201
|
Abstract
With the recent advances in magnetic resonance (MR) labeling of cellular therapeutics, it is natural that interventional MRI techniques for targeting would be developed. This review provides an overview of the current methods of stem cell labeling and the challenges that are created with respect to interventional MRI administration. In particular, stem cell therapies will require specialized, MR-compatible devices as well as integration of graphical user interfaces with pulse sequences designed for interactive, real-time delivery in many organs. Specific applications that are being developed will be reviewed as well as strategies for future translation to the clinical realm.
Collapse
Affiliation(s)
- Dara L Kraitchman
- Johns Hopkins University, School of Medicine, Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD 21287, USA.
| | | | | |
Collapse
|
202
|
Elder JB, Liu CY, Apuzzo ML. NEUROSURGERY IN THE REALM OF 10−9, PART 2. Neurosurgery 2008; 62:269-84; discussion 284-5. [DOI: 10.1227/01.neu.0000315995.73269.c3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- James B. Elder
- Department of Neurological Surgery, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Charles Y. Liu
- Department of Neurological Surgery, University of Southern California, Keck School of Medicine, Los Angeles, California
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California
| | - Michael L.J. Apuzzo
- Department of Neurological Surgery, University of Southern California, Keck School of Medicine, Los Angeles, California
| |
Collapse
|
203
|
Neri M, Maderna C, Cavazzin C, Deidda-Vigoriti V, Politi LS, Scotti G, Marzola P, Sbarbati A, Vescovi AL, Gritti A. Efficient in vitro labeling of human neural precursor cells with superparamagnetic iron oxide particles: relevance for in vivo cell tracking. Stem Cells 2008; 26:505-16. [PMID: 17975226 DOI: 10.1634/stemcells.2007-0251] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Recent studies have raised appealing possibilities of replacing damaged or lost neural cells by transplanting in vitro-expanded neural precursor cells (NPCs) and/or their progeny. Magnetic resonance (MR) tracking of superparamagnetic iron oxide (SPIO)-labeled cells is a noninvasive technique to track transplanted cells in longitudinal studies on living animals. Murine NPCs and human mesenchymal or hematopoietic stem cells can be efficiently labeled by SPIOs. However, the validation of SPIO-based protocols to label human neural precursor cells (hNPCs) has not been extensively addressed. Here, we report the development and validation of optimized protocols using two SPIOs (Sinerem and Endorem) to label human hNPCs that display bona fide stem cell features in vitro. A careful titration of both SPIOs was required to set the conditions resulting in efficient cell labeling without impairment of cell survival, proliferation, self-renewal, and multipotency. In vivo magnetic resonance imaging (MRI) combined with histology and confocal microscopy indicated that low numbers (5 x 10(3) to 1 x 10(4)) of viable SPIO-labeled hNPCs could be efficiently detected in the short term after transplantation in the adult murine brain and could be tracked for at least 1 month in longitudinal studies. By using this approach, we also clarified the impact of donor cell death to the MR signal. This study describes a simple protocol to label NPCs of human origin using SPIOs at optimized low dosages and demonstrates the feasibility of noninvasive imaging of labeled cells after transplantation in the brain; it also evidentiates potential limitations of the technique that have to be considered, particularly in the perspective of neural cell-based clinical applications.
Collapse
Affiliation(s)
- Margherita Neri
- aStem Cell Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Abstract
Tumour responses to treatment are still largely assessed from imaging measurements of reductions in tumour size. However, this can take several weeks to become manifest and in some cases may not occur at all, despite a positive response to treatment. There has been considerable interest, therefore, in non-invasive techniques for imaging tissue function that can give early evidence of response. These can be used in clinical trials of new drugs to give an early indication of drug efficacy, and subsequently in the clinic to select the most effective therapy at an early stage of treatment.
Collapse
Affiliation(s)
- Kevin Brindle
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK.
| |
Collapse
|
205
|
Hoehn M, Himmelreich U, Kruttwig K, Wiedermann D. Molecular and cellular MR imaging: Potentials and challenges for neurological applications. J Magn Reson Imaging 2008; 27:941-54. [DOI: 10.1002/jmri.21280] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
206
|
Abstract
Cell-based therapies may gain future importance in defeating different kinds of diseases, including cancer, immunological disorders, neurodegenerative diseases, cardiac infarction and stroke. In this context, the noninvasive localization of the transplanted cells and the monitoring of their migration can facilitate basic research on the underlying mechanism and improve clinical translation. In this chapter, different ways to label and track cells in vivo are described. The oldest and only clinically established method is leukocyte scintigraphy, which enables a (semi)quantitative assessment of cell assemblies and, thus, the localization of inflammation foci. Noninvasive imaging of fewer or even single cells succeeds with MRI after labeling of the cells with (ultrasmall) superparamagentic iron oxide particles (SPIO and USPIO). However, in order to gain an acceptable signal-to-noise ratio, at a sufficiently high spatial resolution of the MR sequence to visualize a small amount of cells, experimental MR scanners working at high magnetic fields are usually required. Nevertheless, feasibility of clinical translation has been achieved by showing the localization of USPIO-labeled dendritic cells in cervical lymph nodes of patients by clinical MRI.Cell-tracking approaches using optical methods are important for preclinical research. Here, cells are labeled either with fluorescent dyes or quantum dots, or transfected with plasmids coding for fluorescent proteins such as green fluorescent protein (GFP) or red fluorescent protein (RFP). The advantage of the latter approach is that the label does not get lost during cell division and, thus, makes imaging of proliferating transplanted cells (e.g., tumor cells) possible. In summary, there are several promising options for noninvasive cell tracking, which have different strengths and limitations that should be considered when planning cell-tracking experiments.
Collapse
Affiliation(s)
- Fabian Kiessling
- Abteilung Medizinische Physik in der Radiologie, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg.
| |
Collapse
|
207
|
Callera F, de Melo CMTP. Magnetic resonance tracking of magnetically labeled autologous bone marrow CD34+ cells transplanted into the spinal cord via lumbar puncture technique in patients with chronic spinal cord injury: CD34+ cells' migration into the injured site. Stem Cells Dev 2007; 16:461-6. [PMID: 17610376 DOI: 10.1089/scd.2007.0083] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The purpose of this study was to demonstrate the possibility of delivering autologous bone marrow precursor cells into the spinal cord via lumbar puncture technique (LP) in patients with spinal cord injury (SCI). Magnetic resonance imaging provides a noninvasive method for studying the fate of transplanted cells in vivo. Considering these propositions, we studied magnetic resonance tracking of autologous bone marrow CD34(+) cells labeled with magnetic nanoparticles delivered into the spinal cord via LP in patients with SCI. Sixteen patients with chronic SCI were enrolled and divided into two groups; one group got their own labeled-CD34(+) cells injected into the spinal cord via LP (n = 10); the others received an injection, but it contained magnetic beads without stem cells (controls, n = 6). CD34(+) cells were magnetically labeled with magnetic beads coated with a monoclonal antibody specific for the CD34 cell membrane antigen. Magnetic resonance images were obtained by a standard turbospin echo-T2 weighted sequences before and 20 and 35 days after post-transplantation. The median number of CD34(+) cells injected via LP was 0.7 x 10(6) (range 0.45 to 1.22 x 10(6)). Magnetically labeled CD34(+) cells were visible at the lesion site as hypointense signals in five patients of the labeled-CD34(+) group 20 and 35 days after transplantation; these signals were not visible in any patient of the control group. We suggested for the first time that autologous bone marrow CD34(+) cells labeled with magnetic nanoparticles delivered into the spinal cord via LP technique migrated into the injured site in patients with chronic SCI.
Collapse
Affiliation(s)
- Fernando Callera
- Serviço de Hematologia e Hemoterapia de São José Dos Campos, São José Dos Campos-São Paulo, CEP 12210-040, Brasil.
| | | |
Collapse
|
208
|
Soenen SJH, Baert J, De Cuyper M. Optimal Conditions for Labelling of 3T3 Fibroblasts with Magnetoliposomes without Affecting Cellular Viability. Chembiochem 2007; 8:2067-77. [DOI: 10.1002/cbic.200700327] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
209
|
Liu PK, Mandeville JB, Guangping Dai, Jenkins BG, Kim YR, Liu CH. Transcription MRI: a new view of the living brain. Neuroscientist 2007; 14:503-20. [PMID: 18024855 DOI: 10.1177/1073858407309746] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Altered gene activities are underlying causes of many neurological disorders. The ability to detect, image, and report endogenous gene transcription using magnetic resonance (MR) holds great potential for providing significant clinical benefits. In this review, we present the development of conjugates consisting of gene-targeting short nucleic acids (oligodeoxynucleotides, or sODN) and superparamagnetic iron oxide nanoparticles (SPION, an MR susceptibility T(2) agent) for reporting gene activity using transcription MRI (tMRI). We will discuss 1) the target specificity of sODN, 2) selection of contrast agents for tMRI, 3) the distribution and uptake, 4) sequence specificity, 5) histology of SPION and sODN, 6) data acquisition and quantitative analysis for tMRI, and 7) application of gene transcript-targeting nanoparticles in biology and medicine. We will also discuss methods of validating the correlation between results from conventional assays (in situ hybridization, PCR, histology Prussian blue stain and immunohistochemistry) in postmortem samples and retention of SPION-sODN using tMRI. The application of our novel contrast probe to report and target gene transcripts in the mesolimbic pathways of living mouse brains after amphetamine exposure will be discussed. Because of the targeting ability in the nucleic acid sequence, the concept of tMRI probes with complementary nucleic acid (antisense DNA or short interfering RNA) allows not only tracking, targeting, binding to intracellular mRNA, and manipulating gene action but also tracing cells with specific gene action in living brains. Transcription MRI will lend itself to myriad applications in living organs.
