-Jie Liang
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry and Environmental Science, Hebei University, Baoding, China.
| |
Collapse
|
4
|
Ryzhkov A, Raikher Y. Coarse-Grained Molecular Dynamics Modelling of a Magnetic Polymersome. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E763. [PMID: 30261672 PMCID: PMC6215137 DOI: 10.3390/nano8100763] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 09/21/2018] [Accepted: 09/23/2018] [Indexed: 11/16/2022]
Abstract
A coarse-grained molecular dynamics framework is proposed to investigate the equilibrium structure and quasi-static deformational response of a magnetic polymersome, a hollow object whose magnetoactive part is its shell (membrane). In the developed scheme, the shell is modelled as a pair of two concentric interfaces, between which a layer of a linearly viscous fluid filled with magnetic nanoparticles is confined; the thickness of this layer slightly exceeds the nanoparticle diameter. The shell boundaries possess weak bending elasticity, very high surface tension and are impermeable for the nanoparticles. The nanoparticles bear permanent magnetic moments and are translationally and rotationally free inside the layer. The factors favoring the particle aggregation are the magneto-dipole coupling and Zeeman interaction with the external field; the impeding factors are thermal motion and steric restrictions imposed by the boundaries. The volume content of magnetic phase in the shell is sufficiently small (below 11 vol.%) to enable one to clearly observe structure patterns occurring in the basic state and under an applied magnetic field. As shown, both the particle concentration and the level of interparticle interaction strongly affect the extent and type of the aggregation that, in turn, causes overall deformation of the polymersome: stretching along the applied field and shrinking in the transverse plane.
Collapse
Affiliation(s)
- Aleksandr Ryzhkov
- Department of Applied Physics, Perm National Research Polytechnic University, Perm 614990, Russia.
- Laboratory of Physics and Mechanics of Soft Matter, Institute of Continuous Media Mechanics, Russian Academy of Sciences, Ural Branch, Perm 614013, Russia.
| | - Yuriy Raikher
- Laboratory of Physics and Mechanics of Soft Matter, Institute of Continuous Media Mechanics, Russian Academy of Sciences, Ural Branch, Perm 614013, Russia.
- Department of Theoretical and Mathematical Physics, Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620089, Russia.
| |
Collapse
|
5
|
Effects of static magnetic fields on natural or magnetized mesenchymal stromal cells: Repercussions for magnetic targeting. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2075-2085. [PMID: 29933023 DOI: 10.1016/j.nano.2018.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/16/2018] [Accepted: 06/05/2018] [Indexed: 01/12/2023]
Abstract
The magnetic targeting (MT) technique improves delivery of mesenchymal stromal cells (MSCs) to target sites. However, the moderate-intensity static magnetic fields (SMF) used for MT may exert adverse effects on MSCs. Thus, we aimed to evaluate the effects of SMF on MSCs in vitro. Cells were initially magnetized using citrate-coated magnetite nanoparticles. Then, control and magnetized MSCs were transferred to an in vitro MT system and exposed to 0.3-0.45 Tesla SMFs. MSC viability, morphology, ultrastructure, proliferation rates, differentiation, and immunomodulation were evaluated after 24 and 48 hours of exposure. MSCs temporarily lost viability and exhibited ultrastructural changes after exposure to SMFs, regardless of magnetization. Moreover, exposure to SMF reduced magnetized MSC proliferation rates. Nevertheless, MSCs remained functional (i.e., capable of differentiating, secreting repair mediators, and modulating alveolar macrophage phenotype). Thus, the experimental protocol tested in this experiment can be applied in future in vivo MT studies.
Collapse
|
6
|
Mapping the microscale origins of magnetic resonance image contrast with subcellular diamond magnetometry. Nat Commun 2018; 9:131. [PMID: 29317627 PMCID: PMC5760582 DOI: 10.1038/s41467-017-02471-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 12/03/2017] [Indexed: 12/18/2022] Open
Abstract
Magnetic resonance imaging (MRI) is a widely used biomedical imaging modality that derives much of its contrast from microscale magnetic field patterns in tissues. However, the connection between these patterns and the appearance of macroscale MR images has not been the subject of direct experimental study due to a lack of methods to map microscopic fields in biological samples. Here, we optically probe magnetic fields in mammalian cells and tissues with submicron resolution and nanotesla sensitivity using nitrogen-vacancy diamond magnetometry, and combine these measurements with simulations of nuclear spin precession to predict the corresponding MRI contrast. We demonstrate the utility of this technology in an in vitro model of macrophage iron uptake and histological samples from a mouse model of hepatic iron overload. In addition, we follow magnetic particle endocytosis in live cells. This approach bridges a fundamental gap between an MRI voxel and its microscopic constituents. Magnetic resonance imaging derives its contrast from local magnetic fields, however the connection between these fields and macroscale contrast has not been established through direct experiments. Here, Davis et al. use diamond magnetometry to map local magnetic fields within mammalian cells with sub-micron resolution and predict macroscale contrast.
Collapse
|
7
|
Filippi M, Boido M, Pasquino C, Garello F, Boffa C, Terreno E. Successful in vivo MRI tracking of MSCs labeled with Gadoteridol in a Spinal Cord Injury experimental model. Exp Neurol 2016; 282:66-77. [DOI: 10.1016/j.expneurol.2016.05.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/26/2016] [Accepted: 05/18/2016] [Indexed: 01/02/2023]
|
8
|
Chen H, Wang Y, Wang T, Shi D, Sun Z, Xia C, Wang B. Application prospective of nanoprobes with MRI and FI dual-modality imaging on breast cancer stem cells in tumor. J Nanobiotechnology 2016; 14:52. [PMID: 27339420 PMCID: PMC4918029 DOI: 10.1186/s12951-016-0195-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/20/2016] [Indexed: 12/30/2022] Open
Abstract
Breast cancer (BC) is a serious disease to threat lives of women. Numerous studies have proved that BC originates from cancer stem cells (CSCs). But at present, no one approach can quickly and simply identify breast cancer stem cells (BCSCs) in solid tumor. Nanotechnology is probably able to realize this goal. But in study process, scientists find it seems that nanomaterials with one modality, such as magnetic resonance imaging (MRI) or fluorescence imaging (FI), have their own advantages and drawbacks. They cannot meet practical requirements in clinic. The nanoprobe combined MRI with FI modality is a promising tool to accurately detect desired cells with low amount in tissue. In this work, we briefly describe the MRI and FI development history, analyze advantages and disadvantages of nanomaterials with single modality in cancer cell detection. Then the application development of nanomaterials with dual-modality in cancer field is discussed. Finally, the obstacles and prospective of dual-modal nanoparticles in detection field of BCSCs are also pointed out in order to speed up clinical applications of nanoprobes.
Collapse
Affiliation(s)
- Hetao Chen
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, No. 22, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Yu Wang
- Department of Chemistry, Qiqihaer Medical College, Qiqihaer, 161006, Heilongjiang Province, China
| | - Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, No. 22, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Dongxing Shi
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, No. 22, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Zengrong Sun
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, No. 22, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Chunhui Xia
- Department of Chemistry, Qiqihaer Medical College, Qiqihaer, 161006, Heilongjiang Province, China.
| | - Baiqi Wang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, No. 22, Qixiangtai Road, Heping District, Tianjin, 300070, China.
| |
Collapse
|
9
|
Bud-neck scaffolding as a possible driving force in ESCRT-induced membrane budding. Biophys J 2015; 108:833-843. [PMID: 25692588 PMCID: PMC4336374 DOI: 10.1016/j.bpj.2014.12.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 12/20/2014] [Accepted: 12/23/2014] [Indexed: 01/03/2023] Open
Abstract
Membrane budding is essential for processes such as protein sorting and transport. Recent experimental results with ESCRT proteins reveal a novel budding mechanism, with proteins emerging in bud necks but separated from the entire bud surface. Using an elastic model, we show that ESCRT protein shapes are sufficient to spontaneously create experimentally observed structures, with protein-membrane interactions leading to protein scaffolds in bud-neck regions. Furthermore, the model reproduces experimentally observed budding directions and bud sizes. Finally, our results reveal that membrane-mediated sorting has the capability of creating structures more complicated than previously assumed.
