1
|
Garello F, Svenskaya Y, Parakhonskiy B, Filippi M. Micro/Nanosystems for Magnetic Targeted Delivery of Bioagents. Pharmaceutics 2022; 14:pharmaceutics14061132. [PMID: 35745705 PMCID: PMC9230665 DOI: 10.3390/pharmaceutics14061132] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 01/09/2023] Open
Abstract
Targeted delivery of pharmaceuticals is promising for efficient disease treatment and reduction in adverse effects. Nano or microstructured magnetic materials with strong magnetic momentum can be noninvasively controlled via magnetic forces within living beings. These magnetic carriers open perspectives in controlling the delivery of different types of bioagents in humans, including small molecules, nucleic acids, and cells. In the present review, we describe different types of magnetic carriers that can serve as drug delivery platforms, and we show different ways to apply them to magnetic targeted delivery of bioagents. We discuss the magnetic guidance of nano/microsystems or labeled cells upon injection into the systemic circulation or in the tissue; we then highlight emergent applications in tissue engineering, and finally, we show how magnetic targeting can integrate with imaging technologies that serve to assist drug delivery.
Collapse
Affiliation(s)
- Francesca Garello
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy;
| | - Yulia Svenskaya
- Science Medical Center, Saratov State University, 410012 Saratov, Russia;
| | - Bogdan Parakhonskiy
- Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium;
| | - Miriam Filippi
- Soft Robotics Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
- Correspondence:
| |
Collapse
|
2
|
Wang C, Adams SR, Xu H, Zhu W, Ahrens ET. β‑Diketonate-Iron(III) Complex: A Versatile Fluorine-19 MRI Signal Enhancement Agent. ACS APPLIED BIO MATERIALS 2019; 2:3836-3842. [PMID: 33981964 DOI: 10.1021/acsabm.9b00455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fluorine-19 magnetic resonance imaging (MRI) has gained considerable momentum as a promising imaging modality for in vivo tracking of cellular therapies and as a diagnostic for inflammatory disease. To further the utility of this technique, we increase imaging probe sensitivity by merging paramagnetic metal chelates with aqueous perfluorocarbon (PFC) nanoemulsions. We prepared a highly fluorinated ferric tris(β-diketonate) chelate (MW = 1265.2 g/mol) at gram scale. This iron chelate is soluble in multiple PFC oils used for MRI and readily reduces the 19F longitudinal relaxation time (T 1) to <100 ms with modest line broadening and displays superior properties for 19F MRI applications. The sensitivity enhancement by Fe(III) laden PFC nanoemulsion was confirmed in MRI phantom studies, where reduced T 1 speeds data acquisition thereby increasing the 19F image sensitivity per time via signal averaging. Additionally, 19F relaxivity of nanoemulsions incorporating other metal ions, including Gd, Er, Ho, Dy, Mn, Cr, and Ni, were evaluated. High-moment lanthanide ions, such as Gd(III), display severe line broadening, but other ions [e.g., Ho(III)] induce pseudocontact chemical shifts (up to 0.5 ppm) of 19F in nanoemulsion, which makes them potentially useful for multichromatic 19F imaging. Formulated nanoemulsions have a shelf life >200 days. Free β-diketonate or its iron complex in formed PFC nanoemulsion did not induce cytotoxicity in intracellularly labeled macrophages. Overall, ferric tris(β-diketonate) chelate provides a scalable approach for boosting sensitivity of PFC-based 19F MRI probes. More generally, it can functionalize PFC oil, whose chemical modification remains challenging.