Collapse
Affiliation(s)
- Philip K Liu
- Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA.
| | | | | | | | | | | |
Collapse
|
210
|
Abstract
Superparamagnetic iron oxide (SPIO) contrast agents, clinically established for high resolution magnetic resonance imaging of reticuloendothelial system containing anatomical structures, can additionally be exploited for the non-invasive characterization and quantification of pathology down to the molecular level. In this context, SPIOs can be applied for non-invasive cell tracking, quantification of tissue perfusion and target specific imaging, as well as for the detection of gene expression. This article provides an overview of new applications for clinically approved iron oxides as well of new, modified SPIO contrast agents for parametric and molecular imaging.
Collapse
Affiliation(s)
- L Matuszewski
- Institut für Klinische Radiologie, Universitätsklinikum Münster.
| | | | | | | |
Collapse
|
211
|
Abstract
Cell based therapies such as stem cell therapies or adoptive immunotherapies are currently being explored as a potential treatment for a variety of diseases such as Parkinson's disease, diabetes or cancer. However, quantitative and qualitative evaluation of adoptively transferred cells is indispensable for monitoring the efficiency of the treatment. Current approaches mostly analyze transferred cells from peripheral blood, which cannot assess whether transferred cells actually home to and stay in the targeted tissue. Using cell-labeling methods such as direct labeling or transfection with a marker gene in conjunction with various imaging modalities (MRI, optical or nuclear imaging), labeled cells can be followed in vivo in real-time, and their accumulation as well as function in vivo can be monitored and quantified accurately. This method is usually referred to as "cell tracking" or "cell trafficking" and is also being applied in basic biological sciences, exemplified in the evaluation of genes contributing to metastasis. This review focuses on principles of this promising methodology and explains various approaches by highlighting recent examples.
Collapse
Affiliation(s)
- J Grimm
- Dept. of Radiology, Memorial Sloan Kettering Cancer Center,1275 York Avenue, New York, NY 10021, USA.
| | | | | |
Collapse
|
212
|
Wu YJ, Muldoon LL, Varallyay C, Markwardt S, Jones RE, Neuwelt EA. In vivo leukocyte labeling with intravenous ferumoxides/protamine sulfate complex and in vitro characterization for cellular magnetic resonance imaging. Am J Physiol Cell Physiol 2007; 293:C1698-708. [PMID: 17898131 DOI: 10.1152/ajpcell.00215.2007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cellular labeling with ferumoxides (Feridex IV) superparamagnetic iron oxide nanoparticles can be used to monitor cells in vivo by MRI. The objective of this study was to use histology and MRI to evaluate an in vivo, as opposed to in vitro, technique for labeling of mononuclear leukocytes as a means of tracking inflammatory processes in the brain. Long-Evans rats were intravenously injected with 20 mg/kg ferumoxides, ferumoxtran-10, or ferumoxytol with or without protamine sulfate. Leukocytes and splenocytes were evaluated by cell sorting and iron histochemistry or were implanted into the brain for MRI. Injection of ferumoxides/protamine sulfate complex IV resulted in iron labeling of leukocytes (ranging from 7.4 ± 0.5% to 12.5 ± 0.9% with average 9.2 ± 0.8%) compared with ferumoxides (ranging from 3.9 ± 0.4% to 6.3 ± 0.5% with average 5.0 ± 0.5%) or protamine sulfate alone (ranging from 0% to 0.9 ± 0.7% with average 0.3 ± 0.3%). Cell sorting analysis indicated that iron-labeled cells were enriched for cell types positive for the myelomonocytic marker (CD11b/c) and the B lymphocyte marker (CD45RA) and depleted in the T cell marker (CD3). Neither ferumoxtran-10 nor ferumoxytol with protamine sulfate labeled leukocytes. In vivo ferumoxides/protamine sulfate-loaded leukocytes and splenocytes were detected by MRI after intracerebral injection. Ferumoxides/protamine complex labeled CD45RA-positive and CD11b/c-positive leukocytes in vivo without immediate toxicity. The dose of feumoxides in this report is much higher than the approved human dose, so additional animal studies are required before this approach could be translated to the clinic. These results might provide useful information for monitoring leukocyte trafficking into the brain.
Collapse
Affiliation(s)
- Y Jeffrey Wu
- Research Service, Veterans Administration Medical Center, Oregon Health and Sciences University, Portland, Oregon 97239, USA
| | | | | | | | | | | |
Collapse
|
213
|
Dousset V, Tourdias T, Brochet B, Boiziau C, Petry KG. How to trace stem cells for MRI evaluation? J Neurol Sci 2007; 265:122-6. [PMID: 17963784 DOI: 10.1016/j.jns.2007.09.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 09/09/2007] [Accepted: 09/12/2007] [Indexed: 11/20/2022]
Abstract
Applications of imaging techniques to visualize stem cells for monitoring, control and treatment of biological systems, in particular the brain, is at the forefront of investigations. These approaches involve the identification of stem and precursor cells that may be of various origins, but are related to specific clinical conditions, and the choice of the appropriate markers to achieve the required imaging while minimizing the side effects. This article will review examples of the contrast agent design for rational approaches in stem cell imaging. Potential pitfalls or side effects associated with contrast agents, in particular iron oxide nanoparticles, for cell labelling are also discussed.
Collapse
Affiliation(s)
- Vincent Dousset
- Laboratoire de Neurobiologie des Affections de la Myéline EA 2966 Université Victor Segalen Bordeaux 2, 146 rue Léo Saignat 33076 Bordeaux, France.
| | | | | | | | | |
Collapse
|
214
|
Sykova E, Jendelova P. In vivo tracking of stem cells in brain and spinal cord injury. PROGRESS IN BRAIN RESEARCH 2007; 161:367-83. [PMID: 17618991 DOI: 10.1016/s0079-6123(06)61026-1] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cellular magnetic resonance (MR) imaging is a rapidly growing field that aims to visualize and track cells in living organisms. Superparamagnetic iron oxide (SPIO) nanoparticles offer a sufficient signal for T2 weighted MR images. We followed the fate of embryonic stem cells (ESCs) and bone marrow mesenchymal stem cells (MSCs) labeled with iron oxide nanoparticles (Endorem) and human CD34+ cells labeled with magnetic MicroBeads (Miltenyi) in rats with a cortical or spinal cord lesion, models of stroke and spinal cord injury (SCI), respectively. Cells were either grafted intracerebrally, contralaterally to a cortical photochemical lesion, or injected intravenously. During the first post-transplantation week, grafted MSCs or ESCs migrated to the lesion site in the cortex as well as in the spinal cord and were visible in the lesion on MR images as a hypointensive signal, persisting for more than 30 days. In rats with an SCI, we found an increase in functional recovery after the implantation of MSCs or a freshly prepared mononuclear fraction of bone marrow cells (BMCs) or after an injection of granulocyte colony stimulating factor (G-CSF). Morphometric measurements in the center of the lesions showed an increase in white matter volume in cell-treated animals. Prussian blue staining confirmed a large number of iron-positive cells, and the lesions were considerably smaller than in control animals. Additionally, we implanted hydrogels based on poly-hydroxypropylmethacrylamide (HPMA) seeded with nanoparticle-labeled MSCs into hemisected rat spinal cords. Hydrogels seeded with MSCs were visible on MR images as hypointense areas, and subsequent Prussian blue histological staining confirmed positively stained cells within the hydrogels. To obtain better results with cell labeling, new polycation-bound iron oxide superparamagnetic nanoparticles (PC-SPIO) were developed. In comparison with Endorem, PC-SPIO demonstrated a more efficient intracellular uptake into MSCs, with no decrease in cell viability. Our studies demonstrate that magnetic resonance imaging (MRI) of grafted adult as well as ESCs labeled with iron oxide nanoparticles is a useful method for evaluating cellular migration toward a lesion site.
