1
|
Bulte JWM, Wang C, Shakeri-Zadeh A. In Vivo Cellular Magnetic Imaging: Labeled vs. Unlabeled Cells. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2207626. [PMID: 36589903 PMCID: PMC9798832 DOI: 10.1002/adfm.202207626] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Indexed: 06/17/2023]
Abstract
Superparamagnetic iron oxide (SPIO)-labeling of cells has been applied for magnetic resonance imaging (MRI) cell tracking for over 30 years, having resulted in a dozen or so clinical trials. SPIO nanoparticles are biodegradable and can be broken down into elemental iron, and hence the tolerance of cells to magnetic labeling has been overall high. Over the years, however, single reports have accumulated demonstrating that the proliferation, migration, adhesion and differentiation of magnetically labeled cells may differ from unlabeled cells, with inhibition of chondrocytic differentiation of labeled human mesenchymal stem cells (hMSCs) as a notable example. This historical perspective provides an overview of some of the drawbacks that can be encountered with magnetic labeling. Now that magnetic particle imaging (MPI) cell tracking is emerging as a new in vivo cellular imaging modality, there has been a renaissance in the formulation of SPIO nanoparticles this time optimized for MPI. Lessons learned from the occasional past pitfalls encountered with SPIO-labeling of cells for MRI may expedite possible future clinical translation of (combined) MRI/MPI cell tracking.
Collapse
Affiliation(s)
- Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chao Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ali Shakeri-Zadeh
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
2
|
Multifunctional Nanoparticles Based on Iron Oxide and Gold-198 Designed for Magnetic Hyperthermia and Radionuclide Therapy as a Potential Tool for Combined HER2-Positive Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14081680. [PMID: 36015306 PMCID: PMC9415738 DOI: 10.3390/pharmaceutics14081680] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/28/2022] [Accepted: 08/09/2022] [Indexed: 01/17/2023] Open
Abstract
Iron oxide nanoparticles are commonly used in many medical applications as they can be easily modified, have a high surface-to-volume ratio, and are biocompatible and biodegradable. This study was performed to synthesize nanoparticles designed for multimodal HER2-positive cancer treatment involving radionuclide therapy and magnetic hyperthermia. The magnetic core (Fe3O4) was coated with a gold-198 layer creating so-called core-shell nanoparticles. These were then further modified with a bifunctional PEG linker and monoclonal antibody to achieve the targeted therapy. Monoclonal antibody—trastuzumab was used to target specific breast and nipple HER2-positive cancer cells. The nanoparticles measured by transmission electron microscopy were as small as 9 nm. The bioconjugation of trastuzumab was confirmed by two separate methods: thermogravimetric analysis and iodine-131 labeling. Synthesized nanoparticles showed that they are good heat mediators in an alternating magnetic field and exhibit great specific binding and internalization capabilities towards the SKOV-3 (HER2 positive) cancer cell line. Radioactive nanoparticles also exhibit capabilities regarding spheroid degradation without and with the application of magnetic hyperthermia with a greater impact in the case of the latter. Designed radiobioconjugate shows great promise and has great potential for in vivo studies regarding magnetic hyperthermia and radionuclide combined therapy.
Collapse
|
3
|
Arifin DR, Witwer KW, Bulte JWM. Non-Invasive imaging of extracellular vesicles: Quo vaditis in vivo? J Extracell Vesicles 2022; 11:e12241. [PMID: 35844061 PMCID: PMC9289215 DOI: 10.1002/jev2.12241] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/12/2022] [Accepted: 06/16/2022] [Indexed: 01/11/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bilayer delimited vesicles released by nearly all cell types that serve as mediators of intercellular signalling. Recent evidence has shown that EVs play a key role in many normal as well as pathological cellular processes. EVs can be exploited as disease biomarkers and also as targeted, cell-free therapeutic delivery and signalling vehicles for use in regenerative medicine and other clinical settings. Despite this potential, much remains unknown about the in vivo biodistribution and pharmacokinetic profiles of EVs after administration into living subjects. The ability to non-invasively image exogeneous EVs, especially in larger animals, will allow a better understanding of their in vivo homing and retention patterns, blood and tissue half-life, and excretion pathways, all of which are needed to advance clinical diagnostic and/or therapeutic applications of EVs. We present the current state-of-the-art methods for labeling EVs with various diagnostic contrast agents and tracers and the respective imaging modalities that can be used for their in vivo visualization: magnetic resonance imaging (MRI), X-ray computed tomography (CT) imaging, magnetic particle imaging (MPI), single-photon emission computed tomography (SPECT), positron emission tomography (PET), and optical imaging (fluorescence and bioluminescence imaging). We review here the strengths and weaknesses of each of these EV imaging approaches, with special emphasis on clinical translation.
Collapse
Affiliation(s)
- Dian R. Arifin
- Russell H. Morgan Department of Radiology and Radiological ScienceDivision of MR Researchthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Neurologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jeff W. M. Bulte
- Russell H. Morgan Department of Radiology and Radiological ScienceDivision of MR Researchthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Oncologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical & Biomolecular Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
4
|
Żuk M, Gawęda W, Majkowska-Pilip A, Osial M, Wolski M, Bilewicz A, Krysiński P. Hybrid Radiobioconjugated Superparamagnetic Iron Oxide-Based Nanoparticles for Multimodal Cancer Therapy. Pharmaceutics 2021; 13:1843. [PMID: 34834258 PMCID: PMC8619896 DOI: 10.3390/pharmaceutics13111843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 11/21/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) are widely used for biomedical applications for their outstanding properties such as facile functionalization and doping with different metals, high surface-to-volume ratio, superparamagnetism, and biocompatibility. This study was designed to synthesize and investigate multifunctional nanoparticle conjugate to act as both a magnetic agent, anticancer immunological drug, and radiopharmaceutic for anticancer therapy. The carrier, 166Ho doped iron oxide, was coated with an Au layer, creating core-shell nanoparticles ([166Ho] Fe3O4@Au. These nanoparticles were subsequently modified with monoclonal antibody trastuzumab (Tmab) to target HER2+ receptors. We describe the radiobioconjugate preparation involving doping of a radioactive agent and attachment of the organic linker and drug to the SPIONs' surface. The size of the SPIONs coated with an Au shell measured by transmission electron microscopy was about 15 nm. The bioconjugation of trastuzumab onto SPIONs was confirmed by thermogravimetric analysis, and the amount of two molecules per one nanoparticle was estimated with the use of radioiodinated [131I]Tmab. The synthesized bioconjugates showed that they are efficient heat mediators and also exhibit a cytotoxic effect toward SKOV-3 ovarian cancer cells expressing HER2 receptors. Prepared radiobioconjugates reveal the high potential for in vivo application of the proposed multimodal hybrid system, combined with magnetic hyperthermia and immunotherapy against cancer tissues.
Collapse
Affiliation(s)
- Michał Żuk
- Faculty of Chemistry, University of Warsaw, Pasteura 1 Str., 02-093 Warsaw, Poland; (M.Ż.); (M.O.)
| | - Weronika Gawęda
- Institute of Nuclear Chemistry and Technology, Dorodna 16 Str., 03-195 Warsaw, Poland; (W.G.); (A.M.-P.)
| | - Agnieszka Majkowska-Pilip
- Institute of Nuclear Chemistry and Technology, Dorodna 16 Str., 03-195 Warsaw, Poland; (W.G.); (A.M.-P.)
| | - Magdalena Osial
- Faculty of Chemistry, University of Warsaw, Pasteura 1 Str., 02-093 Warsaw, Poland; (M.Ż.); (M.O.)
| | - Marcin Wolski
- Centre for Radiotherapy Amethyst, Lubańska 11-12, 59-900 Zgorzelec, Poland;
| | - Aleksander Bilewicz
- Institute of Nuclear Chemistry and Technology, Dorodna 16 Str., 03-195 Warsaw, Poland; (W.G.); (A.M.-P.)
| | - Paweł Krysiński
- Faculty of Chemistry, University of Warsaw, Pasteura 1 Str., 02-093 Warsaw, Poland; (M.Ż.); (M.O.)
| |
Collapse
|
5
|
Glover JC, Aswendt M, Boulland JL, Lojk J, Stamenković S, Andjus P, Fiori F, Hoehn M, Mitrecic D, Pavlin M, Cavalli S, Frati C, Quaini F. In vivo Cell Tracking Using Non-invasive Imaging of Iron Oxide-Based Particles with Particular Relevance for Stem Cell-Based Treatments of Neurological and Cardiac Disease. Mol Imaging Biol 2021; 22:1469-1488. [PMID: 31802361 DOI: 10.1007/s11307-019-01440-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell-based therapeutics is a rapidly developing field associated with a number of clinical challenges. One such challenge lies in the implementation of methods to track stem cells and stem cell-derived cells in experimental animal models and in the living patient. Here, we provide an overview of cell tracking in the context of cardiac and neurological disease, focusing on the use of iron oxide-based particles (IOPs) visualized in vivo using magnetic resonance imaging (MRI). We discuss the types of IOPs available for such tracking, their advantages and limitations, approaches for labeling cells with IOPs, biological interactions and effects of IOPs at the molecular and cellular levels, and MRI-based and associated approaches for in vivo and histological visualization. We conclude with reviews of the literature on IOP-based cell tracking in cardiac and neurological disease, covering both preclinical and clinical studies.