Collapse
Affiliation(s)
- Chao Wang
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| | - Stephen R Adams
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093, United States
| | - Hongyan Xu
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| | - Wenlian Zhu
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| | - Eric T Ahrens
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
3
|
Hsu FT, Wei ZH, Hsuan YCY, Lin W, Su YC, Liao CH, Hsieh CL. MRI tracking of polyethylene glycol-coated superparamagnetic iron oxide-labelled placenta-derived mesenchymal stem cells toward glioblastoma stem-like cells in a mouse model. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S448-S459. [PMID: 30198338 DOI: 10.1080/21691401.2018.1499661] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Mesenchymal stem cells (MSCs) that display homing and infiltration properties towards tumor cells are a promising cellular targeting vector for brain tumor therapy but are limited to local-regional delivery in current preclinical models. Here, we investigated whether placenta-derived MSCs (P-MSCs) are a superior cellular vector for systemic targeting of glioblastoma stem-like cells (GSCs), with an imaging modality to real-time monitor the trafficking P-MSCs to glioblastoma sites. Results demonstrated that P-MSCs had greater migratory activity towards GSCs and across blood-brain barrier compared with bone marrow-derived MSCs, and this activity was enhanced by hypoxia precondition. Chemokine ligand 5 was identified as a chemoattractant responsible for the glioblastoma tropism of P-MSCs. Polyethylene glycol-coated superparamagnetic iron oxide (PEG-SPIO) was synthesized for cellular labelling and imaging P-MSCs, displaying high cellular uptake and no cytotoxic effect on P-MSCs cell proliferation or stemness property. The homing effects of intravenously administered PEG-SPIO-labelled P-MSCs towards intracerebral GSCs were able to be detected in mice models through T2-weighted magnetic resonance imaging (MRI). This study suggests the possibility of innovative systemic P-MSC-based cell therapy for aggressive GSCs, developing a state-of-the-art theranostic technique for real-time tracking of therapeutic P-MSCs tumor infiltration through cellular MRI.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- a Department of Radiology , School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan.,b Department of Biological Science and Technology , China Medical University , Taichung , Taiwan.,c Department of Medical Imaging , Taipei Medical University Hospital , Taipei , Taiwan.,d Research Center of Translational Imaging , College of Medicine, Taipei Medical University , Taipei , Taiwan
| | - Zung-Hang Wei
- e Department of Power Mechanical Engineering , National Tsing Hua University , Hsinchu , Taiwan
| | | | - Willie Lin
- f Meridigen Biotech Co., Ltd. , Neihu, Taipei City , Taiwan
| | - Yu-Chin Su
- f Meridigen Biotech Co., Ltd. , Neihu, Taipei City , Taiwan
| | - Chia-Hui Liao
- g The PhD Program for Translational Medicine , College of Medical Science and Technology, Taipei Medical University , Taipei , Taiwan
| | - Chia-Ling Hsieh
- g The PhD Program for Translational Medicine , College of Medical Science and Technology, Taipei Medical University , Taipei , Taiwan.,h Clinical Research Center , Taipei Medical University Hospital, Taipei Medical University , Taipei , Taiwan.,i TMU Research Center of Cancer Translational Medicine , Taipei Medical University , Taipei , Taiwan
| |
Collapse
|
4
|
Lai WF, Rogach AL, Wong WT. Chemistry and engineering of cyclodextrins for molecular imaging. Chem Soc Rev 2018; 46:6379-6419. [PMID: 28930330 DOI: 10.1039/c7cs00040e] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cyclodextrins (CDs) are naturally occurring cyclic oligosaccharides bearing a basket-shaped topology with an "inner-outer" amphiphilic character. The abundance of hydroxyl groups enables CDs to be functionalized with multiple targeting ligands and imaging elements. The imaging time, and the payload of different imaging elements, can be tuned by taking advantage of the commercial availability of CDs with different sizes of the cavity. This review aims to offer an outlook of the chemistry and engineering of CDs for the development of molecular probes. Complexation thermodynamics of CDs, and the corresponding implications for probe design, are also presented with examples demonstrating the structural and physiochemical roles played by CDs in the full ambit of molecular imaging. We hope that this review not only offers a synopsis of the current development of CD-based molecular probes, but can also facilitate translation of the incremental advancements from the laboratory to real biomedical applications by illuminating opportunities and challenges for future research.
Collapse
Affiliation(s)
- Wing-Fu Lai
- School of Pharmaceutical Sciences, Health Science Centre, Shenzhen University, Shenzhen, China.