Collapse
|
10
|
A refined theory of magnetoelastic buckling matches experiments with ferromagnetic and superparamagnetic rods. Proc Natl Acad Sci U S A 2015; 112:7135-40. [PMID: 26015577 DOI: 10.1073/pnas.1422534112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In its simplest form the magnetoelastic buckling instability refers to the sudden bending transition of an elastic rod experiencing a uniform induction field applied at a normal angle with respect to its long axis. This fundamental physics phenomenon was initially documented in 1968, and, surprisingly, despite many refinements, a gap has always remained between the observations and the theoretical expectations. Here, we first renew the theory with a simple model based on the assumption that the magnetization follows the rod axis as soon as it bends. We demonstrate that the magnetoelastic buckling corresponds to a classical Landau second-order transition. Our model yields a solution for the critical field as well as the shape of the deformed rods which we compare with experiments on flexible ferromagnetic nickel rods at the centimeter scale. We also report this instability at the micrometer scale with specially designed rods made of nanoparticles. We characterized our samples by determining all of the relevant parameters (radius, length, Young modulus, magnetic susceptibility) and, using these values, we found that the theory fits extremely well the experimental results for both systems without any adjustable parameter. The superparamagnetic feature of the microrods also highlights the fact that ferromagnetic systems break the symmetry before the buckling. We propose a magnetic "stick-slip" model to explain this peculiar feature, which was visible in past reports but never detailed.
Collapse
|
11
|
Kolosnjaj-Tabi J, Wilhelm C, Clément O, Gazeau F. Cell labeling with magnetic nanoparticles: opportunity for magnetic cell imaging and cell manipulation. J Nanobiotechnology 2013; 11 Suppl 1:S7. [PMID: 24564857 PMCID: PMC4029272 DOI: 10.1186/1477-3155-11-s1-s7] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This tutorial describes a method of controlled cell labeling with citrate-coated ultra small superparamagnetic iron oxide nanoparticles. This method may provide basically all kinds of cells with sufficient magnetization to allow cell detection by high-resolution magnetic resonance imaging (MRI) and to enable potential magnetic manipulation. In order to efficiently exploit labeled cells, quantify the magnetic load and deliver or follow-up magnetic cells, we herein describe the main requirements that should be applied during the labeling procedure. Moreover we present some recommendations for cell detection and quantification by MRI and detail magnetic guiding on some real-case studies in vitro and in vivo.
Collapse
|
12
|
Wilhelm C, Gazeau F. [Magnetic nanoparticles as tools for cell therapy]. Biol Aujourdhui 2013; 206:273-84. [PMID: 23419254 DOI: 10.1051/jbio/2012024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Indexed: 11/15/2022]
Abstract
Labelling living cells with magnetic nanoparticles creates opportunities for numerous biomedical applications such as Magnetic Resonance Imaging (MRI) cell tracking, cell manipulation, cell patterning for tissue engineering and magnetically-assisted cell delivery. The unique advantage of magnetic-based methods is to activate or monitor cell behavior by a remote stimulus, the magnetic field. Cell labelling methods using superparamagnetic nanoparticles have been widely developed, showing no adverse effect on cell proliferation and functionalities while conferring magnetic properties to various cell types. This paper first describes how cells can become responsive to magnetic field by safely internalizing magnetic nanoparticles. We next show how magnetic cells can be detected by MRI, giving the opportunity for non-invasive in vivo monitoring of cell migration. We exemplify the fact that MRI cell tracking has become a method of choice to follow the fate of administrated cells in cell therapy assay, whether the cells are grafted locally or administrated in the circulation. Finally we give different examples of magnetic manipulation of cells and their applications to regenerative medicine. Magnetic cell manipulation are forecasted to be more and more developed, in order to improve tissue engineering technique and assist cell-based therapies. Owing to the clinical approval of iron-oxide nanoparticles as MRI contrast agent, there is no major obstacle in the translation to human clinics of the magnetic methods summarized in this paper.
Collapse
Affiliation(s)
- Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes, CNRS – Université Paris Diderot, 75205 Paris Cedex 13, France.
| | | |
Collapse
|
13
|
Petrichenko O, Erglis K, Cēbers A, Plotniece A, Pajuste K, Béalle G, Ménager C, Dubois E, Perzynski R. Bilayer properties of giant magnetic liposomes formed by cationic pyridine amphiphile and probed by active deformation under magnetic forces. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2013; 36:9. [PMID: 23359032 DOI: 10.1140/epje/i2013-13009-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 01/16/2013] [Indexed: 06/01/2023]
Abstract
We synthesize giant magnetic liposomes by a reverse-phase evaporation method (REV) using a new self-assembling Cationic Pyridine Amphiphile (CPA) derived from 1,4-dihydropyridine as liposome-forming agent and a magnetic ferrofluid based on γ-Fe(2)O(3) nanoparticles. Having in view the potential interest of CPA in targeted transport by magnetic forces, the mechanical elastic properties of such bilayers are here directly investigated in vesicles loaded with magnetic nanoparticles. Bending elastic modulus K(b) ∼ 0.2 to 5k(B)T and pre-stress τ ∼ 3.2 to 12.10(-6) erg/cm(2) are deduced from the under-field deformations of the giant magnetic liposomes. The obtained K(b) values are discussed in terms of A. Wurgers's theory.
Collapse
Affiliation(s)
- O Petrichenko
- University of Latvia, Zeļļu-8, LV-1002, Rıga, Latvia
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lévy M, Wilhelm C, Devaud M, Levitz P, Gazeau F. How cellular processing of superparamagnetic nanoparticles affects their magnetic behavior and NMR relaxivity. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 7:373-83. [PMID: 22649043 DOI: 10.1002/cmmi.504] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cellular processing of nanomaterials may affect their physical properties at the root of various biomedical applications. When nanoparticles interact with living cells, their spatial distribution is progressively modified by cellular activity, which tends to concentrate them into intracellular compartments, changing in turn their responsivity to physical stimuli. In this paper, we investigate the consequences of cellular uptake on the related magnetic properties and NMR relaxivity of iron oxide nanoparticles. The superparamagnetic behavior (field-dependent and temperature-dependent magnetization curves investigated by SQUID (Superconducting Quantum Interference Device) measurements) and nuclear magnetic relaxation dispersion (NMRD) R(1) profiles of citrate-coated maghemite nanoparticles (mean diameter 8 nm) were characterized in colloidal suspension and after being uptaken by several types of cells (tumor cells, stem cells and macrophages). The temperature-dependent magnetization as well as the NMRD profile were changed following cellular uptake depending on the stage of endocytosis process while the field-dependent magnetization at room temperature remained unchanged. Magnetic coupling between nanoparticles confined in cell lysosomes accounts for the modification in magnetic behavior, thereby reflecting the local organization of nanoparticles. NMR longitudinal relaxivity was directly sensitive to the intracellular distribution of nanoparticles, in line with Transmission Electron Microscopy TEM observations. This study is the first attempt to link up magnetic properties and NMR characterization of iron oxide nanoparticles before and after their cell processing.