Collapse
Affiliation(s)
- Joel C Glover
- Laboratory for Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, Norway. .,Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway.
| | - Markus Aswendt
- Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich, Leo-Brandt-Str. 5, 52425, Jülich, Germany
| | - Jean-Luc Boulland
- Laboratory for Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway
| | - Jasna Lojk
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, Ljubljana, Slovenia
| | - Stefan Stamenković
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, PB 52, 10001 Belgrade, Serbia
| | - Pavle Andjus
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, PB 52, 10001 Belgrade, Serbia
| | - Fabrizio Fiori
- Department of Applied Physics, Università Politecnica delle Marche - Di.S.C.O., Via Brecce Bianche, 60131, Ancona, Italy
| | - Mathias Hoehn
- Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich, Leo-Brandt-Str. 5, 52425, Jülich, Germany
| | - Dinko Mitrecic
- Laboratory for Stem Cells, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mojca Pavlin
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, Ljubljana, Slovenia.,Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Stefano Cavalli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | |
Collapse
|
6
|
Dual responsive cellulose microspheres with high solid-state fluorescence emission. Colloids Surf A Physicochem Eng Asp 2020. [DOI: 10.1016/j.colsurfa.2020.124510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
7
|
Assessing the interactions between radiotherapy and antitumour immunity. Nat Rev Clin Oncol 2019; 16:729-745. [PMID: 31243334 DOI: 10.1038/s41571-019-0238-9] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2019] [Indexed: 12/17/2022]
Abstract
Immunotherapy, specifically the introduction of immune checkpoint inhibitors, has transformed the treatment of cancer, enabling long-term tumour control even in individuals with advanced-stage disease. Unfortunately, only a small subset of patients show a response to currently available immunotherapies. Despite a growing consensus that combining immune checkpoint inhibitors with radiotherapy can increase response rates, this approach might be limited by the development of persistent radiation-induced immunosuppression. The ultimate goal of combining immunotherapy with radiotherapy is to induce a shift from an ineffective, pre-existing immune response to a long-lasting, therapy-induced immune response at all sites of disease. To achieve this goal and enable the adaptation and monitoring of individualized treatment approaches, assessment of the dynamic changes in the immune system at the patient level is essential. In this Review, we summarize the available clinical data, including forthcoming methods to assess the immune response to radiotherapy at the patient level, ranging from serum biomarkers to imaging techniques that enable investigation of immune cell dynamics in patients. Furthermore, we discuss modelling approaches that have been developed to predict the interaction of immunotherapy with radiotherapy, and highlight how they could be combined with biomarkers of antitumour immunity to optimize radiotherapy regimens and maximize their synergy with immunotherapy.
Collapse
|
8
|
Stroh A, Kressel J, Coras R, Dreyer AY, Fröhlich W, Förschler A, Lobsien D, Blümcke I, Zoubaa S, Schlegel J, Zimmer C, Boltze J. A Safe and Effective Magnetic Labeling Protocol for MRI-Based Tracking of Human Adult Neural Stem Cells. Front Neurosci 2019; 13:1092. [PMID: 31680827 PMCID: PMC6797601 DOI: 10.3389/fnins.2019.01092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/27/2019] [Indexed: 01/09/2023] Open
Abstract
Magnetic resonance imaging (MRI) provides a unique tool for in vivo visualization and tracking of stem cells in the brain. This is of particular importance when assessing safety of experimental cell treatments in the preclinical or clinical setup. Yet, specific imaging requires an efficient and non-perturbing cellular magnetic labeling which precludes adverse effects of the tag, e.g., the impact of iron-oxide-nanoparticles on the critical differentiation and integration processes of the respective stem cell population investigated. In this study we investigated the effects of very small superparamagnetic iron oxide particle (VSOP) labeling on viability, stemness, and neuronal differentiation potential of primary human adult neural stem cells (haNSCs). Cytoplasmic VSOP incorporation massively reduced the transverse relaxation time T2, an important parameter determining MR contrast. Cells retained cytoplasmic label for at least a month, indicating stable incorporation, a necessity for long-term imaging. Using a clinical 3T MRI, 1 × 103 haNSCs were visualized upon injection in a gel phantom, but detection limit was much lower (5 × 104 cells) in layer phantoms and using an imaging protocol feasible in a clinical scenario. Transcriptional analysis and fluorescence immunocytochemistry did not reveal a detrimental impact of VSOP labeling on important parameters of cellular physiology with cellular viability, stemness and neuronal differentiation potential remaining unaffected. This represents a pivotal prerequisite with respect to clinical application of this method.
Collapse
Affiliation(s)
- Albrecht Stroh
- Institute for Pathophysiology, Mainz University, Mainz, Germany.,German Resilience Center, Mainz, Germany
| | - Jenny Kressel
- Department of Neuroradiology, Technical University Munich, Munich, Germany.,Helmholtz Center Munich, Institute for Biological and Medical Imaging, Munich, Germany
| | - Roland Coras
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Antje Y Dreyer
- Translational Center for Regenerative Medicine, Fraunhofer Institute for Cell Therapy and Immunology, University of Leipzig, Leipzig, Germany
| | - Wenke Fröhlich
- Translational Center for Regenerative Medicine, Fraunhofer Institute for Cell Therapy and Immunology, University of Leipzig, Leipzig, Germany
| | - Annette Förschler
- Department of Neuroradiology, Technical University Munich, Munich, Germany
| | - Donald Lobsien
- Department of Neuroradiology, University Hospital Leipzig, Leipzig, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Saida Zoubaa
- Division of Neuropathology, Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Jürgen Schlegel
- Division of Neuropathology, Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Technical University Munich, Munich, Germany
| | - Johannes Boltze
- Translational Center for Regenerative Medicine, Fraunhofer Institute for Cell Therapy and Immunology, University of Leipzig, Leipzig, Germany.,School of Life Sciences, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
9
|
Batista Napotnik T, Miklavčič D. In vitro electroporation detection methods – An overview. Bioelectrochemistry 2018; 120:166-182. [DOI: 10.1016/j.bioelechem.2017.12.005] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022]
|
10
|
Advances in Monitoring Cell-Based Therapies with Magnetic Resonance Imaging: Future Perspectives. Int J Mol Sci 2017; 18:ijms18010198. [PMID: 28106829 PMCID: PMC5297829 DOI: 10.3390/ijms18010198] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 01/07/2023] Open
Abstract
Cell-based therapies are currently being developed for applications in both regenerative medicine and in oncology. Preclinical, translational, and clinical research on cell-based therapies will benefit tremendously from novel imaging approaches that enable the effective monitoring of the delivery, survival, migration, biodistribution, and integration of transplanted cells. Magnetic resonance imaging (MRI) offers several advantages over other imaging modalities for elucidating the fate of transplanted cells both preclinically and clinically. These advantages include the ability to image transplanted cells longitudinally at high spatial resolution without exposure to ionizing radiation, and the possibility to co-register anatomical structures with molecular processes and functional changes. However, since cellular MRI is still in its infancy, it currently faces a number of challenges, which provide avenues for future research and development. In this review, we describe the basic principle of cell-tracking with MRI; explain the different approaches currently used to monitor cell-based therapies; describe currently available MRI contrast generation mechanisms and strategies for monitoring transplanted cells; discuss some of the challenges in tracking transplanted cells; and suggest future research directions.
Collapse
|
11
|
Ocampo SM, Rodriguez V, de la Cueva L, Salas G, Carrascosa JL, Josefa Rodríguez M, García-Romero N, Cuñado JLF, Camarero J, Miranda R, Belda-Iniesta C, Ayuso-Sacido A. g-force induced giant efficiency of nanoparticles internalization into living cells. Sci Rep 2015; 5:15160. [PMID: 26477718 PMCID: PMC4609925 DOI: 10.1038/srep15160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/16/2015] [Indexed: 01/20/2023] Open
Abstract
Nanotechnology plays an increasingly important role in the biomedical arena. Iron oxide nanoparticles (IONPs)-labelled cells is one of the most promising approaches for a fast and reliable evaluation of grafted cells in both preclinical studies and clinical trials. Current procedures to label living cells with IONPs are based on direct incubation or physical approaches based on magnetic or electrical fields, which always display very low cellular uptake efficiencies. Here we show that centrifugation-mediated internalization (CMI) promotes a high uptake of IONPs in glioblastoma tumour cells, just in a few minutes, and via clathrin-independent endocytosis pathway. CMI results in controllable cellular uptake efficiencies at least three orders of magnitude larger than current procedures. Similar trends are found in human mesenchymal stem cells, thereby demonstrating the general feasibility of the methodology, which is easily transferable to any laboratory with great potential for the development of improved biomedical applications.
