51
|
Silva AKA, Wilhelm C, Kolosnjaj-Tabi J, Luciani N, Gazeau F. Cellular Transfer of Magnetic Nanoparticles Via Cell Microvesicles: Impact on Cell Tracking by Magnetic Resonance Imaging. Pharm Res 2012; 29:1392-403. [DOI: 10.1007/s11095-012-0680-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 01/06/2012] [Indexed: 01/03/2023]
|
52
|
Mahmoudi M, Hofmann H, Rothen-Rutishauser B, Petri-Fink A. Assessing the in vitro and in vivo toxicity of superparamagnetic iron oxide nanoparticles. Chem Rev 2011; 112:2323-38. [PMID: 22216932 DOI: 10.1021/cr2002596] [Citation(s) in RCA: 366] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Morteza Mahmoudi
- Laboratory of Powder Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | | | | | | |
Collapse
|
53
|
The use of cellular magnetic resonance imaging to track the fate of iron-labeled multipotent stromal cells after direct transplantation in a mouse model of spinal cord injury. Mol Imaging Biol 2011; 13:702-11. [PMID: 20686855 DOI: 10.1007/s11307-010-0393-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The objective of this study was to track the fate of iron-labeled, multipotent stromal cells (MSC) after their direct transplantation into mice with spinal cord injuries using magnetic resonance imaging (MRI). PROCEDURES Mice with spinal cord injuries received a direct transplant of (1) live MSC labeled with micron-sized iron oxide particles (MPIO); (2) dead, MPIO-labeled MSC; (3) unlabeled MSC; or (4) free MPIO and were imaged at 3 T for 6 weeks after transplantation. RESULTS Live, iron-labeled MSC appeared as a well-defined region of signal loss in the mouse spinal cord at the site of transplant. However, the MR appearance of dead, iron-labeled MSC and free iron particles was similar and persisted for the 6 weeks of the study. CONCLUSIONS Iron-labeled stem cells can be detected and monitored in vivo after direct transplantation into the injured spinal cord of mice. However, the fate of the iron label is not clear. Our investigation indicates that caution should be taken when interpreting MR images after direct transplantation of iron-labeled cells.
Collapse
|
54
|
Jiang J, Chen Y, Zhu Y, Yao X, Qi J. Efficient in vitro labeling of human prostate cancer cells with superparamagnetic iron oxide nanoparticles. Cancer Biother Radiopharm 2011; 26:461-7. [PMID: 21812654 DOI: 10.1089/cbr.2010.0934] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The purpose of this study was to investigate the feasibility and optimization of protocols using superparamagnetic iron oxide (SPIO) nanoparticles to label human prostate cancer cell lines PC3 in vitro. The PC3 cells were labeled with different concentrations (28-252 μg Fe/mL) of SPIO and increasing incubation time (6-24 hours), in the presence or absence of a transfection agent poly-l-lysine (PLL). The cell labeling efficiency was analyzed by Prussian blue stain method. The cellular viability was evaluated using trypan blue dye exclusion test. The signal intensity change of the labeled cells was assessed with magnetic resonance imaging (MRI). The results demonstrated that the iron oxide uptake by PC3 cells was dependent on dose and time. The PLL significantly increased the iron load of cells (p<0.01). A final concentration of SPIO nanoparticles of 42-126 μg/mL with 12-24 hours incubation times could be sufficient to label PC3 cells for cellular MRI without impairment of cell viability. This technology may allow for further study into the mechanisms underlying prostate cancer progression as well as permit the real-time imaging of the effectiveness of cancer therapies in vivo.
Collapse
Affiliation(s)
- Jun Jiang
- Department of Ultrasound, Xinhua Hospital/Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, China
| | | | | | | | | |
Collapse
|
55
|
Kallur T, Farr TD, Böhm-Sturm P, Kokaia Z, Hoehn M. Spatio-temporal dynamics, differentiation and viability of human neural stem cells after implantation into neonatal rat brain. Eur J Neurosci 2011; 34:382-93. [DOI: 10.1111/j.1460-9568.2011.07759.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
56
|
Zeng G, Wang G, Guan F, Chang K, Jiao H, Gao W, Xi S, Yang B. Human amniotic membrane-derived mesenchymal stem cells labeled with superparamagnetic iron oxide nanoparticles: the effect on neuron-like differentiation in vitro. Mol Cell Biochem 2011; 357:331-41. [PMID: 21625950 DOI: 10.1007/s11010-011-0904-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 05/19/2011] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells (MSCs) have the potential for self-renewal and multipotential differentiation to regenerate damaged tissues or recover functional absence in diseases. Superparamagnetic iron oxide nanoparticles (SPIONs) are used as contrast agents in magnetic resonance imaging (MRI) for labeling cells in vitro and for tracking SPION-labeled cells after transplantation in vivo. Human amniotic membrane-derived mesenchymal stem cells (hAM-dMSCs) have the capacity for neuron-like differentiation that could be used to cure central nervous system (CNS) diseases. The study investigated the impacts of cytotoxicity of SPIONs on neuron-like differentiation of hAM-dMSCs in both single (1×) and multiple (4×) SPIONs-labeled methods. hAM-dMSCs could be efficiently labeled at safe concentrations of SPIONs (≤14 μg/ml) without significantly affecting their viability (>80% after a MTT assay), special surface antigens (CD29, CD44, CD90, CD105 through flow cytometry), and neuron-like differentiation (nestin and neuron-specific enolase through immunocytochemistry and reverse transcription polymerase chain reaction). Compared with multiple (4×) SPION-labeled methods, a single (1×) SPION-labeled method avoided multiple SPION-labeled hAM-dMSCs and minimized the impact of cytotoxicity of SPIONs on neuron-like differentiation of hAM-dMSCs. Under safe concentrations of SPIONs, a single (1×) SPION-labeled method provided appropriate viability for SPIONs-labeled hAM-dMSCs and facilitated the MRI evaluation of hAM-dMSCs after transplantation.