| | | | | |
Collapse
|
5
|
Hu S, Zhou Y, Zhao Y, Xu Y, Zhang F, Gu N, Ma J, Reynolds MA, Xia Y, Xu HH. Enhanced bone regeneration and visual monitoring via superparamagnetic iron oxide nanoparticle scaffold in rats. J Tissue Eng Regen Med 2018; 12:e2085-e2098. [DOI: 10.1002/term.2641] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 11/28/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Shuying Hu
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical University Nanjing P. R. China
| | - Yi Zhou
- Yixing People's Hospital Yixing P. R. China
| | - Yantao Zhao
- Beijing Engineering Research Center of Orthopaedic ImplantsFirst Affiliated Hospital of CPLA General Hospital Beijing P. R. China
| | - Yang Xu
- Affiliated Stomatology Hospital of Soochow University Suzhou P. R. China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical University Nanjing P. R. China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical EngineeringSoutheast University Nanjing P. R. China
- Suzhou Institute & Collaborative Innovation Center of Suzhou Nano Science and TechnologySoutheast University Suzhou P. R. China
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical University Nanjing P. R. China
| | - Mark A. Reynolds
- Department of Advanced Oral Sciences & TherapeuticsUniversity of Maryland School of Dentistry Baltimore MD USA
| | - Yang Xia
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical University Nanjing P. R. China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical EngineeringSoutheast University Nanjing P. R. China
- Department of Advanced Oral Sciences & TherapeuticsUniversity of Maryland School of Dentistry Baltimore MD USA
| | - Hockin H.K. Xu
- Department of Advanced Oral Sciences & TherapeuticsUniversity of Maryland School of Dentistry Baltimore MD USA
- Center for Stem Cell Biology & Regenerative MedicineUniversity of Maryland School of Medicine Baltimore MD USA
- Department of Mechanical EngineeringUniversity of Maryland Baltimore County Baltimore County MD USA
| |
Collapse
|
6
|
Yoon HY, Jeon S, You DG, Park JH, Kwon IC, Koo H, Kim K. Inorganic Nanoparticles for Image-Guided Therapy. Bioconjug Chem 2016; 28:124-134. [DOI: 10.1021/acs.bioconjchem.6b00512] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hong Yeol Yoon
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sangmin Jeon
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- School
of Chemical Engineering, Sungkyunkwan University, 2066, Seobu-ro,
Jangan-gu, Suwon 16419, Republic of Korea
| | - Dong Gil You
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- School
of Chemical Engineering, Sungkyunkwan University, 2066, Seobu-ro,
Jangan-gu, Suwon 16419, Republic of Korea
| | - Jae Hyung Park
- School
of Chemical Engineering, Sungkyunkwan University, 2066, Seobu-ro,
Jangan-gu, Suwon 16419, Republic of Korea
| | - Ick Chan Kwon
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST
Graduate School of Converging Science and Technology, Korea University, 145
Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Heebeom Koo
- Department
of Medical Lifescience, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Kwangmeyung Kim
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
7
|
Ariza de Schellenberger A, Kratz H, Farr TD, Löwa N, Hauptmann R, Wagner S, Taupitz M, Schnorr J, Schellenberger EA. Labeling of mesenchymal stem cells for MRI with single-cell sensitivity. Int J Nanomedicine 2016; 11:1517-35. [PMID: 27110112 PMCID: PMC4835118 DOI: 10.2147/ijn.s101141] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sensitive cell detection by magnetic resonance imaging (MRI) is an important tool for the development of cell therapies. However, clinically approved contrast agents that allow single-cell detection are currently not available. Therefore, we compared very small iron oxide nanoparticles (VSOP) and new multicore carboxymethyl dextran-coated iron oxide nanoparticles (multicore particles, MCP) designed by our department for magnetic particle imaging (MPI) with discontinued Resovist® regarding their suitability for detection of single mesenchymal stem cells (MSC) by MRI. We achieved an average intracellular nanoparticle (NP) load of >10 pg Fe per cell without the use of transfection agents. NP loading did not lead to significantly different results in proliferation, colony formation, and multilineage in vitro differentiation assays in comparison to controls. MRI allowed single-cell detection using VSOP, MCP, and Resovist® in conjunction with high-resolution T2*-weighted imaging at 7 T with postprocessing of phase images in agarose cell phantoms and in vivo after delivery of 2,000 NP-labeled MSC into mouse brains via the left carotid artery. With optimized labeling conditions, a detection rate of ~45% was achieved; however, the experiments were limited by nonhomogeneous NP loading of the MSC population. Attempts should be made to achieve better cell separation for homogeneous NP loading and to thus improve NP-uptake-dependent biocompatibility studies and cell detection by MRI and future MPI. Additionally, using a 7 T MR imager equipped with a cryocoil resulted in approximately two times higher detection. In conclusion, we established labeling conditions for new high-relaxivity MCP, VSOP, and Resovist® for improved MRI of MSC with single-cell sensitivity.