Collapse
Affiliation(s)
- Sandra M Ocampo
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain
| | - Vanessa Rodriguez
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain
| | - Leonor de la Cueva
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain
| | - Gorka Salas
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain
| | - Jose L Carrascosa
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain.,Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | | | - Noemí García-Romero
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain.,Instituto de Medicina Molecular Aplicada (IMMA), School of Medicine, San Pablo-CEU University, Campus de Montepríncipe, Madrid Spain
| | - Jose Luis F Cuñado
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain.,Departamento de Física de la Materia Condensada, Universidad Autónoma de Madrid &Instituto Nicolás Cabrera, Madrid, Spain
| | - Julio Camarero
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain.,Departamento de Física de la Materia Condensada, Universidad Autónoma de Madrid &Instituto Nicolás Cabrera, Madrid, Spain
| | - Rodolfo Miranda
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain.,Departamento de Física de la Materia Condensada, Universidad Autónoma de Madrid &Instituto Nicolás Cabrera, Madrid, Spain
| | | | - Angel Ayuso-Sacido
- Instituto Madrileño de Estudios Avanzados, IMDEA Nanociencia. Madrid, Spain.,Instituto de Medicina Molecular Aplicada (IMMA), School of Medicine, San Pablo-CEU University, Campus de Montepríncipe, Madrid Spain.,Fundación de Investigación HM Hospitales, Madrid, Spain
| |
Collapse
|
12
|
Granot D, Nkansah MK, Bennewitz MF, Tang KS, Markakis EA, Shapiro EM. Clinically viable magnetic poly(lactide-co-glycolide) particles for MRI-based cell tracking. Magn Reson Med 2015; 71:1238-50. [PMID: 23568825 DOI: 10.1002/mrm.24741] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To design, fabricate, characterize, and in vivo assay clinically viable magnetic particles for MRI-based cell tracking. METHODS Poly(lactide-co-glycolide) (PLGA) encapsulated magnetic nano and microparticles were fabricated. Multiple biologically relevant experiments were performed to assess cell viability, cellular performance, and stem cell differentiation. In vivo MRI experiments were performed to separately test cell transplantation and cell migration paradigms, as well as in vivo biodegradation. RESULTS Highly magnetic nano (∼100 nm) and microparticles (∼1-2 µm) were fabricated. Magnetic cell labeling in culture occurred rapidly achieving 3-50 pg Fe/cell at 3 h for different particles types, and >100 pg Fe/cell after 10 h, without the requirement of a transfection agent, and with no effect on cell viability. The capability of magnetically labeled mesenchymal or neural stem cells to differentiate down multiple lineages, or for magnetically labeled immune cells to release cytokines following stimulation, was uncompromised. An in vivo biodegradation study revealed that NPs degraded ∼80% over the course of 12 weeks. MRI detected as few as 10 magnetically labeled cells, transplanted into the brains of rats. Also, these particles enabled the in vivo monitoring of endogenous neural progenitor cell migration in rat brains over 2 weeks. CONCLUSION The robust MRI properties and benign safety profile of these particles make them promising candidates for clinical translation for MRI-based cell tracking.
Collapse
Affiliation(s)
- Dorit Granot
- Molecular and Cellular MRI Laboratory, Magnetic Resonance Research Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
13
|
Abstract
Stem cell based-therapies are novel therapeutic strategies that hold key for developing new treatments for diseases conditions with very few or no cures. Although there has been an increase in the number of clinical trials involving stem cell-based therapies in the last few years, the long-term risks and benefits of these therapies are still unknown. Detailed in vivo studies are needed to monitor the fate of transplanted cells, including their distribution, differentiation, and longevity over time. Advancements in non-invasive cellular imaging techniques to track engrafted cells in real-time present a powerful tool for determining the efficacy of stem cell-based therapies. In this review, we describe the latest approaches to stem cell labeling and tracking using different imaging modalities.
Collapse
Affiliation(s)
- Amit K Srivastava
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, 217 Traylor Building, 720 Rutland Avenue, Baltimore, MD, 21205-1832, USA
| | | |
Collapse
|
14
|
Lu X, Xia R, Zhang B, Gao F. MRI tracking stem cells transplantation for coronary heart disease. Pak J Med Sci 2014; 30:899-903. [PMID: 25097541 PMCID: PMC4121722 DOI: 10.12669/pjms.304.4936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 02/14/2014] [Accepted: 04/02/2014] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide. Stem cell transplantation has become a new treatment option for cardiovascular disease because the stem cells are able to migrate to damaged cardiac tissue, repair the myocardial infarction area and ultimately reduce the role of the infarct-related mortality. Cardiac magnetic resonance imaging (MRI) is a new robust non-invasive imaging technique that can detect anatomical information and myocardial dysfunction, study the mechanism of stem cells therapy with superb spatial/temporal resolution, relatively safe contrast material and lack of radiation. This review describes the advantages and disadvantages of cardiac MRI applied in stem cells transplantation and discusses how to translate this technique into clinical therapy. Sources of Data/Study Selection: Data from cross-sectional and prospective studies published between the years 2001-2013 on the topic were included. Data searches included both human and animal studies. Data Extraction: The data was extracted from online resources of statistic reports, Pub med, THE MEDLINE, Google Scholar, Medical and Radiological journals. Conclusion: MRI is an appealing technique for cell trafficking depicting engraftment, differentiation and survival.
Collapse
Affiliation(s)
- Xi Lu
- Xi Lu, Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, No.1, Ke Yuan Road 4, Gao Xin District, Chengdu, 610041, Sichuan, China
| | - Rui Xia
- Rui Xia, Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, No.1, Ke Yuan Road 4, Gao Xin District, Chengdu, 610041, Sichuan, China
| | - Bing Zhang
- Bing Zhang, Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, No.1, Ke Yuan Road 4, Gao Xin District, Chengdu, 610041, Sichuan, China
| | - Fabao Gao
- Fabao Gao, Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, No.1, Ke Yuan Road 4, Gao Xin District, Chengdu, 610041, Sichuan, China
| |
Collapse
|
15
|
Hu L, Wickline SA, Hood JL. Magnetic resonance imaging of melanoma exosomes in lymph nodes. Magn Reson Med 2014; 74:266-271. [PMID: 25052384 DOI: 10.1002/mrm.25376] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/03/2014] [Accepted: 06/28/2014] [Indexed: 12/21/2022]
Abstract
PURPOSE Exosomes are cell derived extracellular nanovesicles that relay molecular signals pertinent to both normal physiologic and disease processes. The ability to modify and track exosomes in vivo is essential to understanding exosome pathogenesis, and for utilizing exosomes as effective diagnostic and therapeutic nanocarriers to treat diseases. METHODS We recently reported a new electroporation method that allow exosomes to be loaded with superparamagnetic iron oxide nanoparticles for magnetic resonance tracking. RESULTS Building on this approach, we now demonstrate for the first time using a C57BL/6 mouse model that melanoma exosomes can be imaged in vitro, and within lymph nodes in vivo with the use of standard MRI approaches. CONCLUSION These findings demonstrate proof of principle that exosome biology can be followed in vivo and pave the way for the development of future diagnostic and therapeutic applications. Magn Reson Med 74:266-271, 2015. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lingzhi Hu
- Consortium for Translational Research in Advanced Imaging and Nanomedicine (C-TRAIN), Washington University School of Medicine, St. Louis, Missouri, USA
| | - Samuel A Wickline
- Consortium for Translational Research in Advanced Imaging and Nanomedicine (C-TRAIN), Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joshua L Hood
- Consortium for Translational Research in Advanced Imaging and Nanomedicine (C-TRAIN), Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
16
|
Aryal S, Key J, Stigliano C, Landis MD, Lee DY, Decuzzi P. Positron emitting magnetic nanoconstructs for PET/MR imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:2688-2696. [PMID: 24639392 DOI: 10.1002/smll.201303933] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/10/2014] [Indexed: 06/03/2023]
Abstract
Hybrid PET/MRI scanners have the potential to provide fundamental molecular, cellular, and anatomic information essential for optimizing therapeutic and surgical interventions. However, their full utilization is currently limited by the lack of truly multi-modal contrast agents capable of exploiting the strengths of each modality. Here, we report on the development of long-circulating positron-emitting magnetic nanoconstructs (PEM) designed to image solid tumors for combined PET/MRI. PEMs are synthesized by a modified nano-precipitation method mixing poly(lactic-co-glycolic acid) (PLGA), lipids, and polyethylene glycol (PEG) chains with 5 nm iron oxide nanoparticles (USPIOs). PEM lipids are coupled with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and subsequently chelated to (64)Cu. PEMs show a diameter of 140 ± 7 nm and a transversal relaxivity r2 of 265.0 ± 10.0 (mM × s)(-1), with a r2/r1 ratio of 123. Using a murine xenograft model bearing human breast cancer cell line (MDA-MB-231), intravenously administered PEMs progressively accumulate in tumors reaching a maximum of 3.5 ± 0.25% ID/g tumor at 20 h post-injection. Correlation of PET and MRI signals revealed non-uniform intratumoral distribution of PEMs with focal areas of accumulation at the tumor periphery. These long-circulating PEMs with high transversal relaxivity and tumor accumulation may allow for detailed interrogation over multiple scales in a clinically relevant setting.