Collapse
Affiliation(s)
- Michael Lévy
- Laboratoire Matière et Systèmes Complexes, UMR CNRS/Université Paris - Diderot, France
| | | | | | | | | |
Collapse
|
15
|
Mok H, Zhang M. Superparamagnetic iron oxide nanoparticle-based delivery systems for biotherapeutics. Expert Opin Drug Deliv 2012. [PMID: 23199200 DOI: 10.1517/17425247.2013.747507] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Superparamagnetic iron oxide nanoparticle (SPION)-based carrier systems have many advantages over other nanoparticle-based systems. They are biocompatible, biodegradable, facilely tunable and superparamagnetic and thus controllable by an external magnetic field. These attributes enable their broad biomedical applications. In particular, magnetically driven carriers are drawing considerable interest as an emerging therapeutic delivery system because of their superior delivery efficiency. AREAS COVERED This article reviews the recent advances in use of SPION-based carrier systems to improve the delivery efficiency and target specificity of biotherapeutics. The authors examine various formulations of SPION-based delivery systems, including SPION micelles, clusters, hydrogels, liposomes and micro/nanospheres, as well as their specific applications in delivery of biotherapeutics. EXPERT OPINION Recently, biotherapeutics including therapeutic cells, proteins and genes have been studied as alternative treatments to various diseases. Despite the advantages of high target specificity and low adverse effects, clinical translation of biotherapeutics has been hindered by the poor stability and low delivery efficiency compared with chemical drugs. Accordingly, biotherapeutic delivery systems that can overcome these limitations are actively pursued. SPION-based materials can be ideal candidates for developing such delivery systems because of their excellent biocompatibility and superparamagnetism that enables long-term accumulation/retention at target sites by utilization of a suitable magnet. In addition, synthesis technologies for production of finely tuned, homogeneous SPIONs have been well developed, which may promise their rapid clinical translation.
Collapse
Affiliation(s)
- Hyejung Mok
- Konkuk University, Department of Bioscience and Biotechnology, Seoul 143-701, Republic of Korea
| | | |
Collapse
|
16
|
Różycki B, Boura E, Hurley JH, Hummer G. Membrane-elasticity model of Coatless vesicle budding induced by ESCRT complexes. PLoS Comput Biol 2012; 8:e1002736. [PMID: 23093927 PMCID: PMC3475702 DOI: 10.1371/journal.pcbi.1002736] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 08/23/2012] [Indexed: 01/08/2023] Open
Abstract
The formation of vesicles is essential for many biological processes, in particular for the trafficking of membrane proteins within cells. The Endosomal Sorting Complex Required for Transport (ESCRT) directs membrane budding away from the cytosol. Unlike other vesicle formation pathways, the ESCRT-mediated budding occurs without a protein coat. Here, we propose a minimal model of ESCRT-induced vesicle budding. Our model is based on recent experimental observations from direct fluorescence microscopy imaging that show ESCRT proteins colocalized only in the neck region of membrane buds. The model, cast in the framework of membrane elasticity theory, reproduces the experimentally observed vesicle morphologies with physically meaningful parameters. In this parameter range, the minimum energy configurations of the membrane are coatless buds with ESCRTs localized in the bud neck, consistent with experiment. The minimum energy configurations agree with those seen in the fluorescence images, with respect to both bud shapes and ESCRT protein localization. On the basis of our model, we identify distinct mechanistic pathways for the ESCRT-mediated budding process. The bud size is determined by membrane material parameters, explaining the narrow yet different bud size distributions in vitro and in vivo. Our membrane elasticity model thus sheds light on the energetics and possible mechanisms of ESCRT-induced membrane budding.
Collapse
Affiliation(s)
- Bartosz Różycki
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Max Planck Institute of Colloids and Interfaces, Department of Theory and Bio-Systems, Potsdam, Germany
| | - Evzen Boura
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - James H. Hurley
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gerhard Hummer
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
17
|
Safi M, Courtois J, Seigneuret M, Conjeaud H, Berret JF. The effects of aggregation and protein corona on the cellular internalization of iron oxide nanoparticles. Biomaterials 2011; 32:9353-63. [PMID: 21911254 DOI: 10.1016/j.biomaterials.2011.08.048] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 08/16/2011] [Indexed: 11/16/2022]
Abstract
Engineered inorganic nanoparticles are essential components in the development of nanotechnologies. For applications in nanomedicine, particles need to be functionalized to ensure a good dispersibility in biological fluids. In many cases however, functionalization is not sufficient: the particles become either coated by a corona of serum proteins or precipitate out of the solvent. In the present paper, we show that by changing the coating of iron oxide nanoparticles from a low-molecular weight ligand (citrate ions) to small carboxylated polymers (poly(acrylic acid)), the colloidal stability of the dispersion is improved and the adsorption/internalization of iron toward living mammalian cells is profoundly affected. Citrate-coated particles are shown to destabilize in all fetal-calf-serum based physiological conditions tested, whereas the polymer coated particles exhibit an outstanding dispersibility as well as a structure devoid of protein corona. The interactions between nanoparticles and human lymphoblastoid cells are investigated by transmission electron microscopy and flow cytometry. Two types of nanoparticle/cell interactions are underlined. Iron oxides are found either adsorbed on the cellular membranes, or internalized into membrane-bound endocytosis compartments. For the precipitating citrate-coated particles, the kinetics of interactions reveal a massive and rapid adsorption of iron oxide on the cell surfaces. The quantification of the partition between adsorbed and internalized iron was performed from the cytometry data. The results highlight the importance of resilient adsorbed nanomaterials at the cytoplasmic membrane.
Collapse
Affiliation(s)
- M Safi
- Matière et Systèmes Complexes, UMR 7057 CNRS Université Denis Diderot Paris-VII, Bâtiment Condorcet 10 rue Alice Domon et Léonie Duquet, 75205 Paris, France
| | | | | | | | | |
Collapse
|
18
|
Magnetic nanoparticles as targeted delivery systems in oncology. Radiol Oncol 2011; 45:1-16. [PMID: 22933928 PMCID: PMC3423716 DOI: 10.2478/v10019-011-0001-z] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 01/05/2011] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Many different types of nanoparticles, magnetic nanoparticles being just a category among them, offer exciting opportunities for technologies at the interfaces between chemistry, physics and biology. Some magnetic nanoparticles have already been utilized in clinical practice as contrast enhancing agents for magnetic resonance imaging (MRI). However, their physicochemical properties are constantly being improved upon also for other biological applications, such as magnetically-guided delivery systems for different therapeutics. By exposure of magnetic nanoparticles with attached therapeutics to an external magnetic field with appropriate characteristics, they are concentrated and retained at the preferred site which enables the targeted delivery of therapeutics to the desired spot. CONCLUSIONS The idea of binding chemotherapeutics to magnetic nanoparticles has been around for 30 years, however, no magnetic nanoparticles as delivery systems have yet been approved for clinical practice. Recently, binding of nucleic acids to magnetic nanoparticles has been demonstrated as a successful non-viral transfection method of different cell lines in vitro. With the optimization of this method called magnetofection, it will hopefully become another form of gene delivery for the treatment of cancer.