Collapse
Affiliation(s)
- Guangwei Zeng
- Department of Neurosurgery, First Hospital of Zhengzhou University, Zhengzhou, 450052 Henan Province, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Mahmoudi M, Sahraian MA, Shokrgozar MA, Laurent S. Superparamagnetic iron oxide nanoparticles: promises for diagnosis and treatment of multiple sclerosis. ACS Chem Neurosci 2011; 2:118-40. [PMID: 22778862 PMCID: PMC3369738 DOI: 10.1021/cn100100e] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 12/08/2010] [Indexed: 12/15/2022] Open
Abstract
Smart superparamagnetic iron oxide nanoparticles (SPIONs) are the most promising candidate for theragnosis (i.e., diagnosis and treatment) of multiple sclerosis. A deep understanding of the dynamics of the in vivo neuropathology of multiple sclerosis can be achieved by improving the efficiency of various medical techniques (e.g., positron emission tomography and magnetic resonance imaging) using multimodal SPIONs. In this Review, recent advances and challenges in the development of smart SPIONs for theragnostic applications are comprehensively described. In addition, critical outlines of emerging developments are provided from the points of view of both clinicians and nanotechnologists.
Collapse
Affiliation(s)
- Morteza Mahmoudi
- National Cell Bank, Pasteur Institute of Iran, Tehran, 11365-8639, Iran.
| | | | | | | |
Collapse
|
58
|
Moon SH, Kim JS, Park SJ, Lim JJ, Lee HJ, Lee SM, Chung HM. Effect of chromosome instability on the maintenance and differentiation of human embryonic stem cells in vitro and in vivo. Stem Cell Res 2011; 6:50-9. [PMID: 20920899 DOI: 10.1016/j.scr.2010.08.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2010] [Revised: 08/24/2010] [Accepted: 08/26/2010] [Indexed: 11/18/2022] Open
Abstract
The therapeutic potential of human embryonic stem cells (hESCs) has long been appreciated, and the recent FDA approval of hESC derivatives for cell-based therapy encourages the clinical application of hESCs. Here, using CHA3-hESCs with normal and abnormal karyotypes, we report the importance of maintaining normal chromosomes during in vitro culture and the differentiation of hESCs for minimization of posttransplantation complications. We found that undifferentiated CHA3-hESCs with trisomy chromosome 12 undergo abnormal cell division with multiple spindles in comparison to the bipolar cell division of the karyotypically normal CHA3-hESCs. Transplanted karyotypically abnormal CHA3-hESC derivatives formed a tumor-like tissue 6weeks after transplantation in two out of seven mice tested. Our results demonstrate that the preservation of normal chromosomes is indispensable for maintaining the true properties of hESCs in vitro and abolishing adverse effects posttransplantation. Thus, the development of optimized techniques for stabilizing the chromosome state during in vitro hESC culture is a prerequisite for the therapeutic application of hESCs.
Collapse
|
59
|
Mahmoudi M, Hosseinkhani H, Hosseinkhani M, Boutry S, Simchi A, Journeay WS, Subramani K, Laurent S. Magnetic resonance imaging tracking of stem cells in vivo using iron oxide nanoparticles as a tool for the advancement of clinical regenerative medicine. Chem Rev 2010; 111:253-80. [PMID: 21077606 DOI: 10.1021/cr1001832] [Citation(s) in RCA: 271] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
60
|
Tang C, Russell PJ, Martiniello-Wilks R, Rasko JEJ, Khatri A. Concise review: Nanoparticles and cellular carriers-allies in cancer imaging and cellular gene therapy? Stem Cells 2010; 28:1686-702. [PMID: 20629172 PMCID: PMC2996089 DOI: 10.1002/stem.473] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ineffective treatment and poor patient management continue to plague the arena of clinical oncology. The crucial issues include inadequate treatment efficacy due to ineffective targeting of cancer deposits, systemic toxicities, suboptimal cancer detection and disease monitoring. This has led to the quest for clinically relevant, innovative multifaceted solutions such as development of targeted and traceable therapies. Mesenchymal stem cells (MSCs) have the intrinsic ability to "home" to growing tumors and are hypoimmunogenic. Therefore, these can be used as (a) "Trojan Horses" to deliver gene therapy directly into the tumors and (b) carriers of nanoparticles to allow cell tracking and simultaneous cancer detection. The camouflage of MSC carriers can potentially tackle the issues of safety, vector, and/or transgene immunogenicity as well as nanoparticle clearance and toxicity. The versatility of the nanotechnology platform could allow cellular tracking using single or multimodal imaging modalities. Toward that end, noninvasive magnetic resonance imaging (MRI) is fast becoming a clinical favorite, though there is scope for improvement in its accuracy and sensitivity. In that, use of superparamagnetic iron-oxide nanoparticles (SPION) as MRI contrast enhancers may be the best option for tracking therapeutic MSC. The prospects and consequences of synergistic approaches using MSC carriers, gene therapy, and SPION in developing cancer diagnostics and therapeutics are discussed.