Collapse
Affiliation(s)
| | - Harald Kratz
- Department of Radiology, Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tracy D Farr
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany; School of Life Sciences, University of Nottingham, Medical School, Nottingham, UK
| | - Norbert Löwa
- Department of Biomagnetic Signals, Physikalisch-Technische Bundesanstalt Berlin, Berlin, Germany
| | - Ralf Hauptmann
- Department of Radiology, Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne Wagner
- Department of Radiology, Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Taupitz
- Department of Radiology, Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg Schnorr
- Department of Radiology, Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eyk A Schellenberger
- Department of Radiology, Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Sharifi S, Seyednejad H, Laurent S, Atyabi F, Saei AA, Mahmoudi M. Superparamagnetic iron oxide nanoparticles for in vivo molecular and cellular imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 10:329-55. [PMID: 25882768 DOI: 10.1002/cmmi.1638] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 01/30/2015] [Accepted: 02/06/2015] [Indexed: 12/16/2022]
Abstract
In the last decade, the biomedical applications of nanoparticles (NPs) (e.g. cell tracking, biosensing, magnetic resonance imaging (MRI), targeted drug delivery, and tissue engineering) have been increasingly developed. Among the various NP types, superparamagnetic iron oxide NPs (SPIONs) have attracted considerable attention for early detection of diseases due to their specific physicochemical properties and their molecular imaging capabilities. A comprehensive review is presented on the recent advances in the development of in vitro and in vivo SPION applications for molecular imaging, along with opportunities and challenges.
Collapse
Affiliation(s)
- Shahriar Sharifi
- Department of Biomaterials Science and Technology, University of Twente, The Netherlands
| | - Hajar Seyednejad
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA
| | - Sophie Laurent
- Department of General, Organic, and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, B-7000, Mons, Belgium.,CMMI - Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland 8, B-6041, Gosselies, Belgium
| | - Fatemeh Atyabi
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ata Saei
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Morteza Mahmoudi
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
9
|
Xiang L, Chen Y. Stem cell transplantation for treating spinal cord injury: A literature comparison between studies of stem cells obtained from various sources. Neural Regen Res 2015; 7:1256-63. [PMID: 25709624 PMCID: PMC4336961 DOI: 10.3969/j.issn.1673-5374.2012.16.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 04/23/2012] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE: To identify global research trends of stem cell transplantation for treating spinal cord injury using a bibliometric analysis of the Web of Science. DATA RETRIEVAL: We performed a bibliometric analysis of data retrievals for stem cell transplantation for treating spinal cord injury from 2002 to 2011 using the Web of Science. SELECTION CRITERIA: Inclusion criteria: (a) peer-reviewed articles on stem cell transplantation for treating spinal cord injury that were published and indexed in the Web of Science; (b) type of articles: original research articles, reviews, meeting abstracts, proceedings papers, book chapters, editorial material, and news items; and (c) year of publication: 2002–2011. Exclusion criteria: (a) articles that required manual searching or telephone access; (b) documents that were not published in the public domain; and (c) a number of corrected papers from the total number of articles. MAIN OUTCOME MEASURES: (1) Annual publication output; (2) distribution according to country; (3) distribution according to institution; (4) distribution according to journals; (5) distribution according to funding agencies; and (6) top cited articles over the last 10 years. RESULTS: Bone marrow mesenchymal stem cells and embryonic stem cells have been widely used for treating spinal cord injury. In total, 191 studies of bone marrow mesenchymal stem cell transplantation and 236 studies of embryonic stem cell transplantation for treating spinal cord injury appeared in the Web of Science from 2002 to 2011, and almost half of which were derived from American or Japanese authors and institutes. The number of studies of stem cell transplantation for treating spinal cord injury has gradually increased over the past 10 years. Most papers on stem cell transplantation for treating spinal cord injury appeared in journals with a particular focus on stem cell research, such as Stem Cells and Cell Transplantation. Although umbilical cord blood stem cells and adipose-derived stem cells have been studied for treating spinal cord injury, the number of published papers was much smaller, with only 21 and 17 records, respectively, in the Web of Science. CONCLUSION: Based on our analysis of the literature and research trends, we found that stem cells transplantation obtained from various sources have been studied for treating spinal cord injury; however, it is difficult for researchers to reach a consensus on this theme.
Collapse
Affiliation(s)
- Liangbi Xiang
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang 110016, Liaoning Province, China
| | - Yu Chen
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang 110016, Liaoning Province, China
| |
Collapse
|
10
|
|
11
|
Naumova AV, Modo M, Moore A, Murry CE, Frank JA. Clinical imaging in regenerative medicine. Nat Biotechnol 2014; 32:804-18. [PMID: 25093889 PMCID: PMC4164232 DOI: 10.1038/nbt.2993] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/15/2014] [Indexed: 01/09/2023]
Abstract
In regenerative medicine, clinical imaging is indispensable for characterizing damaged tissue and for measuring the safety and efficacy of therapy. However, the ability to track the fate and function of transplanted cells with current technologies is limited. Exogenous contrast labels such as nanoparticles give a strong signal in the short term but are unreliable long term. Genetically encoded labels are good both short- and long-term in animals, but in the human setting they raise regulatory issues related to the safety of genomic integration and potential immunogenicity of reporter proteins. Imaging studies in brain, heart and islets share a common set of challenges, including developing novel labeling approaches to improve detection thresholds and early delineation of toxicity and function. Key areas for future research include addressing safety concerns associated with genetic labels and developing methods to follow cell survival, differentiation and integration with host tissue. Imaging may bridge the gap between cell therapies and health outcomes by elucidating mechanisms of action through longitudinal monitoring.