Collapse
Affiliation(s)
- Santosh Aryal
- Department of Translational Imaging, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | | | | | | | | | | |
Collapse
|
17
|
Hood JL, Scott MJ, Wickline SA. Maximizing exosome colloidal stability following electroporation. Anal Biochem 2013; 448:41-9. [PMID: 24333249 DOI: 10.1016/j.ab.2013.12.001] [Citation(s) in RCA: 226] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 11/25/2013] [Accepted: 12/02/2013] [Indexed: 12/21/2022]
Abstract
Development of exosome-based semisynthetic nanovesicles for diagnostic and therapeutic purposes requires novel approaches to load exosomes with cargo. Electroporation has previously been used to load exosomes with RNA. However, investigations into exosome colloidal stability following electroporation have not been considered. Herein, we report the development of a unique trehalose pulse media (TPM) that minimizes exosome aggregation following electroporation. Dynamic light scattering (DLS) and RNA absorbance were employed to determine the extent of exosome aggregation and electroextraction post electroporation in TPM compared to common PBS pulse media or sucrose pulse media (SPM). Use of TPM to disaggregate melanoma exosomes post electroporation was dependent on both exosome concentration and electric field strength. TPM maximized exosome dispersal post electroporation for both homogenous B16 melanoma and heterogeneous human serum-derived populations of exosomes. Moreover, TPM enabled heavy cargo loading of melanoma exosomes with 5nm superparamagnetic iron oxide nanoparticles (SPION5) while maintaining original exosome size and minimizing exosome aggregation as evidenced by transmission electron microscopy. Loading exosomes with SPION5 increased exosome density on sucrose gradients. This provides a simple, label-free means of enriching exogenously modified exosomes and introduces the potential for MRI-driven theranostic exosome investigations in vivo.
Collapse
Affiliation(s)
- Joshua L Hood
- Consortium for Translational Research in Advanced Imaging and Nanomedicine (C-TRAIN), Cardiovascular Division, Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Suite 101, Campus Box 8215, St. Louis, MO 63108, USA.
| | - Michael J Scott
- Consortium for Translational Research in Advanced Imaging and Nanomedicine (C-TRAIN), Cardiovascular Division, Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Suite 101, Campus Box 8215, St. Louis, MO 63108, USA.
| | - Samuel A Wickline
- Consortium for Translational Research in Advanced Imaging and Nanomedicine (C-TRAIN), Cardiovascular Division, Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Suite 101, Campus Box 8215, St. Louis, MO 63108, USA.
| |
Collapse
|
18
|
Abstract
The increasing complexity of in vivo imaging technologies, coupled with the development of cell therapies, has fuelled a revolution in immune cell tracking in vivo. Powerful magnetic resonance imaging (MRI) methods are now being developed that use iron oxide- and ¹⁹F-based probes. These MRI technologies can be used for image-guided immune cell delivery and for the visualization of immune cell homing and engraftment, inflammation, cell physiology and gene expression. MRI-based cell tracking is now also being applied to evaluate therapeutics that modulate endogenous immune cell recruitment and to monitor emerging cellular immunotherapies. These recent uses show that MRI has the potential to be developed in many applications to follow the fate of immune cells in vivo.
Collapse
|
19
|
Edmundson M, Thanh NTK, Song B. Nanoparticles based stem cell tracking in regenerative medicine. Theranostics 2013; 3:573-82. [PMID: 23946823 PMCID: PMC3741606 DOI: 10.7150/thno.5477] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 01/07/2013] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapies offer great potentials in the treatment for a wide range of diseases and conditions. With so many stem cell replacement therapies going through clinical trials currently, there is a great need to understand the mechanisms behind a successful therapy, and one of the critical points of discovering them is to track stem cell migration, proliferation and differentiation in vivo. To be of most use tracking methods should ideally be non-invasive, high resolution and allow tracking in three dimensions. Magnetic resonance imaging (MRI) is one of the ideal methods, but requires a suitable contrast agent to be loaded to the cells to be tracked, and one of the most wide-spread in stem cell tracking is a group of agents known as magnetic nanoparticles. This review will explore the current use of magnetic nanoparticles in developing and performing stem cell therapies, and will investigate their potential limitations and the future directions magnetic nanoparticle tracking is heading in.
Collapse
|
20
|
Wang Z, Cuschieri A. Tumour cell labelling by magnetic nanoparticles with determination of intracellular iron content and spatial distribution of the intracellular iron. Int J Mol Sci 2013; 14:9111-25. [PMID: 23624604 PMCID: PMC3676776 DOI: 10.3390/ijms14059111] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 03/26/2013] [Accepted: 04/02/2013] [Indexed: 01/26/2023] Open
Abstract
Magnetically labelled cells are used for in vivo cell tracking by MRI, used for the clinical translation of cell-base therapies. Studies involving magnetic labelled cells may include separation of labelled cells, targeted delivery and controlled release of drugs, contrast enhanced MRI and magnetic hyperthermia for the in situ ablation of tumours. Dextran-coated super-paramagnetic iron oxide (SPIO) ferumoxides are used clinically as an MR contrast agents primarily for hepatic imaging. The material is also widely used for in vitro cell labelling, as are other SPIO-based particles. Our results on the uptake by human cancer cell lines of ferumoxides indicate that electroporation in the presence of protamine sulphate (PS) results in rapid high uptake of SPIO nanoparticles (SPIONs) by parenchymal tumour cells without significant impairment of cell viability. Quantitative determination of cellular iron uptake performed by colorimetric assay is in agreement with data from the literature. These results on intracellular iron content together with the intracellular distribution of SPIONs by magnetic force microscopy (MFM) following in vitro uptake by parenchymal tumour cells confirm the potential of this technique for clinical tumour cell detection and destruction.
Collapse
Affiliation(s)
- Zhigang Wang
- Authors to whom correspondence should be addressed; E-Mails: (Z.W.); (A.C.); Tel.: +44-1382-381-030 (Z.W.); +44-1382-381-009 (A.C.); Fax: +44-1382-386-588 (Z.W. & A.C.)
| | - Alfred Cuschieri
- Authors to whom correspondence should be addressed; E-Mails: (Z.W.); (A.C.); Tel.: +44-1382-381-030 (Z.W.); +44-1382-381-009 (A.C.); Fax: +44-1382-386-588 (Z.W. & A.C.)
| |
Collapse
|
21
|
Manley NC, Steinberg GK. Tracking stem cells for cellular therapy in stroke. Curr Pharm Des 2012; 18:3685-93. [PMID: 22571604 DOI: 10.2174/138161212802002643] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/06/2012] [Indexed: 01/06/2023]
Abstract
Stem cell transplantation has emerged as a promising treatment strategy for stroke. The development of effective ways to monitor transplanted stem cells is essential to understand how stem cell transplantation enhances stroke recovery and ultimately will be an indispensable tool for advancing stem cell therapy to the clinic. In this review, we describe existing methods of tracking transplanted stem cells in vivo, including optical imaging, magnetic resonance imaging (MRI), and positron emission tomography (PET), with emphasis on the benefits and drawbacks of each imaging approach. Key considerations such as the potential impact of each tracking system on stem cell function, as well as its relative applicability to humans are discussed. Finally, we describe multi-modal imaging strategies as a more comprehensive method to track transplanted stem cells in the stroke-injured brain.