Collapse
Affiliation(s)
- Catherine Tang
- Oncology Research Centre, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia
| | | | | | | | | |
Collapse
|
61
|
Fu Y, Kraitchman DL. Stem cell labeling for noninvasive delivery and tracking in cardiovascular regenerative therapy. Expert Rev Cardiovasc Ther 2010; 8:1149-60. [PMID: 20670192 DOI: 10.1586/erc.10.106] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Clinical and basic scientific studies of stem cell-based therapies have shown promising results for cardiovascular diseases. Despite a rapid transition from animal studies to clinical trials, the mechanisms by which stem cells improve heart function are yet to be fully elucidated. To optimize cell therapies in patients will require a noninvasive means to evaluate cell survival, biodistribution and fate in the same subject over time. Cell labeling offers the ability to image distinct cell lineages in vivo and investigate the efficacy of these therapies using standard noninvasive imaging techniques. In this article, we will discuss the most promising cell labeling techniques for translation to clinical cardiovascular applications.
Collapse
Affiliation(s)
- Yingli Fu
- The Johns Hopkins University School of Medicine, Russell H. Morgan Department of Radiology and Radiological Science, 600 N. Wolfe Street, 314 Park Building, Baltimore, MD 21287, USA
| | | |
Collapse
|
62
|
Lunov O, Zablotskii V, Syrovets T, Röcker C, Tron K, Nienhaus GU, Simmet T. Modeling receptor-mediated endocytosis of polymer-functionalized iron oxide nanoparticles by human macrophages. Biomaterials 2010; 32:547-55. [PMID: 20880574 DOI: 10.1016/j.biomaterials.2010.08.111] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 08/31/2010] [Indexed: 01/04/2023]
Abstract
Although systemically applied nanoparticles are quickly taken up by phagocytic cells, mainly macrophages, the interactions between engineered nanoparticles and macrophages are still not well defined. We therefore analyzed the uptake of diagnostically used carboxydextran-coated superparamagnetic iron oxide nanoparticles of 60 nm (SPIO) and 20 nm (USPIO) by human macrophages. By pharmacological and in vitro knockdown approaches, the principal uptake mechanism for both particles was identified as clathrin-mediated, scavenger receptor A-dependent endocytosis. We developed a mathematical model of the uptake process that allows determination of key parameters of endocytosis, including the rate of uptake, the number of nanoparticles per cell in saturation, the mean uptake time, and the correlation between the number of internalized nanoparticles and their extracellular concentration. The calculated parameters correlate well with experimental data obtained by confocal microscopy. Moreover, the model predicts the individual and collective wrapping times of different nanoparticles, describes the relation between cytoskeletal forces, membrane elasticity and the uptake time. We also introduced a new physical parameter 'a' governing the collective uptake process, a reflecting minimal linear spacing between simultaneously acting neighboring endocytotic pits.
Collapse
Affiliation(s)
- Oleg Lunov
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
63
|
Terreno E, Castelli DD, Viale A, Aime S. Challenges for molecular magnetic resonance imaging. Chem Rev 2010; 110:3019-42. [PMID: 20415475 DOI: 10.1021/cr100025t] [Citation(s) in RCA: 567] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Enzo Terreno
- Department of Chemistry IFM and Molecular Imaging Center, University of Torino, Torino, Italy
| | | | | | | |
Collapse
|
64
|
Cohen ME, Muja N, Fainstein N, Bulte JWM, Ben-Hur T. Conserved fate and function of ferumoxides-labeled neural precursor cells in vitro and in vivo. J Neurosci Res 2010; 88:936-44. [PMID: 19885865 DOI: 10.1002/jnr.22277] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Recent progress in cell therapy research for brain diseases has raised the need for non-invasive monitoring of transplanted cells. For therapeutic application in multiple sclerosis, transplanted cells need to be tracked both spatially and temporally, in order to assess their migration and survival in the host tissue. Magnetic resonance imaging (MRI) of superparamagnetic iron oxide-(SPIO)-labeled cells has been widely used for high resolution monitoring of the biodistribution of cells after transplantation into the central nervous system (CNS). Here we labeled mouse glial-committed neural precursor cells (NPCs) with the clinically approved SPIO contrast agent ferumoxides and examined their survival and differentiation in vitro, as well as their functional response to environmental signals present within the inflamed brain of experimental autoimmune encephalomyelitis (EAE) mice in vivo. We show that ferumoxides labeling does not affect NPC survival and pluripotency in vitro. Following intracerebroventricular (ICV) transplantation in EAE mice, ferumoxides-labeled NPCs responded to inflammatory cues in a similar fashion as unlabeled cells. Ferumoxides-labeled NPCs migrated over comparable distances in white matter tracts and differentiated equally into the glial lineages. Furthermore, ferumoxides-labeled NPCs inhibited lymph node cell proliferation in vitro, similarly to non-labeled cells, suggesting a preserved immunomodulatory function. These results demonstrate that ferumoxides-based MRI cell tracking is well suited for non-invasive monitoring of NPC transplantation.