Collapse
|
19
|
Kim JS, Ligler FS. Utilization of microparticles in next-generation assays for microflow cytometers. Anal Bioanal Chem 2010; 398:2373-82. [PMID: 20526882 PMCID: PMC2965807 DOI: 10.1007/s00216-010-3848-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 05/11/2010] [Accepted: 05/13/2010] [Indexed: 11/26/2022]
Abstract
Micron-sized particles have primarily been used in microfabricated flow cytometers for calibration purposes and proof-of-concept experiments. With increasing frequency, microparticles are serving as a platform for assays measured in these small analytical devices. Light scattering has been used to measure the agglomeration of antibody-coated particles in the presence of an antigen. Impedance detection is another technology being integrated into microflow cytometers for microparticle-based assays. Fluorescence is the most popular detection method in flow cytometry, enabling highly sensitive multiplexed assays. Finally, magnetic particles have also been used to measure antigen levels using a magnetophoretic micro-device. We review the progress of microparticle-based assays in microflow cytometry in terms of the advantages and limitations of each approach.
Collapse
Affiliation(s)
- Jason S. Kim
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC 20375-5348, USA
| | - Frances S. Ligler
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC 20375-5348, USA
| |
Collapse
|
20
|
Lévy M, Lagarde F, Maraloiu VA, Blanchin MG, Gendron F, Wilhelm C, Gazeau F. Degradability of superparamagnetic nanoparticles in a model of intracellular environment: follow-up of magnetic, structural and chemical properties. NANOTECHNOLOGY 2010; 21:395103. [PMID: 20820094 DOI: 10.1088/0957-4484/21/39/395103] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The unique magnetic properties of iron oxide nanoparticles have paved the way for various biomedical applications, such as magnetic resonance cellular imaging or magnetically induced therapeutic hyperthermia. Living cells interact with nanoparticles by internalizing them within intracellular acidic compartments. Although no acute toxicity of iron oxide nanoparticles has been reported up to now, the mechanisms of nanoparticle degradation by the cellular environment are still unknown. In the organism, the long term integrity and physical state of iron-based nanoparticles are challenged by iron homeostasis. In this study, we monitored the degradation of 7 nm sized maghemite nanoparticles in a medium mimicking the intracellular environment. Magnetic nanoparticles with three distinct surface coatings, currently evaluated as MRI contrast agents, were shown to exhibit different kinetics of dissolution at an acidic pH in the presence of a citrate chelating agent. Our assessment of the physical state of the nanoparticles during degradation revealed that the magnetic properties, size distribution and structure of the remaining nanocrystals were identical to those of the initial suspension. This result suggests a model for nanoparticle degradation with rapidly dissolved nanocrystals and a reservoir of intact nanoparticles.
Collapse
Affiliation(s)
- Michael Lévy
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université Paris Diderot, Paris, France
| | | | | | | | | | | | | |
Collapse
|
21
|
Lunov O, Zablotskii V, Syrovets T, Röcker C, Tron K, Nienhaus GU, Simmet T. Modeling receptor-mediated endocytosis of polymer-functionalized iron oxide nanoparticles by human macrophages. Biomaterials 2010; 32:547-55. [PMID: 20880574 DOI: 10.1016/j.biomaterials.2010.08.111] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 08/31/2010] [Indexed: 01/04/2023]
Abstract
Although systemically applied nanoparticles are quickly taken up by phagocytic cells, mainly macrophages, the interactions between engineered nanoparticles and macrophages are still not well defined. We therefore analyzed the uptake of diagnostically used carboxydextran-coated superparamagnetic iron oxide nanoparticles of 60 nm (SPIO) and 20 nm (USPIO) by human macrophages. By pharmacological and in vitro knockdown approaches, the principal uptake mechanism for both particles was identified as clathrin-mediated, scavenger receptor A-dependent endocytosis. We developed a mathematical model of the uptake process that allows determination of key parameters of endocytosis, including the rate of uptake, the number of nanoparticles per cell in saturation, the mean uptake time, and the correlation between the number of internalized nanoparticles and their extracellular concentration. The calculated parameters correlate well with experimental data obtained by confocal microscopy. Moreover, the model predicts the individual and collective wrapping times of different nanoparticles, describes the relation between cytoskeletal forces, membrane elasticity and the uptake time. We also introduced a new physical parameter 'a' governing the collective uptake process, a reflecting minimal linear spacing between simultaneously acting neighboring endocytotic pits.
Collapse
Affiliation(s)
- Oleg Lunov
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
22
|
Shi Z, Neoh KG, Kang ET, Shuter B, Wang SC. Bifunctional Eu(3+)-doped Gd(2)O(3) nanoparticles as a luminescent and T(1) contrast agent for stem cell labeling. CONTRAST MEDIA & MOLECULAR IMAGING 2010; 5:105-11. [PMID: 20419762 DOI: 10.1002/cmmi.373] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Magnetic resonance tracking of stem cells has recently become an emerging application for investigating cell-tissue interactions and guiding the development of effective stem cell therapies for regeneration of damaged tissues and organs. In this work, anionic Eu(3+)-doped Gd(2)O(3) hybrid nanoparticles were applied as a contrast agent both for fluorescence microscopy and T(1)-weighted MRI. The nanoparticles were synthesized through the polyol method and further modified with citric acid to obtain anionic nanoparticles. These nanoparticles were internalized into human mesenchymal stem cells (hMSCs) as confirmed by confocal laser scanning microscopy and quantified by inductively coupled plasma-mass spectrometry. MTT assay of the labeled cells showed that the nanoparticles did not possess significant cytotoxicity. In addition, the osteogenic, adipogenic and chondrogenic differentiation of the hMSCs was not influenced by the labeling process. With MRI, the in vitro detection threshold of cells after incubation with nanoparticles at a Gd concentration of 0.5 mM for 2 h was estimated to be about 10 000 cells. The results from this study indicate that the biocompatible anionic Gd(2)O(3) nanoparticles doped with Eu(3+) show promise both as a luminescent and T(1) contrast agent for use in visualizing hMSCs.
Collapse
Affiliation(s)
- Zhilong Shi
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore
| | | | | | | | | |
Collapse
|
23
|
Vats N, Wilhelm C, Rautou PE, Poirier-Quinot M, Péchoux C, Devue C, Boulanger CM, Gazeau F. Magnetic tagging of cell-derived microparticles: new prospects for imaging and manipulation of these mediators of biological information. Nanomedicine (Lond) 2010; 5:727-38. [DOI: 10.2217/nnm.10.44] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: Submicron membrane fragments termed microparticles (MPs), which are released by apoptotic or activated cells, are newly considered as vectors of biological information and actors of pathology development. We propose the tagging of MPs with magnetic nanoparticles as a new approach allowing imaging, manipulation and targeting of cell-derived MPs. Materials & methods: MPs generated in vitro from human endothelial cells or isolated from atherosclerotic plaques were labeled using citrate-coated 8 nm iron-oxide nanoparticles. MPs were tagged with magnetic nanoparticles on their surface and detected as Annexin-V positive by flow cytometry. Results: Labeled MPs could be mobilized, isolated and manipulated at a distance in a magnetic field gradient. Magnetic mobility of labeled MPs was quantified by micromagnetophoresis. Interactions of labeled MPs with endothelial cells could be triggered and modulated by magnetic guidance. Nanoparticles served as tracers at different scales: at the subcellular level by electron microscopy, at the cellular level by histology and at the macroscopic level by MRI. Conclusion: Magnetic labeling of biogenic MPs opens new prospects for noninvasive monitoring and distal manipulations of these biological effectors.