Collapse
Affiliation(s)
- Nathan C Manley
- Department of Neurosurgery, Stanford Stroke Center and Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University School of Medicine, 300 Pasteur Drive Stanford, California, CA 94305-5327, USA
| | | |
Collapse
|
22
|
The Use of Nanoparticles as Contrast Media in Neuroimaging: A Statement on Toxicity. World Neurosurg 2012; 78:709-11. [DOI: 10.1016/j.wneu.2011.08.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Accepted: 08/03/2011] [Indexed: 11/19/2022]
|
23
|
Aswendt M, Gianolio E, Pariani G, Napolitano R, Fedeli F, Himmelreich U, Aime S, Hoehn M. In vivo imaging of inhibitory, GABAergic neurons by MRI. Neuroimage 2012; 62:1685-93. [DOI: 10.1016/j.neuroimage.2012.05.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 04/12/2012] [Accepted: 05/12/2012] [Indexed: 10/28/2022] Open
|
24
|
Saulis G, Saulė R. Size of the pores created by an electric pulse: microsecond vs millisecond pulses. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:3032-9. [PMID: 22766475 DOI: 10.1016/j.bbamem.2012.06.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Revised: 06/23/2012] [Accepted: 06/25/2012] [Indexed: 11/30/2022]
Abstract
Here, the sizes of the pores created by square-wave electric pulses with the duration of 100 μs and 2 ms are compared for pulses with the amplitudes close to the threshold of electroporation. Experiments were carried out with three types of cells: mouse hepatoma MH-22A cells, Chinese hamster ovary (CHO) cells, and human erythrocytes. In the case of a short pulse (square-wave with the duration of 100 μs or exponential with the time constant of 22 μs), in the large portion (30-60%) of electroporated (permeable to potassium ions) cells, an electric pulse created only the pores, which were smaller than the molecule of bleomycin (molecular mass of 1450 Da, r≈0.8 nm) or sucrose (molecular mass of 342.3 Da, radius-0.44-0.52 nm). In the case of a long 2-ms duration pulse, in almost all cells, which were electroporated, there were the pores larger than the molecules of bleomycin and/or sucrose. Kinetics of pore resealing depended on the pulse duration and was faster after the shorter pulse. After a short 100-μs duration pulse, the disappearance of the pores permeable to bleomycin was completed after 6-7 min at 24-26°C, while after a long 2-ms duration pulse, this process was slower and lasted 15-20 min. Thus, it can be concluded that a short 100-μs duration pulse created smaller pores than the longer 2-ms duration pulse. This could be attributed to the time inadequacy for pores to grow and expand during the pulse, in the case of short pulses.
Collapse
Affiliation(s)
- Gintautas Saulis
- Laboratory of Biophysics for Bionanotechnology and Medicine, Department of Biology, Faculty of Natural Sciences, Vytautas Magnus University, 58 K. Donelaicio str., Kaunas, LT-44248, Lithuania.
| | | |
Collapse
|
25
|
Wimpenny I, Markides H, El Haj AJ. Orthopaedic applications of nanoparticle-based stem cell therapies. Stem Cell Res Ther 2012; 3:13. [PMID: 22520594 PMCID: PMC3392773 DOI: 10.1186/scrt104] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Stem cells have tremendous applications in the field of regenerative medicine and tissue engineering. These are pioneering fields that aim to create new treatments for disease that currently have limited therapies or cures. A particularly popular avenue of research has been the regeneration of bone and cartilage to combat various orthopaedic diseases. Magnetic nanoparticles (MNPs) have been applied to aid the development and translation of these therapies from research to the clinic. This review highlights contemporary research for the applications of iron-oxide-based MNPs for the therapeutic implementation of stem cells in orthopaedics. These MNPs comprise of an iron oxide core, coated with a choice of biological polymers that can facilitate the uptake of MNPs by cells through improving endocytic activity. The combined use of these oxides and the biological polymer coatings meet biological requirements, effectively encouraging the use of MNPs in regenerative medicine. The association of MNPs with stem cells can be achieved via the process of endocytosis resulting in the internalisation of these particles or the attachment to cell surface receptors. This allows for the investigation of migratory patterns through various tracking studies, the targeting of particle-labelled cells to desired locations via the application of an external magnetic field and, finally, for activation stem cells to initiate various cellular responses to induce the differentiation. Characterisation of cell localisation and associated tissue regeneration can therefore be enhanced, particularly for in vivo applications. MNPs have been shown to have the potential to stimulate differentiation of stem cells for orthopaedic applications, without limiting proliferation. However, careful consideration of the use of active agents associated with the MNP is suggested, for differentiation towards specific lineages. This review aims to broaden the knowledge of current applications, paving the way to translate the in vitro and in vivo work into further orthopaedic clinical studies.
Collapse
Affiliation(s)
- Ian Wimpenny
- Institute of Science and Technology in Medicine, Keele University, The Guy Hilton Research Centre, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire ST4 7QB, UK
| | | | | |
Collapse
|
26
|
Chen CL, Zhang H, Ye Q, Hsieh WY, Hitchens TK, Shen HH, Liu L, Wu YJ, Foley LM, Wang SJ, Ho C. A new nano-sized iron oxide particle with high sensitivity for cellular magnetic resonance imaging. Mol Imaging Biol 2011; 13:825-39. [PMID: 20862612 PMCID: PMC3087837 DOI: 10.1007/s11307-010-0430-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE In this study, we investigated the labeling efficiency and magnetic resonance imaging (MRI) signal sensitivity of a newly synthesized, nano-sized iron oxide particle (IOP) coated with polyethylene glycol (PEG), designed by Industrial Technology Research Institute (ITRI). PROCEDURES Macrophages, bone-marrow-derived dendritic cells, and mesenchymal stem cells (MSCs) were isolated from rats and labeled by incubating with ITRI-IOP, along with three other iron oxide particles in different sizes and coatings as reference. These labeled cells were characterized with transmission electron microscopy (TEM), light and fluorescence microscopy, phantom MRI, and finally in vivo MRI and ex vivo magnetic resonance microscopy (MRM) of transplanted hearts in rats infused with labeled macrophages. RESULTS The longitudinal (r (1)) and transverse (r (2)) relaxivities of ITRI-IOP are 22.71 and 319.2 s(-1) mM(-1), respectively. TEM and microscopic images indicate the uptake of multiple ITRI-IOP particles per cell for all cell types. ITRI-IOP provides sensitivity comparable or higher than the other three particles shown in phantom MRI. In vivo MRI and ex vivo MRM detect punctate spots of hypointensity in rejecting hearts, most likely caused by the accumulation of macrophages labeled by ITRI-IOP. CONCLUSION ITRI-IOP, the nano-sized iron oxide particle, shows high efficiency in cell labeling, including both phagocytic and non-phagocytic cells. Furthermore, it provides excellent sensitivity in T(2)*-weighted MRI, and thus can serve as a promising contrast agent for in vivo cellular MRI.
Collapse
Affiliation(s)
- Chih-Lung Chen
- Biomedical Engineering Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Haosen Zhang
- Pittsburgh NMR Center for Biomedical Research and Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA
| | - Qing Ye
- Pittsburgh NMR Center for Biomedical Research and Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA
| | - Wen-Yuan Hsieh
- Material and Chemical Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - T. Kevin Hitchens
- Pittsburgh NMR Center for Biomedical Research and Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA
| | - Hsin-Hsin Shen
- Biomedical Engineering Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Li Liu
- Pittsburgh NMR Center for Biomedical Research and Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA
| | - Yi-Jen Wu
- Pittsburgh NMR Center for Biomedical Research and Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA
| | - Lesley M. Foley
- Pittsburgh NMR Center for Biomedical Research and Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA
| | - Shian-Jy Wang
- Material and Chemical Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chien Ho
- Pittsburgh NMR Center for Biomedical Research and Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA 15213, USA
| |
Collapse
|
27
|
Guenoun J, Koning GA, Doeswijk G, Bosman L, Wielopolski PA, Krestin GP, Bernsen MR. Cationic Gd-DTPA liposomes for highly efficient labeling of mesenchymal stem cells and cell tracking with MRI. Cell Transplant 2011; 21:191-205. [PMID: 21929868 DOI: 10.3727/096368911x593118] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In the current study cell labeling was performed with water-soluble gadolinium (Gd)-DTPA containing liposomes, to allow for cell tracking by MRI. Liposomes were used to assure a highly concentrated intracellular build up of Gd, aiming to overcome the relatively low MRI sensitivity of Gd (compared to T2 contrast agents). Liposomes were positively charged (cationic) to facilitate uptake by binding to anionic charges in the cell membrane of bone marrow-derived mesenchymal stem cells (MSCs). We determined the cellular Gd load by variations in labeling time (1, 4, and 24 h) and liposome concentration (125, 250, 500, 1000 μM lipid), closely monitoring effects on cell viability, proliferation rate, and differentiation ability. Labeling was both time and dose dependent. Labeling for 4 h was most efficient regarding the combination of processing time and final cellular Gd uptake. Labeling for 4 h with low-dose concentration (125 μM lipid, corresponding to 52 ± 3 μM Gd) yielded an intracellular load of 30 ± 2.5 pg Gd cell(-1), without any effects on cell viability, proliferation, and cell differentiation. Gd liposomes, colabeled with fluorescent dyes, exhibited a prolonged cellular retention, with an endosomal distribution pattern. In vitro assay over 20 days demonstrated a drop in the average Gd load per cell, as a result of mitosis. However, there was no significant change in the sum of the Gd load in all daughter cells at endpoint (20 days), indicating an excellent cellular retention of Gd. MSCs labeled with Gd liposomes were imaged with MRI at both 1.5T and 3.0T, resulting in excellent visualization both in vitro and in vivo. Prolonged in vivo imaging of 500,000 Gd-labeled cells was possible for at least 2 weeks (3.0T). In conclusion, Gd-loaded cationic liposomes (125 μM lipid) are an excellent candidate to label cells, without detrimental effects on cell viability, proliferation, and differentiation, and can be visualized by MRI.