Collapse
Affiliation(s)
- Mikhal E Cohen
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Medical Center, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
65
|
Li SC, Tachiki LML, Luo J, Dethlefs BA, Chen Z, Loudon WG. A biological global positioning system: considerations for tracking stem cell behaviors in the whole body. Stem Cell Rev Rep 2010; 6:317-33. [PMID: 20237964 PMCID: PMC2887536 DOI: 10.1007/s12015-010-9130-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Many recent research studies have proposed stem cell therapy as a treatment for cancer, spinal cord injuries, brain damage, cardiovascular disease, and other conditions. Some of these experimental therapies have been tested in small animals and, in rare cases, in humans. Medical researchers anticipate extensive clinical applications of stem cell therapy in the future. The lack of basic knowledge concerning basic stem cell biology-survival, migration, differentiation, integration in a real time manner when transplanted into damaged CNS remains an absolute bottleneck for attempt to design stem cell therapies for CNS diseases. A major challenge to the development of clinical applied stem cell therapy in medical practice remains the lack of efficient stem cell tracking methods. As a result, the fate of the vast majority of stem cells transplanted in the human central nervous system (CNS), particularly in the detrimental effects, remains unknown. The paucity of knowledge concerning basic stem cell biology--survival, migration, differentiation, integration in real-time when transplanted into damaged CNS remains a bottleneck in the attempt to design stem cell therapies for CNS diseases. Even though excellent histological techniques remain as the gold standard, no good in vivo techniques are currently available to assess the transplanted graft for migration, differentiation, or survival. To address these issues, herein we propose strategies to investigate the lineage fate determination of derived human embryonic stem cells (hESC) transplanted in vivo into the CNS. Here, we describe a comprehensive biological Global Positioning System (bGPS) to track transplanted stem cells. But, first, we review, four currently used standard methods for tracking stem cells in vivo: magnetic resonance imaging (MRI), bioluminescence imaging (BLI), positron emission tomography (PET) imaging and fluorescence imaging (FLI) with quantum dots. We summarize these modalities and propose criteria that can be employed to rank the practical usefulness for specific applications. Based on the results of this review, we argue that additional qualities are still needed to advance these modalities toward clinical applications. We then discuss an ideal procedure for labeling and tracking stem cells in vivo, finally, we present a novel imaging system based on our experiments.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Center for Neuroscience and Stem Cell Research, Children's Hospital of Orange County Research Institute, University of California Irvine, 455 South Main Street, Orange, CA 92868, USA.
| | | | | | | | | | | |
Collapse
|
66
|
Wang J, Zhang S, Rabinovich B, Bidaut L, Soghomonyan S, Alauddin MM, Bankson JA, Shpall E, Willerson JT, Gelovani JG, Yeh ETH. Human CD34+ cells in experimental myocardial infarction: long-term survival, sustained functional improvement, and mechanism of action. Circ Res 2010; 106:1904-11. [PMID: 20448213 DOI: 10.1161/circresaha.110.221762] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
RATIONALE Human CD34(+) cells have been used in clinical trials for treatment of myocardial infarction (MI). However, it is unknown how long the CD34(+) cells persist in hearts, whether the improvement in cardiac function is sustained, or what are the underlying mechanisms. OBJECTIVE We sought to track the fate of injected human CD34(+) cells in the hearts of severe combined immune deficiency (SCID) mice after experimental MI and to determine the mechanisms of action. METHODS AND RESULTS We used multimodality molecular imaging to track the fate of injected human CD34(+) cells in the hearts of SCID mice after experimental MI, and used selective antibody blocking to determine the mechanisms of action. Bioluminescence imaging showed that injected CD34(+) cells survived in the hearts for longer than 12 months. The PET signal from the injected cells was detected in the wall of the left ventricle. Cardiac MRI showed that left ventricular ejection fraction was significantly improved in the treated mice compared to the control mice for up to 52 weeks (P<0.05). Furthermore, treatment with anti-alpha4beta1 showed that generation of human-derived cardiomyocytes was inhibited, whereas anti-vascular endothelial growth factor (VEGF) treatment blocked the production of human-derived endothelial cells. However, the improvement in cardiac function was abolished only in the anti-VEGF, but not anti-alpha4beta1, treated group. CONCLUSIONS Angiogenesis and/or paracrine effect, but not myogenesis, is responsible for functional improvement following CD34(+) cells therapy.
Collapse
Affiliation(s)
- Jingxiong Wang
- University of Texas-MD Anderson Cancer Center, 1400 Pressler Blvd, Box 1451, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Yang Y, Yang Y, Yanasak N, Schumacher A, Hu TCC. Temporal and noninvasive monitoring of inflammatory-cell infiltration to myocardial infarction sites using micrometer-sized iron oxide particles. Magn Reson Med 2010; 63:33-40. [PMID: 19953508 DOI: 10.1002/mrm.22175] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Micrometer-sized iron oxide particles (MPIO) are a more sensitive MRI contrast agent for tracking cell migration compared to ultrasmall iron oxide particles. This study investigated the temporal relationship between inflammation and tissue remodeling due to myocardial infarction (MI) using MPIO-enhanced MRI. C57Bl/6 mice received an intravenous MPIO injection for cell labeling, followed by a surgically induced MI seven days later (n=7). For controls, two groups underwent either sham-operated surgery without inducing an MI post-MPIO injection (n=7) or MI surgery without MPIO injection (n=6). The MRIs performed post-MI showed significant signal attenuation around the MI site for the mice that received an intravenous MPIO injection for cell labeling, followed by a surgically induced MI seven days later, compared to the two control groups (P<0.01). The findings suggested that the prelabeled inflammatory cells mobilized and infiltrated into the MI site. Furthermore, the linear regression of contrast-to-noise ratio at the MI site and left ventricular ejection function suggested a positive correlation between the labeled inflammatory cell infiltration and cardiac function attenuation during post-MI remodeling (r2=0.98). In conclusion, this study demonstrated an MRI technique for noninvasively and temporally monitoring inflammatory cell migration into the myocardium while potentially providing additional insight concerning the pathologic progression of a myocardial infarction.