Collapse
Affiliation(s)
- Anna V Naumova
- 1] Department of Radiology, University of Washington, Seattle, Washington, USA. [2] Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA. [3] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Michel Modo
- 1] McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [2] Centre for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [3] Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [4] Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anna Moore
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Charles E Murry
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA. [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA. [3] Department of Pathology, University of Washington, Seattle, Washington, USA. [4] Department of Bioengineering, University of Washington, Seattle, Washington, USA. [5] Department of Medicine/Cardiology, University of Washington, Seattle, Washington, USA
| | - Joseph A Frank
- 1] Radiology and Imaging Sciences, Clinical, National Institutes of Health, Bethesda, Maryland, USA. [2] National Institutes of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
12
|
Grześkowiak BF, Sánchez-Antequera Y, Hammerschmid E, Döblinger M, Eberbeck D, Woźniak A, Słomski R, Plank C, Mykhaylyk O. Nanomagnetic Activation as a Way to Control the Efficacy of Nucleic Acid Delivery. Pharm Res 2014; 32:103-21. [DOI: 10.1007/s11095-014-1448-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/02/2014] [Indexed: 01/01/2023]
|
13
|
Eggleston H, Panizzi P. Molecular imaging of bacterial infections in vivo: the discrimination of infection from inflammation. INFORMATICS (MDPI) 2014; 1:72-99. [PMID: 26985401 PMCID: PMC4790455 DOI: 10.3390/informatics1010072] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Molecular imaging by definition is the visualization of molecular and cellular processes within a given system. The modalities and reagents described here represent a diverse array spanning both pre-clinical and clinical applications. Innovations in probe design and technologies would greatly benefit therapeutic outcomes by enhancing diagnostic accuracy and assessment of acute therapy. Opportunistic pathogens continue to pose a worldwide threat, despite advancements in treatment strategies, which highlights the continued need for improved diagnostics. In this review, we present a summary of the current clinical protocol for the imaging of a suspected infection, methods currently in development to optimize this imaging process, and finally, insight into endocarditis as a model of infectious disease in immediate need of improved diagnostic methods.
Collapse
Affiliation(s)
- Heather Eggleston
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849
| | - Peter Panizzi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849
| |
Collapse
|
14
|
Kolosnjaj-Tabi J, Wilhelm C, Clément O, Gazeau F. Cell labeling with magnetic nanoparticles: opportunity for magnetic cell imaging and cell manipulation. J Nanobiotechnology 2013; 11 Suppl 1:S7. [PMID: 24564857 PMCID: PMC4029272 DOI: 10.1186/1477-3155-11-s1-s7] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This tutorial describes a method of controlled cell labeling with citrate-coated ultra small superparamagnetic iron oxide nanoparticles. This method may provide basically all kinds of cells with sufficient magnetization to allow cell detection by high-resolution magnetic resonance imaging (MRI) and to enable potential magnetic manipulation. In order to efficiently exploit labeled cells, quantify the magnetic load and deliver or follow-up magnetic cells, we herein describe the main requirements that should be applied during the labeling procedure. Moreover we present some recommendations for cell detection and quantification by MRI and detail magnetic guiding on some real-case studies in vitro and in vivo.