Collapse
Affiliation(s)
- Jamal Guenoun
- Department of Radiology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
28
|
Li XH, Fu YH, Lin QX, Liu ZY, Shan ZX, Deng CY, Zhu JN, Yang M, Lin SG, Li Y, Yu XY. Induced bone marrow mesenchymal stem cells improve cardiac performance of infarcted rat hearts. Mol Biol Rep 2011; 39:1333-42. [PMID: 21667244 DOI: 10.1007/s11033-011-0867-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 05/12/2011] [Indexed: 02/07/2023]
Abstract
We investigated whether transplantation of bone marrow mesenchymal stem cells (BMSC) with induced BMSC (iBMSC) or uninduced BMSC (uBMSC) into the myocardium could improve the performance of post-infarcted rat hearts. BMSCs were specified by flowcytometry. IBMSCs were cocultured with rat cardiomyocyte before transplantation. Cells were injected into borders of cardiac scar tissue 1 week after experimental infarction. Cardiac performance was evaluated by echocardiography at 1, 2, and 4 weeks after cellular or PBS injection. Langendorff working-heart and histological studies were performed 4 weeks after treatment. Myogenesis was detected by quantitative PCR and immunofluorescence. Echocardiography showed a nearly normal ejection fraction (EF) in iBMSC-treated rats and all sham control rats but a lower EF in all PBS-treated animals. The iBMSC-treated heart, assessed by echocardiography, improved fractional shortening compared with PBS-treated hearts. The coronary flow (CF) was decreased obviously in PBS and uBMSC-treated groups, but recovered in iBMSC-treated heart at 4 weeks (P < 0.01). Immunofluorescent microscopy revealed co-localization of Superparamagnetic iron oxide (SPIO)-labeled transplanted cells with cardiac markers for cardiomyocytes, indicating regeneration of damaged myocardium. These data provide strong evidence that iBMSC implantation is of more potential to improve infarcted cardiac performance than uBMSC treatment. It will open new promising therapeutic opportunities for patients with post-infarction heart failure.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Medical Research Center, Guangdong General Hospital, Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Weilun Bldg, Guangzhou 510080, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
High MR sensitive fluorescent magnetite nanocluster for stem cell tracking in ischemic mouse brain. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:1009-19. [PMID: 21530678 DOI: 10.1016/j.nano.2011.03.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 02/17/2011] [Accepted: 03/24/2011] [Indexed: 12/23/2022]
Abstract
UNLABELLED Stem cells have shown a great potential to treat diseases and injuries, including ischemic brain injury. However, developing agents for the long-term tracking of stem cells with few side effects is still challenging. Our aim is to develop a novel fluorescent-magnetite-nanocluster (FMNC) with high MRI sensitivity and to examine its application in the labeling and tracking of mesenchymal stem cells (MSC). For this purpose, we developed FMNC by embedding individual magnetite nanoparticles (NPs) into a polystyrene scaffold coated with two layers of silica and a sandwiched layer of rhodamine. We examined the efficacy of FMNC in MSC labeling and the feasibility of tracking FMNC-labeled MSCs in the ischemic mouse brain. We found that FMNC has high cell-labeling efficiency with no adverse effects on MSCs. In a mouse middle cerebral artery occlusion model, FMNC-labeled MSCs migrated to and accumulated in the ischemic region after FMNC-labeled MSC transplantation. MRI findings highly correlated to immunohistochemistry results. FROM THE CLINICAL EDITOR In this study, the authors report a novel fluorescent-magnetite-nanocluster with high MRI sensitivity and to labeling and tracking of mesenchymal stem cells, and provide in vivo data utilizing a murine stroke model.
Collapse
|
30
|
Cromer Berman SM, Walczak P, Bulte JWM. Tracking stem cells using magnetic nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2011; 3:343-55. [PMID: 21472999 DOI: 10.1002/wnan.140] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Stem cell therapies offer great promise for many diseases, especially those without current effective treatments. It is believed that noninvasive imaging techniques, which offer the ability to track the status of cells after transplantation, will expedite progress in this field and help to achieve maximized therapeutic effect. Today's biomedical imaging technology allows for real-time, noninvasive monitoring of grafted stem cells including their biodistribution, migration, survival, and differentiation, with magnetic resonance imaging (MRI) of nanoparticle-labeled cells being one of the most commonly used techniques. Among the advantages of MR cell tracking are its high spatial resolution, no exposure to ionizing radiation, and clinical applicability. In order to track cells by MRI, the cells need to be labeled with magnetic nanoparticles, for which many types exist. There are several cellular labeling techniques available, including simple incubation, use of transfection agents, magnetoelectroporation, and magnetosonoporation. In this overview article, we will review the use of different magnetic nanoparticles and discuss how these particles can be used to track the distribution of transplanted cells in different organ systems. Caveats and limitations inherent to the tracking of nanoparticle-labeled stem cells are also discussed.
Collapse
Affiliation(s)
- Stacey M Cromer Berman
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
31
|
Abstract
Recently, several protocols for labeling of stem cells with superparamagnetic iron oxides (SPIOs) have been developed, leading to an active and growing field aimed at visualizing stem cells using MRI (magnetic resonance imaging), including image-guided stem cell injections. This development occurred simultaneously with a significant rise in the number of cell therapy clinical trials for cardiovascular applications and their preceding pre-clinical studies in animal models. In this chapter, we will describe several labeling strategies that can be used to label cells with SPIO nanoparticles. This is followed by a discussion of current strategies for using MRI to visualize these cells in myocardial infarct.
Collapse
Affiliation(s)
- Dara L Kraitchman
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | | |
Collapse
|
32
|
Engberink RDO, van der Pol SM, Walczak P, van der Toorn A, Viergever MA, Dijkstra CD, Bulte JW, de Vries HE, Blezer EL. Magnetic Resonance Imaging of Monocytes Labeled with Ultrasmall Superparamagnetic Particles of Iron Oxide Using Magnetoelectroporation in an Animal Model of Multiple Sclerosis. Mol Imaging 2010. [DOI: 10.2310/7290.2010.00016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Raoul D. Oude Engberink
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| | - Susanne M.A. van der Pol
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| | - Pjotr Walczak
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| | - Annette van der Toorn
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| | - Max A. Viergever
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| | - Christine D. Dijkstra
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| | - Jeff W.M. Bulte
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| | - Helga E. de Vries
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| | - Erwin L.A. Blezer
- From the Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands; and Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research; Department of Chemical and Biomolecular Engineering; Department of Biomedical Engineering; and Cellular Imaging Section/Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University
| |
Collapse
|
33
|
Tang Y, Zhang Q, Wang L, Pan PW, Bai G. Preparation of cellulose magnetic microspheres with "the smallest critical size" and their application for microbial immunocapture. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2010; 26:11266-11271. [PMID: 20499913 DOI: 10.1021/la100582t] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The goal of this paper is to introduce a universal method for quantitative control of the particle size of magnetic cellulose microspheres (MCMS) and to produce an optimal antibody absorption capability as an aid in the research of new applications of MCMS in immunomagnetic capture. In this study, "the smallest critical size theory" (TSCS) was proposed, tested, and confirmed by IgG-carrying capability measurements, magnetic response analysis, immunomagnetic capture, and PCR identification of bacteria. A Gaussian expression was proposed and used to guide the preparation of MCMS of the smallest critical size (SCS). The results showed that the diameter of the SCS of MCMS in this study was 5.82 mum, while the IgG absorption capability of the MCMS with SCS was 186.8 mg/mL. In addition, its high sensitivity and the efficiency of immunomagnetic capture of Salmonella bacteria exhibited another new application for MCMS.