Collapse
Affiliation(s)
- Yidong Yang
- Small Animal Imaging, Department of Radiology, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | | | |
Collapse
|
68
|
"Microenvironmental contaminations" induced by fluorescent lipophilic dyes used for noninvasive in vitro and in vivo cell tracking. Blood 2010; 115:5347-54. [PMID: 20215639 DOI: 10.1182/blood-2009-05-224030] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Determining how normal and leukemic stem cells behave in vivo, in a dynamic and noninvasive way, remains a major challenge. Most optical tracking technologies rely on the use of fluorescent or bioluminescent reporter genes, which need to be stably expressed in the cells of interest. Because gene transfer in primary leukemia samples represents a major risk to impair their capability to engraft in a xenogenic context, we evaluated the possibility to use gene transfer-free labeling technologies. The lipophilic dye 3,3,3',3' tetramethylindotricarbocyanine iodide (DiR) was selected among 4 near-infrared (NIR) staining technologies. Unfortunately we report here a massive transfer of the dye occurring toward the neighbor cells both in vivo and in vitro. We further demonstrate that all lipophilic dyes tested in this study (1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine perchlorate [DiI], DiD, DiR, and PKH26) can give rise to microenvironmental contamination, including when used in suboptimal concentration, after extensive washing procedures and in the absence of phagocytosis or marked cell death. This was observed from all cell types tested. Eventually, we show that this microenvironmental contamination is mediated by both direct cell-cell contacts and diffusible microparticles. We conclude that tracking of labeled cells using non-genetically encoded markers should always be accompanied by drastic cross validation using multimodality approaches.
Collapse
|
69
|
Magnetic labeling, imaging and manipulation of endothelial progenitor cells using iron oxide nanoparticles. Future Med Chem 2010; 2:397-408. [DOI: 10.4155/fmc.09.165] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Endothelial progenitor cells (EPCs), originating from bone marrow, play a significant role in the repair of ischemic tissue and injured blood vessels. They are also involved in tumor angiogenesis. The therapeutic potential of EPCs for regenerative medicine and cancer treatment calls for new methods for monitoring and controlling cell migration. This review focuses on promising magnetic methods based on the internalization of magnetic nanoparticles by EPCs. We first describe the cellular uptake of iron oxide nanoparticles depending on their surface properties. We thus review the use of MRI for the detection of labeled cells and for noninvasive follow-up of EPCs homing in sites of endothelium regeneration. Finally, we show that remotely applied magnetic forces may enable intracellular manipulation and may optimize cell-delivery strategies for localizing cell therapy to target sites.
Collapse
|
70
|
van Buul GM, Farrell E, Kops N, van Tiel ST, Bos PK, Weinans H, Krestin GP, van Osch GJVM, Bernsen MR. Ferumoxides-protamine sulfate is more effective than ferucarbotran for cell labeling: implications for clinically applicable cell tracking using MRI. CONTRAST MEDIA & MOLECULAR IMAGING 2010; 4:230-6. [PMID: 19839030 DOI: 10.1002/cmmi.289] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The use of superparamagnetic iron oxide (SPIO) for labeling cells holds great promise for clinically applicable cell tracking using magnetic resonance imaging. For clinical application, an effectively and specifically labeled cell preparation is highly desired (i.e. a large amount of intracellular iron and a negligible amount of extracellular iron). In this study we performed a direct comparison of two SPIO labeling strategies that have both been reported as efficient and clinically translatable approaches. These approaches are cell labeling using ferumoxides-protamine complexes or ferucarabotran particles. Cell labeling was performed on primary human bone marrow stromal cells (hBMSCs) and chondrocytes. For both cell types ferumoxides-protamine resulted in a higher percentage of labeled cells, a higher total iron load, a larger amount of intracellular iron and a lower amount of extracellular iron aggregates, compared with ferucarbotran. Consequently, hBMSC and chondrocyte labeling with ferumoxides-protamine is more effective and results in more specific cell labeling than ferucarbotran.
Collapse
Affiliation(s)
- G M van Buul
- Department of Radiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Abstract
Despite promising preclinical data, the treatment of cardiovascular diseases using embryonic, bone-marrow-derived, and skeletal myoblast stem cells has not yet come to fruition within mainstream clinical practice. Major obstacles in cardiac stem cell investigations include the ability to monitor cell engraftment and survival following implantation within the myocardium. Several cellular imaging modalities, including reporter gene and MRI-based tracking approaches, have emerged that provide the means to identify, localize, and monitor stem cells longitudinally in vivo following implantation. This Review will examine the various cardiac cellular tracking modalities, including the combinatorial use of several probes in multimodality imaging, with a focus on data from the past 5 years.