Collapse
|
15
|
Fayol D, Le Visage C, Ino J, Gazeau F, Letourneur D, Wilhelm C. Design of Biomimetic Vascular Grafts with Magnetic Endothelial Patterning. Cell Transplant 2013; 22:2105-18. [DOI: 10.3727/096368912x661300] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The development of small diameter vascular grafts with a controlled pluricellular organization is still needed for effective vascular tissue engineering. Here, we describe a technological approach combining a tubular scaffold and magnetically labeled cells to create a pluricellular and organized vascular graft, the endothelialization of which could be monitored by MRI prior to transplantation. A novel type of scaffold was developed with a tubular geometry and a porous bulk structure enabling the seeding of cells in the scaffold pores. A homogeneous distribution of human mesenchymal stem cells in the macroporous structure was obtained by seeding the freeze-dried scaffold with the cell suspension. The efficient covering of the luminal surface of the tube was then made possible thanks to the implementation of a magnetic-based patterning technique. Human endothelial cells or endothelial progenitors were magnetically labeled with iron oxide nanoparticles and successfully attracted to the 2-mm lumen where they attached and formed a continuous endothelium. The combination of imaging modalities [fluorescence imaging, histology, and 3D magnetic resonance imaging (MRI)] evidenced the integrity of the vascular construct. In particular, the observation of different cell organizations in a vascular scaffold within the range of resolution of single cells by 4.7 T MRI is reported.
Collapse
Affiliation(s)
- Delphine Fayol
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS and Université Paris Diderot, Paris, France
| | - Catherine Le Visage
- Inserm, U698, Bio-ingénierie Cardiovasculaire, Université Paris Diderot, CHU X. Bichat, Paris, France
| | - Julia Ino
- Inserm, U698, Bio-ingénierie Cardiovasculaire, Université Paris Diderot, CHU X. Bichat, Paris, France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS and Université Paris Diderot, Paris, France
| | - Didier Letourneur
- Inserm, U698, Bio-ingénierie Cardiovasculaire, Université Paris Diderot, CHU X. Bichat, Paris, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS and Université Paris Diderot, Paris, France
| |
Collapse
|
16
|
Martinez C, Henao A, Rodriguez JE, Padgett KR, Ramaswamy S. Monitoring Steady Flow Effects on Cell Distribution in Engineered Valve Tissues by Magnetic Resonance Imaging. Mol Imaging 2013. [DOI: 10.2310/7290.2013.00063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Catalina Martinez
- From the Tissue Engineering Mechanics, Imaging and Materials Laboratory, Department of Biomedical Engineering, College of Engineering and Computing, Florida International University, and Interdisciplinary Stem Cell Institute and Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL
| | - Angela Henao
- From the Tissue Engineering Mechanics, Imaging and Materials Laboratory, Department of Biomedical Engineering, College of Engineering and Computing, Florida International University, and Interdisciplinary Stem Cell Institute and Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL
| | - Jose E. Rodriguez
- From the Tissue Engineering Mechanics, Imaging and Materials Laboratory, Department of Biomedical Engineering, College of Engineering and Computing, Florida International University, and Interdisciplinary Stem Cell Institute and Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL
| | - Kyle R. Padgett
- From the Tissue Engineering Mechanics, Imaging and Materials Laboratory, Department of Biomedical Engineering, College of Engineering and Computing, Florida International University, and Interdisciplinary Stem Cell Institute and Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL
| | - Sharan Ramaswamy
- From the Tissue Engineering Mechanics, Imaging and Materials Laboratory, Department of Biomedical Engineering, College of Engineering and Computing, Florida International University, and Interdisciplinary Stem Cell Institute and Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
17
|
Varma NRS, Shankar A, Iskander A, Janic B, Borin TF, Ali MM, Arbab AS. Differential biodistribution of intravenously administered endothelial progenitor and cytotoxic T-cells in rat bearing orthotopic human glioma. BMC Med Imaging 2013; 13:17. [PMID: 23758888 PMCID: PMC3706264 DOI: 10.1186/1471-2342-13-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/29/2013] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND A major challenge in the development of cell based therapies for glioma is to deliver optimal number of cells (therapeutic dose) to the tumor. Imaging tools such as magnetic resonance imaging (MRI), optical imaging, positron emission tomography (PET) and single-photon emission computed tomography (SPECT) has been used in cell tracking and/or biodistribution studies. In this study, we evaluate the dynamic biodistribution of systemic injected labeled cells [human cord blood derived endothelial progenitor cells (EPCs) and cytotoxic T-cells (CTLs)] in rat glioma model with in vivo SPECT imaging. METHODS Human cord blood EPCs, T-cells and CD14⁺ cells (monocytes/dendritic cells) were isolated using the MidiMACS system. CD14⁺ cells were converted to dendritic cells (DC) and also primed with U251 tumor cell line lysate. T-cells were co-cultured with irradiated primed DCs at 10:1 ratio to make CTLs. Both EPCs and CTLs were labeled with In-111-oxine at 37°C in serum free DMEM media. Glioma bearing animals were randomly assigned into three groups. In-111 labeled cells or In-111 oxine alone were injected through tail vein and SPECT imaging was performed on day 0, 1, and 3. In-111 oxine activity in various organs and tumor area was determined. Histochemical analysis was performed to further confirm the migration and homing of injected cells at the tumor site. RESULTS EPCs and CTLs showed an In-111 labeling efficiency of 87.06 ± 7.75% and 70.8 ± 12.9% respectively. Initially cell migration was observed in lung following inravenous administration of In-111 labeled cells and decreased on day 1 and 3, which indicate re-distribution of labeled cells from lung to other organs. Relatively higher In-111 oxine activity was observed in tumor areas at 24 hours in animals received In-111 labeled cells (EPCs or CTLs). Histiological analysis revealed iron positive cells in and around the tumor area in animals that received labeled cells (CTLs and EPCs). CONCLUSION We observed differential biodistribution of In-111-oxine labeled EPCs and CTLs in different organs and intracranial glioma. This study indicates In-111 oxine based SPECT imaging is an effective tool to study the biodistribution of therapeutically important cells.