Collapse
Affiliation(s)
- Nidhi Vats
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS & Université Paris Diderot, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris cedex 13, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS & Université Paris Diderot, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris cedex 13, France
| | - Pierre-Emmanuel Rautou
- Paris Centre de Recherche Cardiovasculaire, INSERM U970, 56 Rue Leblanc, 75737, Paris cedex 15, France
| | - Marie Poirier-Quinot
- Laboratoire U2R2M, UMR8081, CNRS & Université Paris-Sud, Centre d’Orsay, 91405, Orsay cedex, France
| | - Christine Péchoux
- Centre de Microscopie Électronique, Plateforme MIMA2, INRA, UR1196 Génomique et Physiologie de la Lactation, Domaine de Vilvert, F-78352 Jouy-en-Josas, France
| | - Cécile Devue
- Paris Centre de Recherche Cardiovasculaire, INSERM U970, 56 Rue Leblanc, 75737, Paris cedex 15, France
| | - Chantal M Boulanger
- Paris Centre de Recherche Cardiovasculaire, INSERM U970, 56 Rue Leblanc, 75737, Paris cedex 15, France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS & Université Paris Diderot, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris cedex 13, France
| |
Collapse
|
24
|
Choubey J, Bajpai AK. Investigation on magnetically controlled delivery of doxorubicin from superparamagnetic nanocarriers of gelatin crosslinked with genipin. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2010; 21:1573-1586. [PMID: 20135205 DOI: 10.1007/s10856-010-3997-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 01/08/2010] [Indexed: 05/28/2023]
Abstract
Gelatin (Type B) nanoparticles were prepared by a single W/O emulsion technique and characterized by infrared (IR) spectra, transmission electron micrographs (TEM), surface potential measurements and magnetization studies. Whereas the IR spectra clearly confirmed the presence of gelatin, genipin and doxorubicin in the loaded nanoparticles, the transmission electron micrographs (TEM) image depicts smooth surface, spherical shape and non-uniform size of nanoparticles (up to 100 nm). The prepared nanoparticles were loaded with doxorubicin, a well known anticancer drug, and in vitro release dynamics of entrapped drug was investigated as a function of various experimental factors such as percent loading of the drug, chemical architecture of the nanocarriers, and pH, temperature, ionic strength and nature of the release medium in presence and absence of magnetic field. The nanoparticles were also studied for their water sorption capacity. The drug release process was analyzed kinetically using Ficks power law and a correlation was established between the quantity of released drug and swelling of the nanoparticles.
Collapse
Affiliation(s)
- Jyoti Choubey
- Bose Memorial Research Laboratory, Department of Chemistry, Government Autonomous Science College, Jabalpur, 482 001, India
| | | |
Collapse
|
25
|
Magnetic labeling, imaging and manipulation of endothelial progenitor cells using iron oxide nanoparticles. Future Med Chem 2010; 2:397-408. [DOI: 10.4155/fmc.09.165] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Endothelial progenitor cells (EPCs), originating from bone marrow, play a significant role in the repair of ischemic tissue and injured blood vessels. They are also involved in tumor angiogenesis. The therapeutic potential of EPCs for regenerative medicine and cancer treatment calls for new methods for monitoring and controlling cell migration. This review focuses on promising magnetic methods based on the internalization of magnetic nanoparticles by EPCs. We first describe the cellular uptake of iron oxide nanoparticles depending on their surface properties. We thus review the use of MRI for the detection of labeled cells and for noninvasive follow-up of EPCs homing in sites of endothelium regeneration. Finally, we show that remotely applied magnetic forces may enable intracellular manipulation and may optimize cell-delivery strategies for localizing cell therapy to target sites.
Collapse
|
26
|
Kyrtatos PG, Lehtolainen P, Junemann-Ramirez M, Garcia-Prieto A, Price AN, Martin JF, Gadian DG, Pankhurst QA, Lythgoe MF. Magnetic tagging increases delivery of circulating progenitors in vascular injury. JACC Cardiovasc Interv 2009; 2:794-802. [PMID: 19695550 DOI: 10.1016/j.jcin.2009.05.014] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 05/20/2009] [Indexed: 11/15/2022]
Abstract
OBJECTIVES We sought to magnetically tag endothelial progenitor cells (EPCs) with a clinical agent and target them to a site of arterial injury using a magnetic device positioned outside the body. BACKGROUND Circulating EPCs are involved in physiological processes such as vascular re-endothelialization and post-ischemic neovascularization. However, the success of cell therapies depends on the ability to deliver the cells to the site of injury. METHODS Human EPCs were labeled with iron oxide superparamagnetic nanoparticles. Cell viability and differentiation were tested using flow cytometry. Following finite element modeling computer simulations and flow testing in vitro, angioplasty was performed on rat common carotid arteries to denude the endothelium and EPCs were administered with and without the presence of an external magnetic device for 12 min. RESULTS Computer simulations indicated successful external magnetic cell targeting from a vessel with flow rate similar to a rat common carotid artery; correspondingly there was a 6-fold increase in cell capture in an in vitro flow system. Targeting enhanced cell retention at the site of injury by 5-fold at 24 h after implantation in vivo. CONCLUSIONS Using an externally applied magnetic device, we have been able to enhance EPC localization at a site of common carotid artery injury. This technology could be more widely adapted to localize cells in other organs and may provide a useful tool for the systemic injection of cell therapies.
Collapse
Affiliation(s)
- Panagiotis G Kyrtatos
- Centre for Advanced Biomedical Imaging, University College London Department of Medicine and UCL Institute of Child Health, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Shen J, Zhong XM, Duan XH, Cheng LN, Hong GB, Bi XB, Liu Y. Magnetic resonance imaging of mesenchymal stem cells labeled with dual (MR and fluorescence) agents in rat spinal cord injury. Acad Radiol 2009; 16:1142-1154. [PMID: 19660710 DOI: 10.1016/j.acra.2009.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2008] [Revised: 03/14/2009] [Accepted: 03/17/2009] [Indexed: 12/31/2022]
Abstract
RATIONALE AND OBJECTIVES In vivo tracking cells using gadolinium-based contrast agents have the important advantage of providing a positive contrast on T1-weighted images, which is less likely to be confused with artifacts because of postoperative local signal voids such as metal, hemorrhage, or air. The aim of this study is to paramagnetically and fluorescently label marrow with dual agents (gadolinium-diethylene triamine penta-acetic acid [Gd-DTPA] and PEI-FluoR) and track them after transplantation into spinal cord injury (SCI) with magnetic resonance imaging (MRI). MATERIALS AND METHODS Marrow mesenchymal stem cells (MSCs) from Sprague-Dawley rats were incubated with PEI-FluoR (rhodamine-conjugated PEI-FluoR) and Gd-DTPA complex for labeling. After labeling, cellular viability, proliferation, and apoptosis were evaluated. T1 value and longevity of intracellular Gd-DTPA retention were measured on a 1.5 T MRI scanner. Thirty-six SCI rats were implanted with labeled and unlabeled MSCs and phosphate-buffered saline. Then, serial MRI and Basso-Beattie-Bresnehan (BBB) locomotor tests were performed and correlated with fluorescent microscopy. The relative signal intensity (RSL) of the engraftment in relation to normal cord was measured and the linear mixed model followed by post-hoc Bonferroni test was used to identify significant differences in RSL as well as BBB score. RESULTS MSCs could be paramagnetically and fluorescently labeled by the dual agents. The labeling did not influence the cellular viability, proliferation, and apoptosis. The longevity of Gd-DTPA retention in labeled MSCs was up to 21 days. The distribution and migration of labeled MSCs in SCI lesions could be tracked until 7 days after implantation on MRI. The relative signal intensities of SCI rats treated with labeled cells at 1 day and 3 days (1.34 +/- 0.02, 1.27 +/- 0.03) were significantly higher than rats treated with unlabeled cells (0.94 +/- 0.01, 0.99 +/- 0.02) and phosphate-buffered saline (0.91 +/- 0.01, 0.95 +/- 0.01) (P < .05). Rats treated with labeled MSCs or unlabeled MSCs achieved significantly higher BBB scores than controls at 14, 21, 28, and 35 days after injury (P < .05). CONCLUSIONS Labeling MSCs with the dual agents may enable cellular MRI and tracking in experimental spinal cord injury.