Collapse
Affiliation(s)
- Eva Sykova
- Institute of Experimental Medicine ASCR, EU Centre of Excellence, Prague, Czech Republic.
| | | |
Collapse
|
215
|
Walczak P, Kedziorek DA, Gilad AA, Barnett BP, Bulte JWM. Applicability and limitations of MR tracking of neural stem cells with asymmetric cell division and rapid turnover: the case of the shiverer dysmyelinated mouse brain. Magn Reson Med 2007; 58:261-9. [PMID: 17654572 DOI: 10.1002/mrm.21280] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
LacZ-transfected C17.2 neural stem cells (NSCs) were labeled with the superparamagnetic iron oxide formulation Feridex prior to ICV injection in shi/shi neonates. Feridex labeling did not alter cell differentiation in vitro and in vivo. Initially, MR images obtained at 11.7T correlated closely to NSC distribution as assessed with anti-dextran and anti-beta-galactosidase double-fluorescent immunostaining. However, at 6 days postgrafting there was already a pronounced mismatch between the hypointense MR signal and the histologically determined cell distribution, with a surprisingly sharp cutoff rather than a gradual decrease of signal. Positive in vivo BrdU labeling of NSCs showed that significant cell replication occurred post-transplantation, causing rapid dilution of Feridex particles between mother and daughter cells toward undetectable levels. Neural differentiation experiments demonstrated asymmetric cell division, explaining the observed sharp cutoff. At later time points (2 weeks), the mismatch further increased by the presence of non-cell-associated Feridex particles resulting from active excretion or cell death. These results are a first demonstration of the inability of MRI to track rapidly dividing and self-renewing, asymmetrically dividing SCs. Therefore, MR cell tracking should only be applied for nonproliferating cells or short-term monitoring of highly-proliferative cells, with mitotic symmetry or asymmetry being important for determining its applicability.
Collapse
Affiliation(s)
- P Walczak
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
216
|
Walczak P, Bulte JWM. The role of noninvasive cellular imaging in developing cell-based therapies for neurodegenerative disorders. NEURODEGENER DIS 2007; 4:306-13. [PMID: 17627134 DOI: 10.1159/000101887] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Stem and progenitor cells from various sources are currently recognized as entities with potential for the treatment of numerous neurodegenerative diseases. It has been observed in many animal models that transplantation of stem cells induces functional improvement. As a result of these findings, the first clinical cell transplantation trials were initiated, including those for Parkinson's disease and cerebral ischemia patients. However, in many patients, although modest improvements have been observed, these improvements were not sufficient to warrant invasive and possibly risky cell therapy. Thus, it is apparent that therapeutic success requires a better understanding of the mechanisms of action and the ability to control these mechanisms that underlie functional improvements, permitting amplification of the therapeutic effect. Considering the complexity of the nervous system, the task of repairing damaged or dysfunctional brain tissue with naïve cellular elements that require spatially and temporally accurate governance may seem daunting. However, the hope for faster and more inclusive progress in this field arises from recent developments in medical biotechnology that offers scientists increasingly sophisticated tools to study and control biological processes. One such technology with great potential for neurotransplantation is noninvasive cellular imaging. This tool allows real-time 'supervision' of grafted cells, as well as monitoring biodistribution and development. In this review, we highlight the current challenges in the field of cell-based therapy for neurodegenerative disorders and outline the role and capabilities of different cellular imaging techniques in addressing those issues.
Collapse
Affiliation(s)
- Piotr Walczak
- Russell H Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University, Baltimore, MD 21205-2195, USA
| | | |
Collapse
|
217
|
Ye Y, Bogaert J. Cell therapy in myocardial infarction: emphasis on the role of MRI. Eur Radiol 2007; 18:548-69. [DOI: 10.1007/s00330-007-0777-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 08/31/2007] [Accepted: 09/04/2007] [Indexed: 01/14/2023]
|
218
|
Hamasaki T, Tanaka N, Kamei N, Ishida O, Yanada S, Nakanishi K, Nishida K, Oishi Y, Kawamata S, Sakai N, Ochi M. Magnetically labeled neural progenitor cells, which are localized by magnetic force, promote axon growth in organotypic cocultures. Spine (Phila Pa 1976) 2007; 32:2300-5. [PMID: 17906569 DOI: 10.1097/brs.0b013e318154c651] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN An in vitro, rat animal study was conducted. OBJECTIVE To assess the corticospinal axon growth potential in varying concentrations of neural progenitor cells (NPCs) and in magnetically localized labeled NPCs, quantitatively using our original organotypic coculture system. SUMMARY OF BACKGROUND DATA Transplantation of NPCs for spinal cord injury has been anticipated as a possible future treatment. It is important not only to illuminate the mechanism of NPCs for spinal cord injury, but also to develop an effective cell delivery system for clinical use. In order to develop more effective, efficient, and minimally invasive cell delivery systems, we established a new system using magnetic targeting. METHODS Magnetically labeled NPCs were suspended with activated magnetic beads and individual NPCs, and were compared the characterization to nonlabeled NPCs in vitro. We transplanted varying concentrations of 10, 10, 10, 10, and 10 NPCs in 1 microL medium to coculture models. Then the 10 labeled NPCs were transplanted with or without magnet to the cocultures. RESULTS Magnetically labeled NPCs had similar potential in axon growth compared with nonlabeled NPCs, so there were few toxic effects of magnetically labeling NPCs. The differential potentials were not changed whether they were localized or scattered in vitro. Corticospinal axon growth was promoted in accordance with the transplanted NPC numbers around the organotypic coculture. Localized labeled NPCs with a magnet promoted axon growth much more than scattered labeled NPCs without a magnet, so magnetically localized labeled NPCs expressed higher potential in axon growth. CONCLUSION Magnetically labeled NPCs, which were localized by magnetic force, could promote axon growth in this organotypic coculture system.
Collapse
Affiliation(s)
- Takahiko Hamasaki
- Department of Orthopaedic Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Abstract
The use of molecular imaging techniques in the central nervous system (CNS) has a rich history. Most of the important developments in imaging-such as computed tomography, magnetic resonance imaging, single photon emission computed tomography, and positron emission tomography-began with neuropsychiatric applications. These techniques and modalities were then found to be useful for imaging other organs involved with various disease processes. Molecular imaging of the CNS has enabled scientists and researchers to understand better the basic biology of brain function and the way in which various disease processes affect the brain. Unlike other organs, the brain is not easily accessible, and it has a highly selective barrier at the endothelial cell level known as the blood-brain barrier. Furthermore, the brain is the most complex cellular network known to exist. Various neurotransmitters act in either an excitatory or an inhibitory fashion on adjacent neurons through a multitude of mechanisms. The various neuronal systems and the myriad of neurotransmitter systems become altered in many diseases. Some of the most devastating diseases, including Alzheimer disease, Parkinson disease, brain tumors, psychiatric disease, and numerous degenerative neurologic diseases, affect only the brain. Molecular neuroimaging will be critical to the future understanding and treatment of these diseases. Molecular neuroimaging of the brain shows tremendous promise for clinical application. In this article, the current state and clinical applications of molecular neuroimaging will be reviewed.
Collapse
Affiliation(s)
- Dima A Hammoud
- Department of Radiology, Johns Hopkins University School of Medicine, 1550 Orleans St, CRB-2, Room 492, Baltimore, MD 21231, USA
| | | | | |
Collapse
|
220
|
Scolding N, Marks D, Rice C. Autologous mesenchymal bone marrow stem cells: practical considerations. J Neurol Sci 2007; 265:111-5. [PMID: 17904159 DOI: 10.1016/j.jns.2007.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 08/08/2007] [Accepted: 08/09/2007] [Indexed: 10/22/2022]
Abstract
A number of practical problems need to be addressed before any form of cell therapy can be widely applied in patients with multiple sclerosis. The choice of cell type is one considered elsewhere in this issue; others include the question of axon loss, that of continuing inflammatory disease activity, the mode of delivery of cells (bearing in mind the presence of innumerable lesions scattered throughout the CNS), the problem of measuring directly or indirectly the impact (if any) of an intervention, the timing of any treatment and perhaps above all the safety of the patient. All converge on the one increasingly relevant underlying question: when should stem cell treatments begin to be tested in patients? Here we review the progress in various of these practical problems in order to explain how we have arrived at the conclusion that the clinical science has progressed to a stage where the 'translation threshold' can be safely and appropriately crossed, and therefore why we have already commenced in Bristol a small pilot/feasibility study of autologous bone marrow cell treatment in patients with multiple sclerosis.
Collapse
Affiliation(s)
- Neil Scolding
- University of Bristol Institute of Clinical Neurosciences, Department of Neurology, Frenchay Hospital, Bristol BS16 1LE, UK.
| | | | | |
Collapse
|
221
|
Gupta AK, Naregalkar RR, Vaidya VD, Gupta M. Recent advances on surface engineering of magnetic iron oxide nanoparticles and their biomedical applications. Nanomedicine (Lond) 2007; 2:23-39. [PMID: 17716188 DOI: 10.2217/17435889.2.1.23] [Citation(s) in RCA: 402] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Magnetic nanoparticles with appropriate surface coatings are increasingly being used clinically for various biomedical applications, such as magnetic resonance imaging, hyperthermia, drug delivery, tissue repair, cell and tissue targeting and transfection. This is because of the nontoxicity and biocompatibility demand that mainly iron oxide-based materials are predominantly used, despite some attempts to develop 'more magnetic nanomaterials' based on cobalt, nickel, gadolinium and other compounds. For all these applications, the material used for surface coating of the magnetic particles must not only be nontoxic and biocompatible but also allow a targetable delivery with particle localization in a specific area. Magnetic nanoparticles can bind to drugs and an external magnetic field can be applied to trap them in the target site. By attaching the targeting molecules, such as proteins or antibodies, at particles surfaces, the latter may be directed to any cell, tissue or tumor in the body. In this review, different polymers/molecules that can be used for nanoparticle coating to stabilize the suspensions of magnetic nanoparticles under in vitro and in vivo situations are discussed. Some selected proteins/targeting ligands that could be used for derivatizing magnetic nanoparticles are also explored. We have reviewed the various biomedical applications with some of the most recent uses of magnetic nanoparticles for early detection of cancer, diabetes and atherosclerosis.