Collapse
|
72
|
Thu MS, Najbauer J, Kendall SE, Harutyunyan I, Sangalang N, Gutova M, Metz MZ, Garcia E, Frank RT, Kim SU, Moats RA, Aboody KS. Iron labeling and pre-clinical MRI visualization of therapeutic human neural stem cells in a murine glioma model. PLoS One 2009; 4:e7218. [PMID: 19787043 PMCID: PMC2746284 DOI: 10.1371/journal.pone.0007218] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Accepted: 08/05/2009] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Treatment strategies for the highly invasive brain tumor, glioblastoma multiforme, require that cells which have invaded into the surrounding brain be specifically targeted. The inherent tumor-tropism of neural stem cells (NSCs) to primary and invasive tumor foci can be exploited to deliver therapeutics to invasive brain tumor cells in humans. Use of the strategy of converting prodrug to drug via therapeutic transgenes delivered by immortalized therapeutic NSC lines have shown efficacy in animal models. Thus therapeutic NSCs are being proposed for use in human brain tumor clinical trials. In the context of NSC-based therapies, MRI can be used both to non-invasively follow dynamic spatio-temporal patterns of the NSC tumor targeting allowing for the optimization of treatment strategies and to assess efficacy of the therapy. Iron-labeling of cells allows their presence to be visualized and tracked by MRI. Thus we aimed to iron-label therapeutic NSCs without affecting their cellular physiology using a method likely to gain United States Federal Drug Administration (FDA) approval. METHODOLOGY For human use, the characteristics of therapeutic Neural Stem Cells must be clearly defined with any pertubation to the cell including iron labeling requiring reanalysis of cellular physiology. Here, we studied the effect of iron-loading of the therapeutic NSCs, with ferumoxide-protamine sulfate complex (FE-Pro) on viability, proliferation, migratory properties and transgene expression, when compared to non-labeled cells. FE-Pro labeled NSCs were imaged by MRI at tumor sites, after intracranial administration into the hemisphere contralateral to the tumor, in an orthotopic human glioma xenograft mouse model. CONCLUSION FE-Pro labeled NSCs retain their proliferative status, tumor tropism, and maintain stem cell character, while allowing in vivo cellular MRI tracking at 7 Tesla, to monitor their real-time migration and distribution at brain tumor sites. Of significance, this work directly supports the use of FE-Pro-labeled NSCs for real-time tracking in the clinical trial under development: "A Pilot Feasibility Study of Oral 5-Fluorocytosine and Genetically modified Neural Stem Cells Expressing Escherichia coli Cytosine Deaminase for Treatment of Recurrent High-Grade Gliomas".
Collapse
Affiliation(s)
- Mya S. Thu
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail: (MYT); (KSA)
| | - Joseph Najbauer
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Stephen E. Kendall
- Division of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Ira Harutyunyan
- Radiology MS 81, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Nicole Sangalang
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Margarita Gutova
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Marianne Z. Metz
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Elizabeth Garcia
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Richard T. Frank
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Seung U. Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, British Columbia, Canada
- Institute for Regenerative Medicine, Gachon University Gil Hospital, Inchon, Korea
| | - Rex A. Moats
- Radiology MS 81, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Karen S. Aboody
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- Division of Neuroscience, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail: (MYT); (KSA)
| |
Collapse
|
73
|
Pawelczyk E, Jordan EK, Balakumaran A, Chaudhry A, Gormley N, Smith M, Lewis BK, Childs R, Robey PG, Frank JA. In vivo transfer of intracellular labels from locally implanted bone marrow stromal cells to resident tissue macrophages. PLoS One 2009; 4:e6712. [PMID: 19696933 PMCID: PMC2726631 DOI: 10.1371/journal.pone.0006712] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 07/07/2009] [Indexed: 02/06/2023] Open
Abstract
Intracellular labels such as dextran coated superparamagnetic iron oxide nanoparticles (SPION), bromodeoxyuridine (BrdU) or green fluorescent protein (GFP) are frequently used to study the fate of transplanted cells by in vivo magnetic resonance imaging or fluorescent microscopy. Bystander uptake of labeled cells by resident tissue macrophages (TM) can confound the interpretation of the presence of intracellular labels especially during direct implantation of cells, which can result in more than 70% cell death. In this study we determined the percentages of TM that took up SPION, BrdU or GFP from labeled bone marrow stromal cells (BMSCs) that were placed into areas of angiogenesis and inflammation in a mouse model known as Matrigel™ plaque perfusion assay. Cells recovered from digested plaques at various time points were analyzed by fluorescence microscopy and flow cytometry. The analysis of harvested plaques revealed 5% of BrdU+, 5–10% of GFP+ and 5–15% of dextran+ macrophages. The transfer of the label was not dependent on cell dose or viability. Collectively, this study suggests that care should be taken to validate donor origin of cells using an independent marker by histology and to assess transplanted cells for TM markers prior to drawing conclusions about the in vivo behavior of transplanted cells.
Collapse
Affiliation(s)
- Edyta Pawelczyk
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Gonzalez-Lara LE, Xu X, Hofstetrova K, Pniak A, Brown A, Foster PJ. In vivo magnetic resonance imaging of spinal cord injury in the mouse. J Neurotrauma 2009; 26:753-62. [PMID: 19397403 DOI: 10.1089/neu.2008.0704] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The feasibility of performing high-resolution in vivo magnetic resonance imaging (MRI) to visualize the injured mouse spinal cord using a three-dimensional (3D)-FIESTA (fast imaging employing steady state acquisition) pulse sequence, in a clip compression injury model, is presented. Images were acquired using a 3-Tesla clinical whole-body MR system equipped with a high-performance gradient coil insert. High-resolution mouse cord images were used to detect and monitor the cord lesions for 6 weeks after spinal cord injury (SCI). The epicenter of the injury appeared as a region of mixed signal intensities on day 2 post-SCI. Regions of signal hypointensity appeared at the lesion site by 2 weeks post-SCI and became more apparent with time. In some mice, large cyst-like lesions were detected rostral to the lesion epicenter, as early as 2 weeks post-SCI, and increased in volume with time. In addition, MRI was used to detect and monitor iron-labeled mesenchymal stem cells (MSCs) after their transplantation into the injured cord. MSCs appeared as large, obvious regions of signal loss in the cord, which decreased in size over time.