Collapse
Affiliation(s)
- Jun Shen
- Department of Radiology, The Second Affiliated Hospital, Sun Yat-sen University, 107 Yanjiang Road West, Guangzhou 510120, Guangdong, China.
| | | | | | | | | | | | | |
Collapse
|
28
|
Shubayev VI, Pisanic TR, Jin S. Magnetic nanoparticles for theragnostics. Adv Drug Deliv Rev 2009; 61:467-77. [PMID: 19389434 DOI: 10.1016/j.addr.2009.03.007] [Citation(s) in RCA: 611] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Accepted: 03/30/2009] [Indexed: 12/11/2022]
Abstract
Engineered magnetic nanoparticles (MNPs) represent a cutting-edge tool in medicine because they can be simultaneously functionalized and guided by a magnetic field. Use of MNPs has advanced magnetic resonance imaging (MRI), guided drug and gene delivery, magnetic hyperthermia cancer therapy, tissue engineering, cell tracking and bioseparation. Integrative therapeutic and diagnostic (i.e., theragnostic) applications have emerged with MNP use, such as MRI-guided cell replacement therapy or MRI-based imaging of cancer-specific gene delivery. However, mounting evidence suggests that certain properties of nanoparticles (e.g., enhanced reactive area, ability to cross cell and tissue barriers, resistance to biodegradation) amplify their cytotoxic potential relative to molecular or bulk counterparts. Oxidative stress, a 3-tier paradigm of nanotoxicity, manifests in activation of reactive oxygen species (ROS) (tier I), followed by a proinflammatory response (tier II) and DNA damage leading to cellular apoptosis and mutagenesis (tier III). Invivo administered MNPs are quickly challenged by macrophages of the reticuloendothelial system (RES), resulting in not only neutralization of potential MNP toxicity but also reduced circulation time necessary for MNP efficacy. We discuss the role of MNP size, composition and surface chemistry in their intracellular uptake, biodistribution, macrophage recognition and cytotoxicity, and review current studies on MNP toxicity, caveats of nanotoxicity assessments and engineering strategies to optimize MNPs for biomedical use.
Collapse
Affiliation(s)
- Veronica I Shubayev
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093-0629, USA.
| | | | | |
Collapse
|
29
|
Duits MHG, Li Y, Vanapalli SA, Mugele F. Mapping of spatiotemporal heterogeneous particle dynamics in living cells. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2009; 79:051910. [PMID: 19518483 DOI: 10.1103/physreve.79.051910] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 02/17/2009] [Indexed: 05/27/2023]
Abstract
Colloidal particles embedded in the cytoplasm of living mammalian cells have been found to display remarkable heterogeneity in their amplitude of motion. However, consensus on the significance and origin of this phenomenon is still lacking. We conducted experiments on Hmec-1 cells loaded with about 100 particles to reveal the intracellular particle dynamics as a function of both location and time. Central quantity in our analysis is the amplitude (A) of the individual mean-squared displacement (iMSD), averaged over a short time. Histograms of A were measured, (1) over all particles present at the same time and (2) for individual particles as a function of time. Both distributions showed significant broadening compared to particles in Newtonian liquid, indicating that the particle dynamics varies with both location and time. However, no systematic dependence of A on intracellular location was found. Both the (strong) spatial and (weak) temporal variations were further analyzed by correlation functions of A . Spatial cross correlations were rather weak down to interparticle distances of 1 microm , suggesting that the precise intracellular probe distribution is not crucial for observing a dynamic behavior that is representative for the whole cell. Temporal correlations of A decayed at approximately 10 s , possibly suggesting an intracellular reorganization at this time scale. These findings imply (1) that both individual particle dynamics and the ensemble averaged behavior in a given cell can be measured if there are enough particles per cell and (2) that the amplitude and power-law exponent of iMSDs can be used to reveal local dynamics. We illustrate this by showing how superdiffusive and subdiffusive behaviors may be hidden under an apparently diffusive global dynamics.
Collapse
Affiliation(s)
- Michael H G Duits
- Physics of Complex Fluids group, Faculty of Science and Technology and MESA+ Institute for Nanotechnology, University of Twente, 7500 AE Enschede, The Netherlands
| | | | | | | |
Collapse
|
30
|
Smirnov P, Poirier-Quinot M, Wilhelm C, Lavergne E, Ginefri JC, Combadière B, Clément O, Darrasse L, Gazeau F. In vivo single cell detection of tumor-infiltrating lymphocytes with a clinical 1.5 Tesla MRI system. Magn Reson Med 2008; 60:1292-7. [DOI: 10.1002/mrm.21812] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
31
|
Wilhelm C, Gazeau F. Universal cell labelling with anionic magnetic nanoparticles. Biomaterials 2008; 29:3161-74. [PMID: 18455232 DOI: 10.1016/j.biomaterials.2008.04.016] [Citation(s) in RCA: 239] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Accepted: 04/01/2008] [Indexed: 01/08/2023]
Abstract
Magnetic labelling of living cells creates opportunities for numerous biomedical applications, from individual cell manipulation to MRI tracking. Here we describe a non-specific labelling method based on anionic magnetic nanoparticles (AMNPs). These particles first adsorb electrostatically to the outer membrane before being internalized within endosomes. We compared the labelling mechanism, uptake efficiency and biocompatibility with 14 different cell types, including adult cells, progenitor cells, immune cells and tumour cells. A single model was found to describe cell/nanoparticle interactions and to predict uptake efficiency by all the cell types. The potential impact of the AMNP label on cell functions, in vitro and in vivo, is discussed according to cellular specificities. We also show that the same label provides sufficient magnetization for MRI detection and distal manipulation.
Collapse
Affiliation(s)
- Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS et Université Paris-Diderot, Paris, France.
| | | |
Collapse
|
32
|
Wilhelm C, Lavialle F, Péchoux C, Tatischeff I, Gazeau F. Intracellular trafficking of magnetic nanoparticles to design multifunctional biovesicles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2008; 4:577-582. [PMID: 18383444 DOI: 10.1002/smll.200700523] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Affiliation(s)
- Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes (MSC) Université Denis Diderot-Paris 7 CNRS UMR 7057, Bâtiment Condorcet, CC 7056 10 rue Alice Domon et Léonie Duquet, 75205 Paris cedex, France.
| | | | | | | | | |
Collapse
|
33
|
Wilhelm C, Bal L, Smirnov P, Galy-Fauroux I, Clément O, Gazeau F, Emmerich J. Magnetic control of vascular network formation with magnetically labeled endothelial progenitor cells. Biomaterials 2007; 28:3797-806. [PMID: 17544118 DOI: 10.1016/j.biomaterials.2007.04.047] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Accepted: 04/27/2007] [Indexed: 10/23/2022]
Abstract
We describe the applications of new cellular magnetic labeling method to endothelial progenitor cells (EPC), which have therapeutic potential for revascularization. Via their negative surface charges, anionic magnetic nanoparticles adsorb non-specifically to the EPC plasma membrane, thereby triggering efficient spontaneous endocytosis. The label is non-toxic and does not affect the cells' proliferative capacity. The expression of major membrane proteins involved in neovascularisation is preserved. Labeled cells continue to differentiate in vitro and to form tubular structures in Matrigel (an in vitro model of neovascularization). This process was followed in situ by using high-resolution MRI. Finally, we show that magnetic forces can be used to move magnetically labeled EPC in vitro and to modify their organization in Matrigel both in vitro an in vivo. Magnetic cell targeting opens up new possibilities for vascular tissue engineering and for delivering localized cell-based therapies.