Collapse
Affiliation(s)
- Ajay Kumar Gupta
- Laboratory for Nanoparticle Research, Formulation & Development Department, Torrent Research Centre, TPL, Village Bhat, Dist. Gandhinagar-382 428, Gujarat, India.
| | | | | | | |
Collapse
|
222
|
Shokouhimehr M, Piao Y, Kim J, Jang Y, Hyeon T. A Magnetically Recyclable Nanocomposite Catalyst for Olefin Epoxidation. Angew Chem Int Ed Engl 2007. [DOI: 10.1002/ange.200702386] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
223
|
Kuhlpeter R, Dahnke H, Matuszewski L, Persigehl T, von Wallbrunn A, Allkemper T, Heindel WL, Schaeffter T, Bremer C. R2 and R2* mapping for sensing cell-bound superparamagnetic nanoparticles: in vitro and murine in vivo testing. Radiology 2007; 245:449-57. [PMID: 17848680 DOI: 10.1148/radiol.2451061345] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To prospectively determine the cellular iron uptake by using R2 and R2* mapping with multiecho readout gradient-echo and spin-echo sequences. MATERIALS AND METHODS All experiments were approved by the institutional animal care committee. Lung carcinoma cells were lipofected with superparamagnetic iron oxides (SPIOs). Agarose gel phantoms containing (a) 1 x 10(5) CCL-185 cells per milliliter of agarose gel with increasing SPIO load (0.01-5.00 mg of iron per milliliter in the medium), (b) different amounts (5.0 x 10(3) to 2.5 x 10(5) cells per milliliter of agarose gel) of identically loaded cells, and (c) free (non-cell-bound) SPIOs at the iron concentrations described for (b) were analyzed with 3.0-T R2 and R2* relaxometry. Iron uptake was analyzed with light microscopy, quantified with atomic emission spectroscopy (AES), and compared with MR data. For in vivo relaxometry, agarose gel pellets containing SPIO-labeled cells, free SPIO, unlabeled control cells, and pure agarose gel were injected into three nude mice each. Linear and nonlinear regression analyses were performed. RESULTS Light microscopy and AES revealed efficient SPIO particle uptake (mean uptake: 0.22 pg of iron per cell +/- 0.1 [standard deviation] for unlabeled cells, 31.17 pg of iron per cell +/- 4.63 for cells incubated with 0.5 mg/mL iron). R2 and R2* values were linearly correlated with cellular iron load, number of iron-loaded cells, and content of freely dissolved iron (r(2) range, 0.92-0.99; P < .001). For cell-bound SPIO, R2* effects were significantly greater than R2 effects (P < .01); for free SPIO, R2 and R2* effects were similar. In vivo relaxometry enabled accurate prediction of the number of labeled cells. R2' (R2* - R2) mapping enabled differentiation between cell-bound and free iron in vitro and in vivo. CONCLUSION Quantitative R2 and R2* mapping enables noninvasive estimations of cellular iron load and number of iron-labeled cells. Cell-bound SPIOs can be differentiated from free SPIOs with R2' imaging.
Collapse
Affiliation(s)
- Rebecca Kuhlpeter
- Department of Clinical Radiology, University Hospital of Muenster, Albert-Schweitzer-Str 33, D-48129, Muenster, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Sumner JP, Conroy R, Shapiro E, Moreland J, Koretsky AP. Delivery of fluorescent probes using iron oxide particles as carriers enables in-vivo labeling of migrating neural precursors for magnetic resonance imaging and optical imaging. JOURNAL OF BIOMEDICAL OPTICS 2007; 12:051504. [PMID: 17994868 PMCID: PMC3529473 DOI: 10.1117/1.2800294] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Iron oxide particles are becoming an important contrast agent for magnetic resonance imaging (MRI) cell tracking studies. Simultaneous delivery of fluorescence indicators with the particles to individual cells offers the possibility of correlating optical images and MRI. In this work, it is demonstrated that micron-sized iron oxide particles (MPIOs) can be used as a carrier to deliver fluorescent probes to cells in culture as well as to migrating neural progenitors in vivo. Migrating progenitors were tracked with MRI and easily identified by histology because of the fluorescent probe. These data suggest that using MPIOs to deliver fluorescent probes should make it possible to combine MRI and optical imaging for in vivo cell tracking.
Collapse
Affiliation(s)
- James P. Sumner
- Laboratory of Functional and Molecular Imaging, NINDS, NIH, Bethesda, MD
| | - Richard Conroy
- Laboratory of Functional and Molecular Imaging, NINDS, NIH, Bethesda, MD
- National Institutes of Standards and Technology, Boulder, CO
| | - Erik Shapiro
- Department of Radiology, Yale Medical School, New Haven, CT
| | - John Moreland
- National Institutes of Standards and Technology, Boulder, CO
| | - Alan P. Koretsky
- Laboratory of Functional and Molecular Imaging, NINDS, NIH, Bethesda, MD
| |
Collapse
|
225
|
Ju S, Teng GJ, Lu H, Zhang Y, Zhang A, Chen F, Ni Y. In vivo MR tracking of mesenchymal stem cells in rat liver after intrasplenic transplantation. Radiology 2007; 245:206-15. [PMID: 17717324 DOI: 10.1148/radiol.2443061290] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To prospectively track in vivo in rats intrasplenically transplanted stem cells labeled with superparamagnetic particles by using magnetic resonance (MR) imaging. MATERIALS AND METHODS The study was approved by the institutional Committee on Animal Research. Liver damage in 12 rats was induced with subcutaneous injection of carbon tetrachloride (CCl4). Intrasplenic transplantation of 6x10(6) rodent bone mesenchymal stem cells (BMSCs) with (n=6) and without (n=6) superparamagnetic particle Fe2O3-poly-L-lysine (PLL) labeling was performed via direct puncture. Cell labeling efficiency was assessed in vitro by using Prussian blue stain and an atomic absorption spectrometer. MR examinations were performed immediately before and 3 hours and 3, 7, and 14 days after transplantation. Liver-to-muscle contrast-to-noise ratios (CNRs) on T2*-weighted MR images obtained before and after injection were measured and correlated with histomorphologic studies. Statistical analyses were performed by using repeated-measures analysis of variance. RESULTS Rat BMSCs could be effectively labeled with approximately 100% efficiency. Migration of transplanted labeled cells to the liver was successfully documented with in vivo MR imaging. CNRs on T2*-weighted images decreased significantly in the liver 3 hours after injection of BMSCs (P<.05) and returned gradually to the level achieved without labeled cell injection in 14 days. Histologic analyses confirmed the presence of BMSCs in the liver. The labeled cells primarily localized in the sinusoids of periportal areas and the foci of CCl4-induced liver damage. Quantitative analysis of Prussian blue-stained cells indicated gradual decrease of dye pigments from 3 hours to 3, 7, and 14 days after injection. No free iron particles were found in the interstitium or within hepatic microvessels. CONCLUSION The rat BMSCs could be efficiently labeled with Fe2O3-PLL and the relocation of the labeled cells to rat livers after intrasplenic transplantation could be depicted at in vivo MR imaging.