Collapse
Affiliation(s)
- Yu Tang
- College of Pharmacy, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | | | | | | | | |
Collapse
|
34
|
Kotková Z, Kotek J, Jirák D, Jendelová P, Herynek V, Berková Z, Hermann P, Lukeš I. Cyclodextrin-Based Bimodal Fluorescence/MRI Contrast Agents: An Efficient Approach to Cellular Imaging. Chemistry 2010; 16:10094-102. [DOI: 10.1002/chem.200903519] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
35
|
Glaus C, Rossin R, Welch MJ, Bao G. In vivo evaluation of (64)Cu-labeled magnetic nanoparticles as a dual-modality PET/MR imaging agent. Bioconjug Chem 2010; 21:715-22. [PMID: 20353170 PMCID: PMC2865436 DOI: 10.1021/bc900511j] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A novel nanoparticle-based dual-modality positron emission tomograph/magnetic resonance imaging (PET/MRI) contrast agent was developed. The probe consisted of a superparamagnetic iron oxide (SPIO) core coated with PEGylated phospholipids. The chelator 1,4,7,10-tetraazacyclo-dodecane-1,4,7,10-tetraacetic acid (DOTA) was conjugated to PEG termini to allow labeling with positron-emitting (64)Cu. Radiolabeling with (64)Cu at high yield and high purity was readily achieved. The (64)Cu-SPIO probes produced strong MR and PET signals and were stable in mouse serum for 24 h at 37 degrees C. Biodistribution and in vivo PET/CT imaging studies of the probes showed a circulation half-life of 143 min and high initial blood retention with moderate liver uptake, making them an attractive contrast agent for disease studies.
Collapse
Affiliation(s)
- Charles Glaus
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Raffaella Rossin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael J. Welch
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gang Bao
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| |
Collapse
|
36
|
Gera A, Steinberg GK, Guzman R. In vivo neural stem cell imaging: current modalities and future directions. Regen Med 2010; 5:73-86. [PMID: 20017696 DOI: 10.2217/rme.09.79] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells have been proposed as a promising therapy for treating a wide variety of neuropathologies. While several studies have demonstrated the therapeutic benefits of neural stem cells, the exact mechanism remains elusive. In order to facilitate research efforts to understand these mechanisms, and before neural stem cell-based therapies can be utilized in a clinical context, we must develop means of monitoring these cells in vivo. However, because of tissue depth and the blood-brain barrier, in vivo imaging of neural stem cells in the brain has unique challenges that do not apply to stem cells for other purposes. In this paper, we review contemporary methods for in vivo neural stem cell imaging, including MRI, PET and optical imaging techniques.
Collapse
Affiliation(s)
- Atul Gera
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, 300 Pasteur Drive, R2111, Stanford, CA 94305-95327, USA
| | | | | |
Collapse
|
37
|
Schäfer R. Labeling and Imaging of Stem Cells - Promises and Concerns. ACTA ACUST UNITED AC 2010; 37:85-89. [PMID: 20737050 DOI: 10.1159/000287271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Accepted: 02/08/2010] [Indexed: 11/19/2022]
Abstract
For experimental and clinical applications of stem cells cell labeling and tracking is interesting to evaluate cell distribution and homing. Tracking of cells after transplantation can be performed for monitoring the delivery and faith of the graft. Furthermore, imaging techniques are part of the evaluation of the functional effects of cellular therapy in the organism of the host. Therefore, systematic investigations on the development and clinical evaluation of imaging techniques and their possible biological impact on stem cells is an emerging topic of today's applied stem cell research. This article refers to different labeling techniques of stem cells highlighting their promises for clinical use and discussing their possible risks.
Collapse
Affiliation(s)
- Richard Schäfer
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital Tuebingen, Germany
| |
Collapse
|
38
|
Abstract
Adoptive transfer of T cells with restricted tumor specificity provides a promising approach to immunotherapy of cancers. However, the isolation of autologous cytotoxic T cells that recognize tumor-associated antigens is time consuming and fails in many instances. Alternatively, gene modification with tumor antigen-specific T-cell receptors (TCR) or chimeric antigen receptors (CARs) can be used to redirect the specificity of large numbers of immune cells toward the malignant cells. Chimeric antigen receptors are composed of the single-chain variable fragment (scFv) of a tumor-recognizing antibody cloned in frame with human T-cell signaling domains (e.g., CD3zeta, CD28, OX40, 4-1BB), thus combining the specificity of antibodies with the effector functions of cytotoxic T cells. Upon antigen binding, the intracellular signaling domains of the CAR initiate cellular activation mechanisms including cytokine secretion and cytolysis of the antigen-positive target cell.In this chapter, we provide detailed protocols for large-scale ex vivo expansion of T cells and manufacturing of medium-scale batches of CAR-expressing T cells for translational research by mRNA electroporation. An anti-CD19 chimeric receptor for the targeting of leukemias and lymphomas was used as a model system. We are currently scaling up the protocols to adapt them to cGMP production of a large number of redirected T cells for clinical applications.
Collapse
Affiliation(s)
- Hilde Almåsbak
- Department of Immunology, Radiumhospitalet-Rikshospitalet, University Hospital, Oslo, Norway
| | | | | |
Collapse
|
39
|
Kedziorek DA, Kraitchman DL. Superparamagnetic iron oxide labeling of stem cells for MRI tracking and delivery in cardiovascular disease. Methods Mol Biol 2010; 660:171-83. [PMID: 20680819 PMCID: PMC3096997 DOI: 10.1007/978-1-60761-705-1_11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In the mid-1980s, iron oxide nanoparticles were developed as contrast agents for diagnostic imaging. In the last two decades, established methods to label cells with superparamagnetic iron oxides (SPIOs) have been developed to aid in targeted delivery and tracking of stem cell therapies. The surge in cellular therapy clinical trials for cardiovascular applications has seen a similar rise in the number of preclinical animal studies of SPIO-labeled stem cells in an effort to understand the mechanisms of cardiovascular regenerative therapy and stem cell biodistribution. The adoption of a limited number of methods of direct labeling of stem cells with SPIOs is due in large part to the desire to rapidly translate these techniques to clinical trials. In this review, we will outline the most commonly adopted methods for iron oxide labeling of stem cells for cardiovascular applications and describe strategies for magnetic resonance imaging (MRI) of magnetically labeled cells in the heart.
Collapse
Affiliation(s)
- Dorota A Kedziorek
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
40
|
Bernsen MR, Moelker AD, Wielopolski PA, van Tiel ST, Krestin GP. Labelling of mammalian cells for visualisation by MRI. Eur Radiol 2009; 20:255-74. [DOI: 10.1007/s00330-009-1540-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 06/11/2009] [Accepted: 06/23/2009] [Indexed: 12/21/2022]
|
41
|
Henning TD, Wendland MF, Golovko D, Sutton EJ, Sennino B, Malek F, Bauer JS, McDonald DM, Daldrup-Link H. Relaxation effects of ferucarbotran-labeled mesenchymal stem cells at 1.5T and 3T: discrimination of viable from lysed cells. Magn Reson Med 2009; 62:325-32. [PMID: 19353670 PMCID: PMC2931823 DOI: 10.1002/mrm.22011] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Accepted: 02/09/2009] [Indexed: 02/06/2023]
Abstract
Human mesenchymal stem cells (hMSCs) were labeled with Ferucarbotran by simple incubation and cultured for up to 14 d. Iron content was determined by spectrometry and the intracellular localization of the contrast agent uptake was studied by electron and confocal microscopy. At various time points after labeling, ranging from 1 to 14 d, samples with viable or lysed labeled hMSCs, as well as nonlabeled controls, underwent MRI. Spin-echo (SE) and gradient-echo (GE) sequences with multiple TRs and TEs were used at 1.5T and 3T on a clinical scanner. Spectrometry showed an initial iron oxide uptake of 7.08 pg per cell. Microscopy studies revealed lysosomal compartmentalization. Contrast agent effects of hMSCs were persistent for up to 14 d after labeling. A marked difference in the T(2) effect of compartmentalized iron oxides compared to free iron oxides was found on T(2)-weighted sequences, but not on T(2)*-weighted sequences. The observed differences may be explained by the loss of compartmentalization of iron oxide particles, the uniformity of distribution, and the subsequent increase in dephasing of protons on SE images. These results show that viable cells with compartmentalized iron oxides may-in principle-be distinguished from lysed cells or released iron oxides.