Collapse
Affiliation(s)
- C Wilhelm
- Laboratoire Matière et Systèmes Complexes, CNRS UMR 7057, Université Paris 7, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
34
|
Fortin JP, Gazeau F, Wilhelm C. Intracellular heating of living cells through Néel relaxation of magnetic nanoparticles. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2007; 37:223-8. [PMID: 17641885 DOI: 10.1007/s00249-007-0197-4] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 05/23/2007] [Accepted: 05/28/2007] [Indexed: 10/23/2022]
Abstract
Maghemite and cobalt ferrite anionic magnetic nanoparticles enter tumor cells and can be used as heat sources when exposed to a high-frequency magnetic field. Comparative studies of the two particles enable to unravel the magnetic heating mechanisms (Néel relaxation vs. Brown relaxation) responsible for the cellular temperature rise, and also to establish a simple model, adjusted to the experimental results, allowing to predict the intracellular heating efficiency of iron oxide nanoparticles. Hence, we are able to derive the best nanoparticle design for a given material with a view to intracellular hyperthermia-based applications.
Collapse
|
35
|
Wilhelm C, Rivière C, Biais N. Magnetic control of Dictyostelium aggregation. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2007; 75:041906. [PMID: 17500920 DOI: 10.1103/physreve.75.041906] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 01/22/2007] [Indexed: 05/15/2023]
Abstract
We report the control of cell migration by external magnetic forces during the early stage of Dictysostelium discoideum morphogenesis. Magnetically labeled aggregating cells respond to the presence of a magnetic field created by a thin magnetic tip: forces as low as 30 pN are sufficient to elicit the aggregation of the cells at the extremity of the tip. This induced magnetotaxis is competitive to classical chemotaxis. We therefore underline the interplay between external mechanical forces and morphogenesis. This magnetic assay will open new possibilities in the study of morphogenesis in Dictyostelium.
Collapse
Affiliation(s)
- C Wilhelm
- Laboratoire Matière et Systèmes Complexes, CNRS UMR 7057-Université Paris 7, Paris, France.
| | | | | |
Collapse
|
36
|
Smirnov P, Gazeau F, Beloeil JC, Doan BT, Wilhelm C, Gillet B. Single-cell detection by gradient echo 9.4 T MRI: a parametric study. CONTRAST MEDIA & MOLECULAR IMAGING 2007; 1:165-74. [PMID: 17193693 DOI: 10.1002/cmmi.104] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recent studies have shown that cell migration can be monitored in vivo by magnetic resonance imaging after intracellular contrast agent incorporation. This is due to the dephasing effect on proton magnetization of the local magnetic field created by a labelled cell. Anionic iron oxide nanoparticles (AMNP) are among the most efficient and non-toxic contrast agents to be spontaneously taken up by a wide variety of cells. Here we measured the iron load and magnetization of HeLa tumour cells labelled with AMNP, as a function of the external magnetic field. High-resolution gradient echo 9.4 T MRI detected individual labelled cells, whereas spin echo sequences were poorly sensitive. We then conducted a systematic study in order to determine the gradient echo sequence parameters (echo time, cell magnetization and resolution) most suitable for in vivo identification of single cells.
Collapse
Affiliation(s)
- P Smirnov
- Laboratoire Matière et Systèmes Complexes, Université Paris 7, Denis Diderot and CNRS UMR 7057, 140, rue de Lourmel, 75015 Paris, France.
| | | | | | | | | | | |
Collapse
|
37
|
Fortin JP, Wilhelm C, Servais J, Ménager C, Bacri JC, Gazeau F. Size-sorted anionic iron oxide nanomagnets as colloidal mediators for magnetic hyperthermia. J Am Chem Soc 2007; 129:2628-35. [PMID: 17266310 DOI: 10.1021/ja067457e] [Citation(s) in RCA: 542] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Iron oxide colloidal nanomagnets generate heat when subjected to an alternating magnetic field. Their heating power, governed by the mechanisms of magnetic energy dissipation for single-domain particles (Brown and Néel relaxations), is highly sensitive to the crystal size, the material, and the solvent properties. This study was designed to distinguish between the contributions of Néel and Brownian mechanisms to heat generation. Anionic nanocrystals of maghemite and cobalt ferrite, differing by their magnetic anisotropy, were chemically synthesized and dispersed in an aqueous suspension by electrostatic stabilization. The particles were size-sorted by successive electrostatic phase separation steps. Parameters governing the efficiency of nanomagnets as heat mediators were varied independently; these comprised the particle size (from 5 to 16.5 nm), the solvent viscosity, magnetic anisotropy, and the magnetic field frequency and amplitude. The measured specific loss powers (SLPs) were in quantitative agreement with the results of a predictive model taking into account both Néel and Brown loss processes and the whole particle size distribution. By varying the carrier fluid viscosity, we found that Brownian friction within the carrier fluid was the main contributor to the heating power of cobalt ferrite particles. In contrast, Néel internal rotation of the magnetic moment accounted for most of the loss power of maghemite particles. Specific loss powers were varied by 3 orders of magnitude with increasing maghemite crystal size (from 4 to 1650 W/g at 700 kHz and 24.8 kA/m). This comprehensive parametric study provides the groundwork for the use of anionic colloidal nanocrystals to generate magnetically induced hyperthermia in various media, including complex systems and biological materials.
Collapse
Affiliation(s)
- Jean-Paul Fortin
- Laboratoire Matière et Systèmes Complexes, UMR CNRS 7057, University Paris 7, 140 rue de Lourmel, 75015 Paris, France
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
We discuss herein the theory as well as some design considerations of magnetic tweezers. This method of generating force on magnetic particles bound to biological entities is shown to have a number of advantages over other techniques: forces are exerted in noncontact mode, they can be large in magnitude (order of 10 nanonewtons), and adjustable in direction, static or oscillatory. One apparatus built in our laboratory is described in detail, along with examples of experimental applications and results.