Collapse
Affiliation(s)
- Shenghong Ju
- Laboratory of Molecular Imaging, Department of Radiology, Zhongda Hospital, Laboratory of Molecular and Biomolecular Electronics, and School of Basic Medical Science, Southeast University, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | | | | | | | | | | | | |
Collapse
|
226
|
Ittrich H, Lange C, Tögel F, Zander AR, Dahnke H, Westenfelder C, Adam G, Nolte-Ernsting C. In vivo magnetic resonance imaging of iron oxide-labeled, arterially-injected mesenchymal stem cells in kidneys of rats with acute ischemic kidney injury: detection and monitoring at 3T. J Magn Reson Imaging 2007; 25:1179-91. [PMID: 17520738 DOI: 10.1002/jmri.20925] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To evaluate MRI for a qualitative and quantitative in vivo tracking of intraaortal injected iron oxide-labeled mesenchymal stem cells (MSC) into rats with acute kidney injury (AKI). MATERIALS AND METHODS In vitro MRI and R2* measurement of nonlabeled and superparamagnetic iron oxide (SPIO)-labeled MSC (MSC(SPIO)) was performed in correlation to cellular iron content and cytological examination (Prussian blue, electron microscopy). In vivo MRI and R2* evaluation were performed before and after ischemic/reperfusion AKI (N = 14) and intraaortal injection of 1.5 x 10(6) MSC(SPIO) (N = 7), fetal calf serum (FCS) (medium, N = 6), and SPIO alone (N = 1) up to 14 days using a clinical 3T scanner. Signal to noise ratios (SNR), R2* of kidneys, liver, spleen, and bone marrow, renal function (creatinine [CREA], blood urea nitrogen [BUN]), and kidney volume were measured and tested for statistical significance (Student's t-test, P < 0.05) in comparison histology (hematoxylin and eosin [H&E], Prussian blue, periodic acid-Schiff [PAS], CD68). RESULTS In vitro, MSC(SPIO) showed a reduction of SNR and T2* with R2* approximately number of MSC(SPIO) (R2 = 0.98). In vivo MSC(SPIO) administration resulted in a SNR decrease (35 +/- 15%) and R2* increase (101 +/- 18.3%) in renal cortex caused by MSC(SPIO) accumulation in contrast to control animals (P < 0.01). Liver, spleen, and bone marrow (MSC(SPIO)) showed a delayed SNR decline/R2* increase (P < 0.05) resulting from MSC(SPIO) migration. The increase of kidney volume and the decrease in renal function (P < 0.05) was reduced in MSC-treated animals. CONCLUSION Qualitative and quantitative in vivo cell-tracking and monitoring of organ distribution of intraaortal injected MSC(SPIO) in AKI is feasible in MRI at 3T.
Collapse
Affiliation(s)
- Harald Ittrich
- Department of Diagnostic and Interventional Radiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, DE-20246 Hamburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
227
|
Hoehn M, Wiedermann D, Justicia C, Ramos-Cabrer P, Kruttwig K, Farr T, Himmelreich U. Cell tracking using magnetic resonance imaging. J Physiol 2007; 584:25-30. [PMID: 17690140 PMCID: PMC2277052 DOI: 10.1113/jphysiol.2007.139451] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Cell tracking by in vivo magnetic resonance imaging (MRI) requires strategies of labelling the cells with MRI contrast agents. The principal routes to achieve efficient cell labelling for neurological applications are discussed with methodological advantages and caveats. Beyond temporo-spatial localization of labelled cells, the investigation of functional cell status is of great interest to allow studies of functional cell dynamics. The two major approaches to reach this goal, use of responsive contrast agents and generation of transgenic cell lines, are discussed.
Collapse
Affiliation(s)
- Mathias Hoehn
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research, Gleueler Strasse 50, D-50931 Köln, Germany.
| | | | | | | | | | | | | |
Collapse
|
228
|
Rad AM, Arbab AS, Iskander ASM, Jiang Q, Soltanian-Zadeh H. Quantification of superparamagnetic iron oxide (SPIO)-labeled cells using MRI. J Magn Reson Imaging 2007; 26:366-74. [PMID: 17623892 PMCID: PMC4509786 DOI: 10.1002/jmri.20978] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE To show the feasibility of using magnetic resonance imaging (MRI) to quantify superparamagnetic iron oxide (SPIO)-labeled cells. MATERIALS AND METHODS Lymphocytes and 9L rat gliosarcoma cells were labeled with ferumoxides-protamine sulfate complex (FE-PRO). The cells were labeled efficiently (more than 95%) and the iron concentration inside each cell was measured by spectrophotometry (4.77-30.21 pg). Phantom tubes containing different numbers of labeled or unlabeled cells, as well as different concentrations of FE-PRO, were made. In addition, labeled and unlabeled cells were injected into fresh and fixed rat brains. RESULTS Cellular viability and proliferation of labeled and unlabeled cells were shown to be similar. T2-weighted images were acquired using 7T and 3T MRI systems, and R2 maps of the tubes containing cells, free FE-PRO, and brains were made. There was a strong linear correlation between R2 values and labeled cell numbers, but the regression lines were different for the lymphocytes and gliosarcoma cells. Similarly, there was strong correlation between R2 values and free iron. However, free iron had higher R2 values than the labeled cells for the same concentration of iron. CONCLUSION Our data indicate that in vivo quantification of labeled cells can be done by careful consideration of different factors and specific control groups.
Collapse
Affiliation(s)
- Ali M Rad
- Department of Radiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Ali S Arbab
- Department of Radiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - ASM Iskander
- Department of Radiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Hamid Soltanian-Zadeh
- Department of Radiology, Henry Ford Health System, Detroit, MI 48202, USA
- Control and Intelligent Processing Center of Excellence, Department of the Electrical and Computer Engineering, University of Tehran, Tehran, Iran
| |
Collapse
|
229
|
Fortin JP, Gazeau F, Wilhelm C. Intracellular heating of living cells through Néel relaxation of magnetic nanoparticles. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2007; 37:223-8. [PMID: 17641885 DOI: 10.1007/s00249-007-0197-4] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 05/23/2007] [Accepted: 05/28/2007] [Indexed: 10/23/2022]
Abstract
Maghemite and cobalt ferrite anionic magnetic nanoparticles enter tumor cells and can be used as heat sources when exposed to a high-frequency magnetic field. Comparative studies of the two particles enable to unravel the magnetic heating mechanisms (Néel relaxation vs. Brown relaxation) responsible for the cellular temperature rise, and also to establish a simple model, adjusted to the experimental results, allowing to predict the intracellular heating efficiency of iron oxide nanoparticles. Hence, we are able to derive the best nanoparticle design for a given material with a view to intracellular hyperthermia-based applications.
Collapse
|
230
|
Na H, Lee J, An K, Park Y, Park M, Lee I, Nam DH, Kim S, Kim SH, Kim SW, Lim KH, Kim KS, Kim SO, Hyeon T. Development of aT1 Contrast Agent for Magnetic Resonance Imaging Using MnO Nanoparticles. Angew Chem Int Ed Engl 2007. [DOI: 10.1002/ange.200604775] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
231
|
Na HB, Lee JH, An K, Park YI, Park M, Lee IS, Nam DH, Kim ST, Kim SH, Kim SW, Lim KH, Kim KS, Kim SO, Hyeon T. Development of aT1 Contrast Agent for Magnetic Resonance Imaging Using MnO Nanoparticles. Angew Chem Int Ed Engl 2007; 46:5397-401. [PMID: 17357103 DOI: 10.1002/anie.200604775] [Citation(s) in RCA: 402] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hyon Bin Na
- National Creative Research Initiative Center for Oxide Nanocrystalline Materials and School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Slotkin JR, Cahill KS, Tharin SA, Shapiro EM. Cellular magnetic resonance imaging: nanometer and micrometer size particles for noninvasive cell localization. Neurotherapeutics 2007; 4:428-33. [PMID: 17599708 PMCID: PMC7479728 DOI: 10.1016/j.nurt.2007.05.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The use of nanometer and micrometer-sized superparamagnetic iron oxide particles as cellular contrast agents allows for the noninvasive detection of labeled cells on high-resolution magnetic resonance images. The development and application of these techniques to neurologic disorders is likely to accelerate the development of cell transplantation therapies and allow for the detailed study of in vivo cellular biology. This review summarizes the early development of iron oxide-based cellular contrast agents and the more recent application of this technology to noninvasive imaging of cellular transplants. The ability of this technique to allow for the noninvasive detection of in vivo transplants on the single-cell level is highlighted.