Collapse
Affiliation(s)
- Nadimpalli Ravi S Varma
- Cellular and Molecular Imaging Laboratory, Radiology, Henry Ford Hospital, Detroit, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
18
|
Noad J, Gonzalez-Lara LE, Broughton HC, McFadden C, Chen Y, Hess DA, Foster PJ. MRI tracking of transplanted iron-labeled mesenchymal stromal cells in an immune-compromised mouse model of critical limb ischemia. NMR IN BIOMEDICINE 2013; 26:458-467. [PMID: 23165968 DOI: 10.1002/nbm.2884] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 08/30/2012] [Accepted: 09/29/2012] [Indexed: 06/01/2023]
Abstract
Peripheral arterial disease is a clinical problem in which mesenchymal stromal cell (MSC) transplantation may offer substantial benefit by promoting the generation of new blood vessels and improving limb ischemia and wound healing via their potent paracrine activities. MRI allows for the noninvasive tracking of cells over time using iron oxide contrast agents to label cells before they are injected or transplanted. However, a major limitation of the tracking of iron oxide-labeled cells with MRI is the possibility that dead or dying cells will transfer the iron oxide label to local bystander macrophages, making it very difficult to distinguish between viable transplanted cells and endogenous macrophages in the images. In this study, a severely immune-compromised mouse, with limited macrophage activity, was investigated to examine cell tracking in a system in which bystander cell uptake of dead, iron-labeled cells or free iron particles was minimized. MRI was used to track the fate of MSCs over 21 days after their intramuscular transplantation in mice with a femoral artery ligation. In all mice, a region of signal loss was observed at the injection site and the volume of signal hypointensity diminished over time. Fluorescence and light microscopy showed that iron-positive MSCs persisted at the transplant site and often appeared to be integrated in perivascular niches. This was compared with MSC transplantation in immune-competent mice with femoral artery ligation. In these mice, the regions of signal loss caused by iron-labeled MSC cleared more slowly, and histology revealed iron particles trapped at the site of cell transplantation and associated with areas of inflammation.
Collapse
Affiliation(s)
- Jennifer Noad
- Robarts Research Institute, London, ON, Canada; Department of Medical Biophysics, The University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
19
|
Wilhelm C, Gazeau F. [Magnetic nanoparticles as tools for cell therapy]. Biol Aujourdhui 2013; 206:273-84. [PMID: 23419254 DOI: 10.1051/jbio/2012024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Indexed: 11/15/2022]
Abstract
Labelling living cells with magnetic nanoparticles creates opportunities for numerous biomedical applications such as Magnetic Resonance Imaging (MRI) cell tracking, cell manipulation, cell patterning for tissue engineering and magnetically-assisted cell delivery. The unique advantage of magnetic-based methods is to activate or monitor cell behavior by a remote stimulus, the magnetic field. Cell labelling methods using superparamagnetic nanoparticles have been widely developed, showing no adverse effect on cell proliferation and functionalities while conferring magnetic properties to various cell types. This paper first describes how cells can become responsive to magnetic field by safely internalizing magnetic nanoparticles. We next show how magnetic cells can be detected by MRI, giving the opportunity for non-invasive in vivo monitoring of cell migration. We exemplify the fact that MRI cell tracking has become a method of choice to follow the fate of administrated cells in cell therapy assay, whether the cells are grafted locally or administrated in the circulation. Finally we give different examples of magnetic manipulation of cells and their applications to regenerative medicine. Magnetic cell manipulation are forecasted to be more and more developed, in order to improve tissue engineering technique and assist cell-based therapies. Owing to the clinical approval of iron-oxide nanoparticles as MRI contrast agent, there is no major obstacle in the translation to human clinics of the magnetic methods summarized in this paper.