Collapse
Affiliation(s)
- Monica Tanase
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | | | | |
Collapse
|
39
|
Rivière C, Wilhelm C, Cousin F, Dupuis V, Gazeau F, Perzynski R. Internal structure of magnetic endosomes. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2007; 22:1-10. [PMID: 17334684 DOI: 10.1140/epje/e2007-00014-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Indexed: 05/12/2023]
Abstract
The internal structure of biological vesicles filled with magnetic nanoparticles is investigated using the following complementary analyses: electronic transmission microscopy, dynamic probing by magneto-optical birefringence and structural probing by Small Angle Neutron Scattering (SANS). These magnetic vesicles are magnetic endosomes obtained via a non-specific interaction between cells and anionic magnetic iron oxide nanoparticles. Thanks to a magnetic purification process, they are probed at two different stages of their formation within HeLa cells: (i) adsorption of nanoparticles onto the cellular membrane and (ii) their subsequent internalisation within endosomes. Differences in the microenvironment of the magnetic nanoparticles at those two different stages are highlighted here. The dynamics of magnetic nanoparticles adsorbed onto cellular membranes and confined within endosomes is respectively 3 and 5 orders of magnitude slower than for isolated magnetic nanoparticles in aqueous media. Interestingly, SANS experiments show that magnetic endosomes have an internal structure close to decorated vesicles, with magnetic nanoparticles locally decorating the endosome membrane, inside their inner-sphere. These results, important for future biomedical applications, suggest that multiple fusions of decorated vesicles are the biological processes underlying the endocytosis of that kind of nanometric materials.
Collapse
Affiliation(s)
- C Rivière
- Laboratoire des Liquides Ioniques et Interfaces Chargées, UMR CNRS-UPMC-ESPCI 7612, 140 rue de Lourmel, 75015 Paris, France
| | | | | | | | | | | |
Collapse
|
40
|
Modo M, Hoehn M, Bulte JWM. Cellular MR Imaging. Mol Imaging 2005; 4:143-64. [PMID: 16194447 DOI: 10.1162/15353500200505145] [Citation(s) in RCA: 240] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Revised: 04/25/2005] [Accepted: 04/29/2005] [Indexed: 11/04/2022] Open
Abstract
Cellular MR imaging is a young field that aims to visualize targeted cells in living organisms. In order to provide a different signal intensity of the targeted cell, they are either labeled with MR contrast agents in vivo or prelabeled in vitro. Either (ultrasmall) superparamagnetic iron oxide [(U)SPIO] particles or (polymeric) paramagnetic chelates can be used for this purpose. For in vivo cellular labeling, Gd3+- and Mn2+- chelates have mainly been used for targeted hepatobiliary imaging, and (U)SPIO-based cellular imaging has been focused on imaging of macrophage activity. Several of these magneto-pharmaceuticals have been FDA-approved or are in late-phase clinical trials. As for prelabeling of cells in vitro, a challenge has been to induce a sufficient uptake of contrast agents into nonphagocytic cells, without affecting normal cellular function. It appears that this issue has now largely been resolved, leading to an active research on monitoring the cellular biodistribution in vivo following transplantation or transfusion of these cells, including cell migration and trafficking. New applications of cellular MR imaging will be directed, for instance, towards our understanding of hematopoietic (immune) cell trafficking and of novel guided (stem) cell-based therapies aimed to be translated to the clinic in the future.
Collapse
|
41
|
Davila AF, Winklhofer M, Shcherbakov VP, Petersen N. Magnetic pulse affects a putative magnetoreceptor mechanism. Biophys J 2005; 89:56-63. [PMID: 15863473 PMCID: PMC1366555 DOI: 10.1529/biophysj.104.049346] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Clusters of superparamagnetic (SP) magnetite crystals have recently been identified in free nerve endings in the upper-beak skin of homing pigeons and are interpreted as being part of a putative magnetoreceptor system. Motivated by these findings, we developed a physical model that accurately predicts the dynamics of interacting SP clusters in a magnetic field. The main predictions are: 1), under a magnetic field, a group of SP clusters self-assembles into a chain-like structure that behaves like a compass needle under slowly rotating fields; 2), in a frequently changing field as encountered by a moving bird, a stacked chain is a structurally more stable configuration than a single chain; 3), chain-like structures of SP clusters disrupt under strong fields applied at oblique angles; and 4), reassemble on a timescale of hours to days (assuming a viscosity of the cell plasma eta approximately 1 P). Our results offer a novel mechanism for magnetic field perception and are in agreement with the response of birds observed after magnetic-pulse treatments, which have been conducted in the past to specifically test if ferrimagnetic material is involved in magnetoreception, but which have defied explanation so far. Our theoretical results are supported by experiments on a technical SP model system using a high-speed camera. We also offer new predictions that can be tested experimentally.
Collapse
Affiliation(s)
- Alfonso F Davila
- Department für Geo- und Umweltwissenschaften, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | | |
Collapse
|
42
|
Bulte JWM, Kraitchman DL. Iron oxide MR contrast agents for molecular and cellular imaging. NMR IN BIOMEDICINE 2004; 17:484-99. [PMID: 15526347 DOI: 10.1002/nbm.924] [Citation(s) in RCA: 1028] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Molecular and cellular MR imaging is a rapidly growing field that aims to visualize targeted macromolecules or cells in living organisms. In order to provide a different signal intensity of the target, gadolinium-based MR contrast agents can be employed although they suffer from an inherent high threshold of detectability. Superparamagnetic iron oxide (SPIO) particles can be detected at micromolar concentrations of iron, and offer sufficient sensitivity for T2(*)-weighted imaging. Over the past two decades, biocompatible particles have been linked to specific ligands for molecular imaging. However, due to their relatively large size and clearance by the reticuloendothelial system (RES), widespread biomedical molecular applications have yet to be implemented and few studies have been reproduced between different laboratories. SPIO-based cellular imaging, on the other hand, has now become an established technique to label and detect the cells of interest. Imaging of macrophage activity was the initial and still is the most significant application, in particular for tumor staging of the liver and lymph nodes, with several products either approved or in clinical trials. The ability to now also label non-phagocytic cells in culture using derivatized particles, followed by transplantation or transfusion in living organisms, has led to an active research interest to monitor the cellular biodistribution in vivo including cell migration and trafficking. While most of these studies to date have been mere of the 'proof-of-principle' type, further exploitation of this technique will be aimed at obtaining a deeper insight into the dynamics of in vivo cell biology, including lymphocyte trafficking, and at monitoring therapies that are based on the use of stem cells and progenitors.
Collapse
Affiliation(s)
- Jeff W M Bulte
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
43
|
Crich SG, Biancone L, Cantaluppi V, Duò D, Esposito G, Russo S, Camussi G, Aime S. Improved route for the visualization of stem cells labeled with a Gd-/Eu-chelate as dual (MRI and fluorescence) agent. Magn Reson Med 2004; 51:938-44. [PMID: 15122675 DOI: 10.1002/mrm.20072] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A simple labeling procedure of stem/progenitor cells based on the use of Gd-HPDO3A and Eu-HPDO3A, respectively, is described. The Gd-chelate acts as T(1)-agent for MRI visualization, whereas the corresponding Eu-chelate acts as reporter in fluorescence microscopy. Owing to their substantial chemical equivalence, the two chelates are equally internalized in EPCs (endothelial progenitor cells), thus allowing their visualization by both techniques. The lanthanide chelates are entrapped in endosomic vesicles and the labeled cells retain biological activity with preservation of viability and pro-angiogenesis capacity. Hyperintense spots in MR have been observed for Gd-labeled EPCs injected under mice kidney capsule or grafted on a subcutaneous Matrigel plug up to 14 days after transplantation.
Collapse
Affiliation(s)
- Simonetta Geninatti Crich
- Department of Chemistry IFM, University of Torino, and Center for Molecular Imaging (CIM), University of Torino, Torino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Bulte JWM, Arbab AS, Douglas T, Frank JA. Preparation of magnetically labeled cells for cell tracking by magnetic resonance imaging. Methods Enzymol 2004; 386:275-99. [PMID: 15120257 DOI: 10.1016/s0076-6879(04)86013-0] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Jeff W M Bulte
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|