Collapse
Affiliation(s)
- Jonathan R. Slotkin
- Department of Neurosurgery, The Brigham and Women’s Hospital, The Children’s Hospital, Boston, Harvard Medical School, 02115 Boston, Massachusetts
| | - Kevin S. Cahill
- Department of Neurosurgery, The Brigham and Women’s Hospital, The Children’s Hospital, Boston, Harvard Medical School, 02115 Boston, Massachusetts
| | - Suzanne A. Tharin
- Department of Neurosurgery, The Brigham and Women’s Hospital, The Children’s Hospital, Boston, Harvard Medical School, 02115 Boston, Massachusetts
| | - Erik M. Shapiro
- Department of Diagnostic Radiology, Yale University School of Medicine, 06510 New Haven, Connecticut
| |
Collapse
|
233
|
Politi LS, Bacigaluppi M, Brambilla E, Cadioli M, Falini A, Comi G, Scotti G, Martino G, Pluchino S. Magnetic-resonance-based tracking and quantification of intravenously injected neural stem cell accumulation in the brains of mice with experimental multiple sclerosis. Stem Cells 2007; 25:2583-92. [PMID: 17600110 DOI: 10.1634/stemcells.2007-0037] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Eliciting the in situ accumulation and persistence patterns of stem cells following transplantation would provide critical insight toward human translation of stem cell-based therapies. To this end, we have developed a strategy to track neural stem/precursor cells (NPCs) in vivo using magnetic resonance (MR) imaging. Initially, we evaluated three different human-grade superparamagnetic iron oxide particles for labeling NPCs and found the optimal labeling to be achieved with Resovist. Next, we carried out in vivo experiments to monitor the accumulation of Resovist-labeled NPCs following i.v. injection in mice with experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis. With a human MR scanner, we were able to visualize transplanted cells as early as 24 hours post-transplantation in up to 80% of the brain demyelinating lesions. Interestingly, continued monitoring of transplanted mice indicated that labeled NPCs were still present 20 days postinjection. Neuropathological analysis confirmed the presence of transplanted NPCs exclusively in inflammatory demyelinating lesions and not in normal-appearing brain areas. Quantification of transplanted cells by means of MR-based ex vivo relaxometry (R2*) showed significantly higher R2* values in focal inflammatory brain lesions from EAE mice transplanted with labeled NPCs as compared with controls. Indeed, sensitive quantification of low numbers of NPCs accumulating into brain inflammatory lesions (33.3-164.4 cells per lesion; r(2) = .998) was also obtained. These studies provide evidence that clinical-grade human MR can be used for noninvasive monitoring and quantification of NPC accumulation in the central nervous system upon systemic cell injection. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Letterio S Politi
- Neuroradiology Unit, Centro Eccellenza Risonanza Magnetica ad Alto Campo, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Kim J, Park S, Lee JE, Jin SM, Lee JH, Lee IS, Yang I, Kim JS, Kim SK, Cho MH, Hyeon T. Designed fabrication of multifunctional magnetic gold nanoshells and their application to magnetic resonance imaging and photothermal therapy. Angew Chem Int Ed Engl 2007; 45:7754-8. [PMID: 17072921 DOI: 10.1002/anie.200602471] [Citation(s) in RCA: 331] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Jaeyun Kim
- National Creative Research Initiative Center for Oxide Nanocrystalline Materials and School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Lai JJ, Hoffman JM, Ebara M, Hoffman AS, Estournès C, Wattiaux A, Stayton PS. Dual magnetic-/temperature-responsive nanoparticles for microfluidic separations and assays. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2007; 23:7385-91. [PMID: 17503854 DOI: 10.1021/la062527g] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
A stimuli-responsive magnetic nanoparticle system for diagnostic target capture and concentration has been developed for microfluidic lab card settings. Telechelic poly(N-isopropylacrylamide) (PNIPAAm) polymer chains were synthesized with dodecyl tails at one end and a reactive carboxylate at the opposite end by the reversible addition fragmentation transfer technique. These PNIPAAm chains self-associate into nanoscale micelles that were used as dimensional confinements to synthesize the magnetic nanoparticles. The resulting superparamagnetic nanoparticles exhibit a gamma-Fe2O3 core ( approximately 5 nm) with a layer of carboxylate-terminated PNIPAAm chains as a corona on the surface. The carboxylate group was used to functionalize the magnetic nanoparticles with biotin and subsequently with streptavidin. The functionalized magnetic nanoparticles can be reversibly aggregated in solution as the temperature is cycled through the PNIPAAm lower critical solution temperature (LCST). While the magnetophoretic mobility of the individual nanoparticles below the LCST is negligible, the aggregates formed above the LCST are large enough to respond to an applied magnetic field. The magnetic nanoparticles can associate with biotinylated targets as individual particles, and then subsequent application of a combined temperature increase and magnetic field can be used to magnetically separate the aggregated particles onto the poly(ethylene glycol)-modified polydimethylsiloxane channel walls of a microfluidic device. When the magnetic field is turned off and the temperature is reversed, the captured aggregates redisperse into the channel flow stream for further downstream processing. The dual magnetic- and temperature-responsive nanoparticles can thus be used as soluble reagents to capture diagnostic targets at a controlled time point and channel position. They can then be isolated and released after the nanoparticles have captured target molecules, overcoming the problem of low magnetophoretic mobility of the individual particle while retaining the advantages of a high surface to volume ratio and faster diffusive properties during target capture.
Collapse
Affiliation(s)
- James J Lai
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | |
Collapse
|
236
|
Park J, Joo J, Kwon S, Jang Y, Hyeon T. Synthese monodisperser sphärischer Nanokristalle. Angew Chem Int Ed Engl 2007. [DOI: 10.1002/ange.200603148] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
237
|
Park J, Joo J, Kwon SG, Jang Y, Hyeon T. Synthesis of Monodisperse Spherical Nanocrystals. Angew Chem Int Ed Engl 2007; 46:4630-60. [PMID: 17525914 DOI: 10.1002/anie.200603148] [Citation(s) in RCA: 1027] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Much progress has been made over the past ten years on the synthesis of monodisperse spherical nanocrystals. Mechanistic studies have shown that monodisperse nanocrystals are produced when the burst of nucleation that enables separation of the nucleation and growth processes is combined with the subsequent diffusion-controlled growth process through which the crystal size is determined. Several chemical methods have been used to synthesize uniform nanocrystals of metals, metal oxides, and metal chalcogenides. Monodisperse nanocrystals of CdSe, Co, and other materials have been generated in surfactant solution by nucleation induced at high temperature, and subsequent aging and size selection. Monodisperse nanocrystals of many metals and metal oxides, including magnetic ferrites, have been synthesized directly by thermal decomposition of metal-surfactant complexes prepared from the metal precursors and surfactants. Nonhydrolytic sol-gel reactions have been used to synthesize various transition-metal-oxide nanocrystals. Monodisperse gold nanocrystals have been obtained from polydisperse samples by digestive-ripening processes. Uniform-sized nanocrystals of gold, silver, platinum, and palladium have been synthesized by polyol processes in which metal salts are reduced by alcohols in the presence of appropriate surfactants.
Collapse
Affiliation(s)
- Jongnam Park
- National Creative Research Initiative Center for Oxide Nanocrystalline Materials, School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Korea
| | | | | | | | | |
Collapse
|
238
|
Guzman R, Uchida N, Bliss TM, He D, Christopherson KK, Stellwagen D, Capela A, Greve J, Malenka RC, Moseley ME, Palmer TD, Steinberg GK. Long-term monitoring of transplanted human neural stem cells in developmental and pathological contexts with MRI. Proc Natl Acad Sci U S A 2007; 104:10211-6. [PMID: 17553967 PMCID: PMC1891235 DOI: 10.1073/pnas.0608519104] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Noninvasive monitoring of stem cells, using high-resolution molecular imaging, will be instrumental to improve clinical neural transplantation strategies. We show that labeling of human central nervous system stem cells grown as neurospheres with magnetic nanoparticles does not adversely affect survival, migration, and differentiation or alter neuronal electrophysiological characteristics. Using MRI, we show that human central nervous system stem cells transplanted either to the neonatal, the adult, or the injured rodent brain respond to cues characteristic for the ambient microenvironment resulting in distinct migration patterns. Nanoparticle-labeled human central nervous system stem cells survive long-term and differentiate in a site-specific manner identical to that seen for transplants of unlabeled cells. We also demonstrate the impact of graft location on cell migration and describe magnetic resonance characteristics of graft cell death and subsequent clearance. Knowledge of migration patterns and implementation of noninvasive stem cell tracking might help to improve the design of future clinical neural stem cell transplantation.
Collapse
Affiliation(s)
- Raphael Guzman
- *Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, R200, Stanford, CA 94305-5327
| | - Nobuko Uchida
- StemCells, Inc., 3155 Porter Drive, Palo Alto, CA 94304-1213
| | - Tonya M. Bliss
- *Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, R200, Stanford, CA 94305-5327
| | - Dongping He
- StemCells, Inc., 3155 Porter Drive, Palo Alto, CA 94304-1213
| | | | - David Stellwagen
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, MSLS P104, Stanford, CA 94305-5485; and
| | - Alexandra Capela
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, MSLS P104, Stanford, CA 94305-5485; and
| | - Joan Greve
- Department of Radiology, Lucas Magnetic Resonance Spectroscopy and Imaging Center, Stanford University School of Medicine, P286, Stanford, CA 94022
| | - Robert C. Malenka
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, MSLS P104, Stanford, CA 94305-5485; and
| | - Michael E. Moseley
- Department of Radiology, Lucas Magnetic Resonance Spectroscopy and Imaging Center, Stanford University School of Medicine, P286, Stanford, CA 94022
| | - Theo D. Palmer
- *Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, R200, Stanford, CA 94305-5327
| | - Gary K. Steinberg
- *Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, R200, Stanford, CA 94305-5327
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
239
|
Liu CH, Huang S, Kim YR, Rosen BR, Liu PK. Forebrain ischemia-reperfusion simulating cardiac arrest in mice induces edema and DNA fragmentation in the brain. Mol Imaging 2007; 6:156-70. [PMID: 17532882 PMCID: PMC2644455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023] Open
Abstract
Brain injury affects one-third of persons who survive after heart attack, even with restoration of spontaneous circulation by cardiopulmonary resuscitation. We studied brain injury resulting from transient bilateral carotid artery occlusion (BCAO) and reperfusion by simulating heart attack and restoration of circulation, respectively, in live C57Black6 mice. This model is known to induce neuronal death in the hippocampus, striatum, and cortex. We report the appearance of edema after transient BCAO of 60 minutes and 1 day of reperfusion. Hyperintensity in diffusion-weighted magnetic resonance imaging (MRI) was detectable in the striatum, thalamus, and cortex but not in the hippocampus. To determine whether damage to the hippocampus can be detected in live animals, we infused a T(2) susceptibility magnetic resonance contrast agent (superparamagnetic iron oxide nanoparticles [SPIONs]) that was linked to single-stranded deoxyribonucleic acid (DNA) complementary in sequence to c-fos messenger ribonucleic acid (SPION-cfos); we acquired in vivo T(2)*-weighted MRI 3 days later. SPION retention was measured as T(2)* (milliseconds) signal reduction or R(2)* value (s(-1)) elevation. We found that animals treated with 60-minute BCAO and 7-day reperfusion exhibited significantly less SPION retention in the hippocampus and cortex than sham-operated animals. These findings suggest that brain injury induced by cardiac arrest can be detected in live animals.