Collapse
|
75
|
Janic B, Rad AM, Jordan EK, Iskander ASM, Ali MM, Varma NRS, Frank JA, Arbab AS. Optimization and validation of FePro cell labeling method. PLoS One 2009; 4:e5873. [PMID: 19517015 PMCID: PMC2690694 DOI: 10.1371/journal.pone.0005873] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 05/11/2009] [Indexed: 02/06/2023] Open
Abstract
Current method to magnetically label cells using ferumoxides (Fe)-protamine (Pro) sulfate (FePro) is based on generating FePro complexes in a serum free media that are then incubated overnight with cells for the efficient labeling. However, this labeling technique requires long (>12-16 hours) incubation time and uses relatively high dose of Pro (5-6 microg/ml) that makes large extracellular FePro complexes. These complexes can be difficult to clean with simple cell washes and may create low signal intensity on T2* weighted MRI that is not desirable. The purpose of this study was to revise the current labeling method by using low dose of Pro and adding Fe and Pro directly to the cells before generating any FePro complexes. Human tumor glioma (U251) and human monocytic leukemia cell (THP-1) lines were used as model systems for attached and suspension cell types, respectively and dose dependent (Fe 25 to 100 microg/ml and Pro 0.75 to 3 microg/ml) and time dependent (2 to 48 h) labeling experiments were performed. Labeling efficiency and cell viability of these cells were assessed. Prussian blue staining revealed that more than 95% of cells were labeled. Intracellular iron concentration in U251 cells reached approximately 30-35 pg-iron/cell at 24 h when labeled with 100 microg/ml of Fe and 3 microg/ml of Pro. However, comparable labeling was observed after 4 h across the described FePro concentrations. Similarly, THP-1 cells achieved approximately 10 pg-iron/cell at 48 h when labeled with 100 microg/ml of Fe and 3 microg/ml of Pro. Again, comparable labeling was observed after 4 h for the described FePro concentrations. FePro labeling did not significantly affect cell viability. There was almost no extracellular FePro complexes observed after simple cell washes. To validate and to determine the effectiveness of the revised technique, human T-cells, human hematopoietic stem cells (hHSC), human bone marrow stromal cells (hMSC) and mouse neuronal stem cells (mNSC C17.2) were labeled. Labeling for 4 hours using 100 microg/ml of Fe and 3 microg/ml of Pro resulted in very efficient labeling of these cells, without impairing their viability and functional capability. The new technique with short incubation time using 100 microg/ml of Fe and 3 microg/ml of Pro is effective in labeling cells for cellular MRI.
Collapse
Affiliation(s)
- Branislava Janic
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, MI, USA.
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Monitoring the survival of islet transplants by MRI using a novel technique for their automated detection and quantification. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2009; 22:257-65. [DOI: 10.1007/s10334-009-0172-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 04/03/2009] [Accepted: 04/06/2009] [Indexed: 10/20/2022]
|
77
|
Liu W, Dahnke H, Rahmer J, Jordan EK, Frank JA. Ultrashort T2* relaxometry for quantitation of highly concentrated superparamagnetic iron oxide (SPIO) nanoparticle labeled cells. Magn Reson Med 2009; 61:761-6. [PMID: 19191285 PMCID: PMC3384488 DOI: 10.1002/mrm.21923] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Accepted: 11/14/2008] [Indexed: 11/12/2022]
Abstract
A new method was developed to measure ultrashort T(2)* relaxation in tissues containing a focal area of superparamagnetic iron oxide (SPIO) nanoparticle-labeled cells in which the T(2)* decay is too short to be accurately measured using regular gradient echo T(2)* mapping. The proposed method utilizes the relatively long T(2) relaxation of SPIO-labeled cells and acquires a series of spin echo images with the readout echo shifted to sample the T(2)* decay curve. MRI experiments in phantoms and rats with SPIO-labeled tumors demonstrated that it can detect ultrashort T(2)* down to 1 ms or less. The measured T(2)* values were about 10% higher than those from the ultrashort TE (UTE) technique. The shorter the TE, the less the measurements deviated from the UTE T(2)* mapping. Combined with the regular T(2)* mapping, this technique is expected to provide quantitation of highly concentrated iron-labeled cells from direct cell transplantation.
Collapse
Affiliation(s)
- Wei Liu
- Philips Research, North America, Briarcliff Manor, New York 10510, USA.
| | | | | | | | | |
Collapse
|
78
|
Abstract
PURPOSE OF REVIEW Transplantation of cells is an urgent clinical need that is increasingly providing an alternative to solid-organ transplants. This review discusses the state-of-the-art in-vivo imaging of cell transplantation with a special focus on recent developments. RECENT FINDINGS Noninvasive imaging modalities, such as magnetic resonance imaging (MRI), nuclear (positron emission tomography and single-photon emission computed tomography), acoustical, and optical imaging can investigate the biodistribution, fate, and functional integration of grafted cells. Especially, multimodal imaging is emerging as an important development to provide complimentary and confirmatory information. SUMMARY The development of noninvasive imaging of transplanted cells has progressed rapidly over the last few years. Translating these techniques into clinical protocols remains the focus of ongoing investigations.
Collapse
|
79
|
Marquis BJ, Love SA, Braun KL, Haynes CL. Analytical methods to assess nanoparticle toxicity. Analyst 2009; 134:425-39. [PMID: 19238274 DOI: 10.1039/b818082b] [Citation(s) in RCA: 317] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
During the past 20 years, improvements in nanoscale materials synthesis and characterization have given scientists great control over the fabrication of materials with features between 1 and 100 nm, unlocking many unique size-dependent properties and, thus, promising many new and/or improved technologies. Recent years have found the integration of such materials into commercial goods; a current estimate suggests there are over 800 nanoparticle-containing consumer products (The Project on Emerging Nanotechnologies Consumer Products Inventory, , accessed Oct. 2008), accounting for 147 billion USD in products in 2007 (Nanomaterials state of the market Q3 2008: stealth success, broad impact, Lux Research Inc., New York, NY, 2008). Despite this increase in the prevalence of engineered nanomaterials, there is little known about their potential impacts on environmental health and safety. The field of nanotoxicology has formed in response to this lack of information and resulted in a flurry of research studies. Nanotoxicology relies on many analytical methods for the characterization of nanomaterials as well as their impacts on in vitro and in vivo function. This review provides a critical overview of these techniques from the perspective of an analytical chemist, and is intended to be used as a reference for scientists interested in conducting nanotoxicological research as well as those interested in nanotoxicological assay development.