Collapse
Affiliation(s)
- Tobias D Henning
- Department of Radiology, University of California, San Francisco (UCSF) Medical Center, UCSF, San Francisco, CA 94143-0628, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Farrell E, Wielopolski P, Pavljasevic P, Kops N, Weinans H, Bernsen MR, van Osch GJVM. Cell labelling with superparamagnetic iron oxide has no effect on chondrocyte behaviour. Osteoarthritis Cartilage 2009; 17:961-7. [PMID: 19147376 DOI: 10.1016/j.joca.2008.11.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 11/28/2008] [Indexed: 02/02/2023]
Abstract
BACKGROUND Tissue engineering and regenerative medicine are two rapidly advancing fields of research offering potential for effective treatment of cartilage lesions. Today, chondrocytes are the cell type of choice for use in cartilage repair approaches such as autologous chondrocyte implantation. To verify the safety and efficacy of such approaches it is necessary to determine the fate of these transplanted cells. One way of doing this is prelabelling cells before implantation and tracking them using imaging techniques. The use of superparamagnetic iron oxide (SPIO) for tracking of cells with magnetic resonance imaging (MRI) is ideal for this purpose. It is non-radioactive, does not require viral transfection and is already approved for clinical use as a contrast agent. OBJECTIVE The purpose of this study was to assess the effect of SPIO labelling on adult human chondrocyte behaviour. METHODS Cells were culture expanded and dedifferentiated for two passages and then labelled with SPIO. Effect on cell proliferation was tested. Furthermore, cells were cultured for 21 days in alginate beads in redifferentiation medium. Following this period, cells were analysed for expression of cartilage-related genes, proteoglycan production and collagen protein expression. RESULTS SPIO labelling did not significantly affect any of these parameters relative to unlabelled controls. We also demonstrated SPIO retention within the cells for the full duration of the experiment. CONCLUSIONS This paper demonstrates for the first time the effects of SPIO labelling on chondrocyte behaviour, illustrating its potential for in vivo tracking of implanted chondrocytes.
Collapse
Affiliation(s)
- E Farrell
- Department of Orthopaedics, Erasmus University Medical Centre, Rotterdam, Netherlands.
| | | | | | | | | | | | | |
Collapse
|
43
|
Wang L, Wang Z, Frank TG, Brown SI, Chudek SA, Cuschieri A. Rapid and efficient cell labeling with a MRI contrast agent by electroporation in the presence of protamine sulfate. Nanomedicine (Lond) 2009; 4:305-15. [PMID: 19331538 DOI: 10.2217/nnm.09.6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIMS To investigate various protocols for magnetic labeling of human cancer cells with ferumoxides with a view to developing an effective and fast technique for potential clinical use in MRI. MATERIALS & METHODS Transfection methods utilizing poly-L-lysine and protamine sulfate (PS), electroporation, and combination of PS with electroporation were evaluated in this in vitro study. RESULTS Although transfection was more effective in terms of uptake rates (95-100%) and intracellular iron concentrations (4.01-7.34 pg/cell), all transfection agents required prolonged incubation. By contrast, electroporation yielded fast labeling but with a lower efficacy (68-75%, 1.63-2.59 pg/cell). The addition of PS to electroporation increased the labeling efficacy (80-91%, 2.84-4.16 pg/cell) and protected cell viability. This combined method also resulted in the best T(2)*-shortening effect in the in vitro cellular MRI. CONCLUSION The combined PS-electroporation method provides a fast and efficient protocol for ferumoxide-based cellular imaging and therapeutic procedure.
Collapse
Affiliation(s)
- Lijun Wang
- Institute for Medical Science & Technology (IMSaT), School of Engineering, Physics & Mathematics, University of Dundee, Dundee, UK
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
BACKGROUND The success of many cell-based therapies is highly dependent on the accurate delivery, dosing and trafficking of the cellular therapeutic. In vivo magnetic resonance (MR) cell tracking provides a means to non-invasively and longitudinally evaluate these parameters for cellular therapy. OBJECTIVE To provide an overview of MR cell tracking and how cellular therapeutics might be improved by utilizing this technology. METHODS We focused on the technologies utilized for stem cell and immunotherapies in preclinical models of disease. RESULTS/CONCLUSION New technologies in MR cell tracking will soon take the field beyond preclinical studies and begin to show benefits in clinical trials of novel experimental cell-based therapies.
Collapse
Affiliation(s)
| | - Jeff WM Bulte
- The Johns Hopkins University, School of Medicine, Baltimore, USA
| |
Collapse
|
45
|
Abstract
Recent advances in stem cell biology have raised expectations that both diseases of, and injuries to, the central nervous system may be ameliorated by cell transplantation. In particular, cell therapy has been studied for inducing efficient remyelination in disorders of myelin, including both the largely pediatric disorders of myelin formation and maintenance and the acquired demyelinations of both children and adults. Potential cell-based treatments of two major groups of disorders include both delivery of myelinogenic replacements and mobilization of residual oligodendrocyte progenitor cells as a means of stimulating endogenous repair; the choice of modality is then predicated upon the disease target. In this review we consider the potential application of cell-based therapeutic strategies to disorders of myelin, highlighting the promises as well as the problems and potential perils of this treatment approach.
Collapse
Affiliation(s)
- Tamir Ben-Hur
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Hospital, Jerusalem, Israel.
| | | |
Collapse
|
46
|
Gilad AA, Walczak P, McMahon MT, Na HB, Lee JH, An K, Hyeon T, van Zijl PC, Bulte JW. MR tracking of transplanted cells with "positive contrast" using manganese oxide nanoparticles. Magn Reson Med 2008; 60:1-7. [PMID: 18581402 PMCID: PMC2575033 DOI: 10.1002/mrm.21622] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 02/14/2008] [Indexed: 11/06/2022]
Abstract
Rat glioma cells were labeled using electroporation with either manganese oxide (MnO) or superparamagnetic iron oxide (SPIO) nanoparticles. The viability and proliferation of SPIO-labeled cells (1.9 mg Fe/ml) or cells electroporated with a low dose of MnO (100 microg Mn/ml) was not significantly different from unlabeled cells; a higher MnO dose (785 microg Mn/ml) was found to be toxic. The cellular ion content was 0.1-0.3 pg Mn/cell and 4.4 pg Fe/cell, respectively, with cellular relaxivities of 2.5-4.8 s(-1) (R(1)) and 45-84 s(-1) (R(2)) for MnO-labeled cells. Labeled cells (SPIO and low-dose MnO) were each transplanted in contralateral brain hemispheres of rats and imaged in vivo at 9.4T. While SPIO-labeled cells produced a strong "negative contrast" due to the increase in R(2), MnO-labeled cells produced "positive contrast" with an increased R(1). Simultaneous imaging of both transplants with opposite contrast offers a method for MR "double labeling" of different cell populations.
Collapse
Affiliation(s)
- Assaf A. Gilad
- Russell H. Morgan Department of Radiology, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Cell Imaging Section The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Piotr Walczak
- Russell H. Morgan Department of Radiology, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Cell Imaging Section The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael T. McMahon
- Russell H. Morgan Department of Radiology, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Hyon Bin Na
- National Creative Research Initiative Center for Oxide Nanocrystalline Materials, Seoul National University, Seoul, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jung Hee Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kwangjin An
- National Creative Research Initiative Center for Oxide Nanocrystalline Materials, Seoul National University, Seoul, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Taegwhan Hyeon
- National Creative Research Initiative Center for Oxide Nanocrystalline Materials, Seoul National University, Seoul, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Peter C.M. van Zijl
- Russell H. Morgan Department of Radiology, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Jeff W.M. Bulte
- Russell H. Morgan Department of Radiology, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Cell Imaging Section The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland, USA
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
47
|
Ramadan SS, Heyn C, MacKenzie LT, Chambers AF, Rutt BK, Foster PJ. Ex-vivo cellular MRI with b-SSFP: quantitative benefits of 3 T over 1.5 T. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2008; 21:251-9. [DOI: 10.1007/s10334-008-0118-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 05/25/2008] [Accepted: 05/26/2008] [Indexed: 10/21/2022]
|
48
|
Elder JB, Hoh DJ, Oh BC, Heller AC, Liu CY, Apuzzo ML. THE FUTURE OF CEREBRAL SURGERY. Neurosurgery 2008; 62:1555-79; discussion 1579-82. [DOI: 10.1227/01.neu.0000333820.33143.0d] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
49
|
|
50
|
Molecular MRI of hematopoietic stem-progenitor cells: in vivo monitoring of gene therapy and atherosclerosis. ACTA ACUST UNITED AC 2008; 5:396-404. [PMID: 18477983 DOI: 10.1038/ncpcardio1217] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Accepted: 02/22/2008] [Indexed: 11/09/2022]
Abstract
A characteristic feature of atherosclerotic cardiovascular disease is the diffuse involvement of arteries across the entire human body and the presence of multiple, simultaneous lesions. The diffuse nature of this disease creates a unique challenge for early diagnosis and effective treatment. We believe that recent progress in the field of molecular MRI has opened new avenues towards solving the problem. A new technology has been developed that uses molecular MRI to monitor the migration and homing of hematopoietic stem-progenitor cells to injured arteries and atherosclerosis. In this Review, we introduce several novel technical developments in the field of molecular MRI of atherosclerosis, including advanced techniques for magnetic labeling of stem-progenitor cells and molecular MRI of hematopoietic bone marrow cells migrating to injured arteries and homing to atherosclerotic plaques. In addition, we examine molecular MRI of vascular gene therapy mediated by stem-progenitor cells. These new techniques provide the basis for the further development of in vivo MRI techniques to monitor stem-cell-mediated vascular gene therapy for multiple and diffuse atherosclerotic cardiovascular lesions.
Collapse
|