Collapse
Affiliation(s)
- Christina H Liu
- A.A. Martinos Center for Biomedical Imaging Charlestown, MA, USA
| | | | | | | | | |
Collapse
|
240
|
Gilad AA, Winnard PT, van Zijl PCM, Bulte JWM. Developing MR reporter genes: promises and pitfalls. NMR IN BIOMEDICINE 2007; 20:275-90. [PMID: 17451181 DOI: 10.1002/nbm.1134] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
MR reporter genes have the potential to monitor transgene expression non-invasively in real time at high resolution. These genes can be applied to interrogate the efficacy of gene therapy, to assess cellular differentiation, cell trafficking, and specific metabolic activity, and also assess changes in the microenvironment. Efforts toward the development of MR reporter genes have been made for at least a decade, but, despite these efforts, the field is still in its early developmental stage. This reflects the fact that there are potential pitfalls, caused by the low sensitivity of detection, the need for substrates with their associated undesirable pharmacokinetics, and/or the difficult and, in some cases, delayed interpretation of signal changes. Nevertheless, significant progress has been made during the last few years. Whereas enzyme-based reporters were initially applied to NMR spectroscopic monitoring of changes in phosphor and fluorine metabolism, MRI-based approaches are now emerging that rely on: (1) enzyme-based cleavage of functional groups that block water (proton) exchange or protein binding of MR contrast agents; (2) expression of surface receptors that enable binding of specific MR contrast agents; (3) expression of para- and anti-ferromagnetic (metallo)proteins involved with iron metabolism, such as tyrosinase, transferrin receptor, and ferritin. After an introduction to the basic principles of designing promoters, expression vectors, and cloning of transgenes, a fresh look is provided on the use of reporter genes for optical (including bioluminescent) and nuclear imaging, with which MR reporter genes compete. Although progress in the use of MR reporter genes has been slow, newer strategies that use metalloproteins or alternative contrast mechanisms, with no need for substrates, promise rapid growth potential for this field.
Collapse
Affiliation(s)
- Assaf A Gilad
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
241
|
Liu CH, Huang S, Kim YR, Rosen BR, Liu PK. Forebrain Ischemia-Reperfusion Simulating Cardiac Arrest in Mice Induces Edema and DNA Fragmentation in the Brain. Mol Imaging 2007. [DOI: 10.2310/7290.2007.00011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Christina H. Liu
- From the A.A. Martinos Center for Biomedical Imaging Charlestown, MA; the Transcript Imaging and NeuroRepair Laboratory, Department of Radiology, Massachusetts General Hospital Charlestown, MA; and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA
| | - Shuning Huang
- From the A.A. Martinos Center for Biomedical Imaging Charlestown, MA; the Transcript Imaging and NeuroRepair Laboratory, Department of Radiology, Massachusetts General Hospital Charlestown, MA; and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA
| | - Young R. Kim
- From the A.A. Martinos Center for Biomedical Imaging Charlestown, MA; the Transcript Imaging and NeuroRepair Laboratory, Department of Radiology, Massachusetts General Hospital Charlestown, MA; and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA
| | - Bruce R. Rosen
- From the A.A. Martinos Center for Biomedical Imaging Charlestown, MA; the Transcript Imaging and NeuroRepair Laboratory, Department of Radiology, Massachusetts General Hospital Charlestown, MA; and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA
| | - Philip K. Liu
- From the A.A. Martinos Center for Biomedical Imaging Charlestown, MA; the Transcript Imaging and NeuroRepair Laboratory, Department of Radiology, Massachusetts General Hospital Charlestown, MA; and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA
| |
Collapse
|
242
|
Syková E, Jendelová P. Migration, fate and in vivo imaging of adult stem cells in the CNS. Cell Death Differ 2007; 14:1336-42. [PMID: 17396130 DOI: 10.1038/sj.cdd.4402140] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adult stem cells have been intensively studied for their potential use in cell therapies for neurodegenerative diseases, ischemia and traumatic injuries. One of the most promising cell sources for autologous cell transplantation is bone marrow, containing a heterogenous cell population that can be roughly divided into hematopoietic stem and progenitor cells and mesenchymal stem cells (MSCs). MSCs are multipotent progenitor cells that, in the case of severe tissue ischemia or damage, can be attracted to the lesion site, where they can secrete bioactive molecules, either naturally or through genetic engineering. They can also serve as vehicles for delivering therapeutic agents. Mobilized from the marrow, sorted or expanded in culture, MSCs can be delivered to the damaged site by direct or systemic application. In addition, MSCs can be labeled with superparamagnetic nanoparticles that allow in vivo cell imaging. Magnetic resonance imaging (MRI) is thus a suitable method for in vivo cell tracking of transplanted cells in the host organism. This review will focus on cell labeling for MRI and the use of MSCs in experimental and clinical studies for the treatment of brain and spinal cord injuries.
Collapse
Affiliation(s)
- E Syková
- Department of Neuroscience, Institute of Experimental Medicine ASCR, Prague, Czech Republic.
| | | |
Collapse
|
243
|
Smirnov P, Gazeau F, Beloeil JC, Doan BT, Wilhelm C, Gillet B. Single-cell detection by gradient echo 9.4 T MRI: a parametric study. CONTRAST MEDIA & MOLECULAR IMAGING 2007; 1:165-74. [PMID: 17193693 DOI: 10.1002/cmmi.104] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recent studies have shown that cell migration can be monitored in vivo by magnetic resonance imaging after intracellular contrast agent incorporation. This is due to the dephasing effect on proton magnetization of the local magnetic field created by a labelled cell. Anionic iron oxide nanoparticles (AMNP) are among the most efficient and non-toxic contrast agents to be spontaneously taken up by a wide variety of cells. Here we measured the iron load and magnetization of HeLa tumour cells labelled with AMNP, as a function of the external magnetic field. High-resolution gradient echo 9.4 T MRI detected individual labelled cells, whereas spin echo sequences were poorly sensitive. We then conducted a systematic study in order to determine the gradient echo sequence parameters (echo time, cell magnetization and resolution) most suitable for in vivo identification of single cells.
Collapse
Affiliation(s)
- P Smirnov
- Laboratoire Matière et Systèmes Complexes, Université Paris 7, Denis Diderot and CNRS UMR 7057, 140, rue de Lourmel, 75015 Paris, France.
| | | | | | | | | | | |
Collapse
|
244
|
Walczak P, Ruiz-Cabello J, Kedziorek DA, Gilad AA, Lin S, Barnett B, Qin L, Levitsky H, Bulte JWM. Magnetoelectroporation: improved labeling of neural stem cells and leukocytes for cellular magnetic resonance imaging using a single FDA-approved agent. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2007; 2:89-94. [PMID: 17292120 DOI: 10.1016/j.nano.2006.01.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Accepted: 01/15/2006] [Indexed: 10/24/2022]
Abstract
Cellular magnetic resonance imaging (MRI) relies on the use of intracellular contrast agents, primarily iron oxide compounds. Several techniques have been used to efficiently shuttle iron oxides into nonphagocytic cells, but all methods used until now require a prolonged incubation of cells. We hypothesized that instant magnetic labeling of cells could be achieved using electroporation. Neural stem cells (NSCs) and leukocytes from spleen and lymph nodes were suspended in a ferumoxide labeling solution, loaded into cuvettes, and subjected to electromechanical permeabilization using electroporation. Magnetically labeled cells were assayed for labeling efficiency, as well as for potential toxicity or altered function. To confirm the method's applicability to detect cells, MRI experiments were performed at 11.7 T. Magnetoelectroporation of NSCs, as demonstrated by Prussian blue staining, anti-dextran immunostaining, and a quantitative iron uptake assay, proved to be an efficient intracellular magnetic labeling method. Leukocytes including lymphocytes, which are notoriously difficult to label because of their membrane properties and small cytoplasmic volume, also demonstrated a pronounced uptake of ferumoxide. MRI experiments showed that labeled NSCs could be visualized as single cells and cell clusters in gelatin phantoms, and as proliferating cell masses in mouse brain. We have developed a convenient technique for instant magnetic labeling of cells. Because magnetoelectroporation allows the use of ferumoxides approved by the US Food and Drug Administration without additional agents, it has excellent potential for clinical translation.