Collapse
Affiliation(s)
- Bryce J Marquis
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
80
|
Arbab AS, Janic B, Haller J, Pawelczyk E, Liu W, Frank JA. In Vivo Cellular Imaging for Translational Medical Research. Curr Med Imaging 2009; 5:19-38. [PMID: 19768136 DOI: 10.2174/157340509787354697] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Personalized treatment using stem, modified or genetically engineered, cells is becoming a reality in the field of medicine, in which allogenic or autologous cells can be used for treatment and possibly for early diagnosis of diseases. Hematopoietic, stromal and organ specific stem cells are under evaluation for cell-based therapies for cardiac, neurological, autoimmune and other disorders. Cytotoxic or genetically altered T-cells are under clinical trial for the treatment of hematopoietic or other malignant diseases. Before using stem cells in clinical trials, translational research in experimental animal models are essential, with a critical emphasis on developing noninvasive methods for tracking the temporal and spatial homing of these cells to target tissues. Moreover, it is necessary to determine the transplanted cell's engraftment efficiency and functional capability. Various in vivo imaging modalities are in use to track the movement and incorporation of administered cells. Tagging cells with reporter genes, fluorescent dyes or different contrast agents transforms them into cellular probes or imaging agents. Recent reports have shown that magnetically labeled cells can be used as cellular magnetic resonance imaging (MRI) probes, demonstrating the cell trafficking to target tissues. In this review, we will discuss the methods to transform cells into probes for in vivo imaging, along with their advantages and disadvantages as well as the future clinical applicability of cellular imaging method and corresponding imaging modality.
Collapse
Affiliation(s)
- Ali S Arbab
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, MI
| | | | | | | | | | | |
Collapse
|
81
|
Abstract
Transplantation of stem cells or immune cells has shown promise for the treatment of several diseases. Monitoring magnetically labeled cells with MRI has furthered our understanding of cellular migration and the pathophysiology of diseases in experimental models. These studies should pave the way for guiding clinical trials using cell-based therapies. This review briefly describes the various methods used to label and track cells with MRI and the potential for such methods to translate to human applications.
Collapse
Affiliation(s)
- Matthew D Budde
- Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
82
|
Liu W, Frank JA. Detection and quantification of magnetically labeled cells by cellular MRI. Eur J Radiol 2008; 70:258-64. [PMID: 18995978 DOI: 10.1016/j.ejrad.2008.09.021] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 09/18/2008] [Indexed: 11/25/2022]
Abstract
Labeling cells with superparamagnetic iron oxide (SPIO) nanoparticles, paramagnetic contrast agent (gadolinium) or perfluorocarbons allows for the possibility of tracking single or clusters of labeled cells within target tissues following either direct implantation or intravenous injection. This review summarizes the practical issues regarding detection and quantification of magnetically labeled cells with various MRI contrast agents with a focus on SPIO nanoparticles.
Collapse
Affiliation(s)
- Wei Liu
- Philips Research North America, Briarcliff Manor, NY 10510, USA
| | | |
Collapse
|
83
|
Arbab AS, Janic B, Knight RA, Anderson SA, Pawelczyk E, Rad AM, Read EJ, Pandit SD, Frank JA. Detection of migration of locally implanted AC133+ stem cells by cellular magnetic resonance imaging with histological findings. FASEB J 2008; 22:3234-46. [PMID: 18556461 DOI: 10.1096/fj.07-105676] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study investigated the factors responsible for migration and homing of magnetically labeled AC133(+) cells at the sites of active angiogenesis in tumor. AC133(+) cells labeled with ferumoxide-protamine sulfate were mixed with either rat glioma or human melanoma cells and implanted in flank of nude mice. An MRI of the tumors including surrounding tissues was performed. Tumor sections were stained for Prussian blue (PB), platelet-derived growth factor (PDGF), hypoxia-inducible factor-1alpha (HIF-1alpha), stromal cell derived factor-1 (SDF-1), matrix metalloproteinase-2 (MMP-2), vascular endothelial growth factor (VEGF), and endothelial markers. Fresh snap-frozen strips from the central and peripheral parts of the tumor were collected for Western blotting. MRIs demonstrated hypointense regions at the periphery of the tumors where the PB(+)/AC133(+) cells were positive for endothelial cells markers. At the sites of PB(+)/AC133(+) cells, both HIF-1alpha and SDF-1 were strongly positive and PDGF and MMP-2 showed generalized expression in the tumor and surrounding tissues. There was no significant association of PB(+)/AC133(+) cell localization and VEGF expression in tumor cells. Western blot demonstrated strong expression of the SDF-1, MMP-2, and PDGF at the peripheral parts of the tumors. HIF-1alpha was expressed at both the periphery and central parts of the tumor. This work demonstrates that magnetically labeled cells can be used as probes for MRI and histological identification of administered cells.
Collapse
Affiliation(s)
- Ali S Arbab
- Department of Radiology, Henry Ford Health System, 1 Ford Pl., 2F, Box 82, Detroit, MI 48202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|