Collapse
Affiliation(s)
- Piotr Walczak
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Berger C, Gremlich HU, Schmidt P, Cannet C, Kneuer R, Hiestand P, Rausch M, Rudin M. In vivo monitoring the fate of Cy5.5-Tat labeled T lymphocytes by quantitative near-infrared fluorescence imaging during acute brain inflammation in a rat model of experimental autoimmune encephalomyelitis. J Immunol Methods 2007; 323:65-77. [PMID: 17433359 DOI: 10.1016/j.jim.2007.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Revised: 02/09/2007] [Accepted: 02/21/2007] [Indexed: 11/28/2022]
Abstract
T cells and macrophages directed against myelin proteins orchestrate the inflammation process in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). So far, assessment of macrophages infiltration or structural alterations has been achieved by in vivo imaging. In this work, we show the infiltration of Cy5.5-labeled T lymphocytes into the brains of EAE rats by reflectance near-infrared fluorescence imaging. T lymphocytes were labeled with Cy5.5-Tat and administered intravenously to naïve or EAE animals. The highest fluorescence signal was observed for EAE animals, which received myelin-activated T cells during the acute phase of the disease. The temporal profile of fluorescence in this group paralleled the pattern of neurological impairment during the acute phase, the remittance and first relapses of EAE. No disease specific fluorescence pattern was observed for EAE animals, which received naïve T cells. However, uptake of Cy5.5-Tat by scavenger cells (e.g. macrophages) following death of labeled T cells in vivo prevents prolonged longitudinal studies. Our work demonstrates that Cy5.5-Tat labeling of T cells is suitable for in vivo fluorescence imaging of inflammation initiation in the EAE model. This approach may particularly be useful for evaluation of novel anti-inflammatory therapies.
Collapse
Affiliation(s)
- Cedric Berger
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
246
|
Abstract
The efficacy of therapies based on neural stem cells (NSC) has been demonstrated in preclinical models of several central nervous system (CNS) diseases. Before any potential human application of such promising therapies can be envisaged, there are some important issues that need to be solved. The most relevant one is the requirement for a noninvasive technique capable of monitoring NSC delivery, homing to target sites and trafficking. Knowledge of the location and temporospatial migration of either transplanted or genetically modified NSC is of the utmost importance in analyzing mechanisms of correction and cell distribution. Further, such a technique may represent a crucial step toward clinical application of NSC-based approaches in humans, for both designing successful protocols and monitoring their outcome. Among the diverse imaging approaches available for noninvasive cell tracking, such as nuclear medicine techniques, fluorescence and bioluminescence, magnetic resonance imaging (MRI) has unique advantages. Its high temporospatial resolution, high sensitivity and specificity render MRI one of the most promising imaging modalities available, since it allows dynamic visualization of migration of transplanted cells in animal models and patients during clinically useful time periods. Different cellular and molecular labeling approaches for MRI depiction of NSC are described and discussed in this review, as well as the most relevant issues to be considered in optimizing molecular imaging techniques for clinical application.
Collapse
Affiliation(s)
- Letterio S Politi
- Neuroradiology Department, San Raffaele Scientific Institute, Via Olgettina 60, Milano, Italy.
| |
Collapse
|
247
|
Rice C, Scolding N. Strategies for achieving and monitoring myelin repair. J Neurol 2007; 254:275-83. [PMID: 17345032 DOI: 10.1007/s00415-006-0455-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Accepted: 11/03/2006] [Indexed: 11/26/2022]
Abstract
A number of factors more or less unique to multiple sclerosis have suggested that this disease may be particularly amenable to cell-based reparative therapies. The relatively focussed damage to oligodendrocytes and myelin at least in early disease implies that only a single population of cells need be replaced-and that the daunting problem of re-establishing connectivity does not apply. The presence of significant though partial spontaneous myelin repair in multiple sclerosis proves there to be no insurmountable barrier to remyelination intrinsic to the CNS: the therapeutic challenge becomes that of supplementing this spontaneous process, rather than creating repair de novo. Finally, the large body of available knowledge concerning the biology of oligodendrocytes, and the success of experimental myelin repair, have allowed cautious optimism that future prospects for such therapies are not unrealistic. Nonetheless, particular and significant problems are not hard to list: the occurrence of innumerable lesions scattered throughout the CNS, axon loss, astrocytosis, and a continuing inflammatory process, to name but a few. Here we review the progress and the areas where difficulties have yet to be resolved in efforts to develop remyelinating therapies for multiple sclerosis.
Collapse
Affiliation(s)
- Claire Rice
- Department of Neurology, University of Bristol, Institute of Clinical Neurosciences, Frenchay Hospital, Bristol, BS16 1LE, UK
| | | |
Collapse
|
248
|
Dietrich J, Kempermann G. Role of Endogenous Neural Stem Cells in Neurological Disease and Brain Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:191-220. [PMID: 16955712 DOI: 10.1007/0-387-30128-3_12] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
These examples show that stem-cell-based therapy of neuro-psychiatric disorders will not follow a single scheme, but rather include widely different approaches. This is in accordance with the notion that the impact of stem cell biology on neurology will be fundamental, providing a shift in perspective, rather than introducing just one novel therapeutic tool. Stem cell biology, much like genomics and proteomics, offers a "view from within" with an emphasis on a theoretical or real potential and thereby the inherent openness, which is central to the concept of stem cells. Thus, stem cell biology influences many other, more traditional therapeutic approaches, rather than introducing one distinct novel form of therapy. Substantial advances have been made i n neural stemcell research during the years. With the identification of stem and progenitor cells in the adult brain and the complex interaction of different stem cell compartments in the CNS--both, under physiological and pathological conditions--new questions arise: What is the lineage relationship between t he different progenitor cells in the CNS and how much lineage plasticity exists? What are the signals controlling proliferation and differentiation of neural stem cells and can these be utilized to allow repair of the CNS? Insights in these questions will help to better understand the role of stem cells during development and aging and the possible relation of impaired or disrupted stem cell function and their impact on both the development and treatment of neurological disease. A number o f studies have indicated a limited neuronal and glial regeneration certain pathological conditions. These fundamental observations have already changed our view on understanding neurological disease and the brain's capacity for endogenous repair. The following years will have to show how we can influence andmodulate endogenous repair nisms by increasing the cellular plasticity in the young and aged CNS.
Collapse
Affiliation(s)
- Jörg Dietrich
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | |
Collapse
|
249
|
Ben-Hur T, van Heeswijk RB, Einstein O, Aharonowiz M, Xue R, Frost EE, Mori S, Reubinoff BE, Bulte JWM. Serial in vivo MR tracking of magnetically labeled neural spheres transplanted in chronic EAE mice. Magn Reson Med 2007; 57:164-71. [PMID: 17191231 DOI: 10.1002/mrm.21116] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Neural stem cell (NSC) transplantation has been shown to attenuate the severity of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Central to the future success of NSC transplantation in MS is the ability of transplanted cells to migrate from the site of transplantation to relevant foci of disease. Using magnetically labeled mouse neurospheres and human embryonic stem cell (hESC)-derived neurospheres, we applied serial magnetic resonance imaging (MRI) to assess the biodynamics of transplanted cell migration in a chronic mouse EAE model. Magnetic labeling did not affect the in vitro and in vivo characteristics of cells as multipotential precursors. Cell migration occurred along white matter (WM) tracts (especially the corpus callosum (CC), fimbria, and internal capsule), predominantly early in the acute phase of disease, and in an asymmetric manner. The distance of cell migration correlated well with clinical severity of disease and the number of microglia in the WM tracts, supporting the notion that inflammatory signals promote transplanted cell migration. This study shows for the first time that hESC-derived neural precursors also respond to tissue signals in an MS model, similarly to rodent cells. The results are directly relevant for designing and optimizing cell therapies for MS, and achieving a better understanding of in vivo cell dynamics and cell-tissue interactions.
Collapse
MESH Headings
- Animals
- Cell Lineage
- Cell Movement
- Cells, Cultured
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Humans
- Magnetic Resonance Imaging
- Mice
- Mice, Inbred C57BL
- Neurons/cytology
- Neurons/physiology
- Neurons/transplantation
- Severity of Illness Index
- Spheroids, Cellular/cytology
- Spheroids, Cellular/transplantation
- Stem Cell Transplantation
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Tamir Ben-Hur
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Abstract
Nanotechnology is a scientific movement that has the potential to transform the diagnosis and treatment of disease in the 21st century. The area of investigation is defined by the study, design, manipulation, manufacture, and control of materials or devices by physical or chemical means at resolutions on the order of one billionth of a meter. The potential for a wide range of clinical applications makes a basic understanding of nanotechnology important to physiatrists. This review presents an introduction to nanotechnology and discusses key developments in tissue engineering, drug delivery, imaging, diagnostics, surface texturing, and biointerfaces that could impact the practice of physiatry in the future.
Collapse
Affiliation(s)
- Assaf T Gordon
- Department of Physical Medicine and Rehabilitation, New York University, Rusk Institute, New York, New York, USA.
| | | | | | | |
Collapse
|