Collapse
Affiliation(s)
- Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes, CNRS – Université Paris Diderot, 75205 Paris Cedex 13, France.
| | | |
Collapse
|
20
|
Spiriev T, Sandu N, Schaller B. Molecular imaging and tracking stem cells in neurosciences. Methods Mol Biol 2013; 1052:195-201. [PMID: 23640257 DOI: 10.1007/7651_2013_27] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Stem cell transplantation is a promising new therapeutic option in different neurological diseases. However, it is not yet possible to translate its potential from animal models to clinical application. One of the main problems of applying stem cell transplantation in clinical medium is the difficulty of detection, localization, and examination of the stem cells in vivo at both cellular and molecular levels. State-of-the-art molecular imaging techniques provide new and better means for noninvasive, repeated, and quantitative tracking of stem cell implant or transplant. From initial deposition to the survival, migration, and differentiation of the transplant/implanted stem cells, current molecular imaging methods allow monitoring of the infused cells in the same live recipient over time. The present review briefly summarizes and compares these molecular imaging methods for cell labeling and imaging in animal models as well as in clinical application and sheds light on consecutive new therapeutic options if appropriate.
Collapse
Affiliation(s)
- Toma Spiriev
- Department of Neurosurgery, Tokuda Hospital Sofia, Sofia, Bulgaria
| | | | | |
Collapse
|
21
|
Dadashzadeh ER, Hobson M, Bryant LH, Dean DD, Frank JA. Rapid spectrophotometric technique for quantifying iron in cells labeled with superparamagnetic iron oxide nanoparticles: potential translation to the clinic. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:50-6. [PMID: 23109392 PMCID: PMC3490434 DOI: 10.1002/cmmi.1493] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Labeling cells with superparamagnetic iron oxide (SPIO) nanoparticles provides the ability to track cells by magnetic resonance imaging. Quantifying intracellular iron concentration in SPIO labeled cells would allow for the comparison of agents and techniques used to magnetically label cells. Here we describe a rapid spectrophotometric technique (ST) to quantify iron content of SPIO-labeled cells, circumventing the previous requirement of an overnight acid digestion. Following lysis with 10% sodium dodecyl sulfate (SDS) of magnetically labeled cells, quantification of SPIO doped or labeled cells was performed using commonly available spectrophotometric instrument(s) by comparing absorptions at 370 and 750 nm with correction for turbidity of cellular products to determine the iron content of each sample. Standard curves demonstrated high linear correlation (R(2) = 0.998) between absorbance spectra of iron oxide nanoparticles and concentration in known SPIO-doped cells. Comparisons of the ST with inductively coupled plasma-mass spectroscopy (ICP-MS) or nuclear magnetic resonance relaxometric (R(2)) determinations of intracellular iron contents in SPIO containing samples resulted in significant linear correlation between the techniques (R(2) vs ST, R(2) > 0.992, p < 0.0001; ST vs ICP-MS, R(2) > 0.995, p < 0.0001) with the limit of detection of ST for iron = 0.66 µg ml(-1) for 10(6) cells ml(-1). We have developed a rapid straightforward protocol that does not require overnight acid digestion for quantifying iron oxide content in magnetically labeled cells using readily available analytic instrumentation that should greatly expedite advances in comparing SPIO agents and protocols for labeling cells.
Collapse
Affiliation(s)
- Esmaeel R. Dadashzadeh
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
- Howard Hughes Medical Scholar, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Matthew Hobson
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - L. Henry Bryant
- Laboratory of Diagnostic Radiology Research, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Dana D. Dean
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Joseph A. Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
- Laboratory of Diagnostic Radiology Research, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
- Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
22
|
Ghosh SK, Uchida M, Yoo B, Ross AW, Gendler SJ, Gong J, Moore A, Medarova Z. Targeted imaging of breast tumor progression and therapeutic response in a human uMUC-1 expressing transgenic mouse model. Int J Cancer 2012; 132:1860-7. [PMID: 23015160 DOI: 10.1002/ijc.27872] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 09/10/2012] [Indexed: 01/12/2023]
Abstract
The ability to monitor breast cancer initiation and progression on the molecular level would provide an effective tool for early diagnosis and therapy. In the present study, we focused on the underglycosylated MUC-1 tumor antigen (uMUC-1), which is directly linked to tumor progression from pre-malignancy to advanced malignancy in breast cancer and has been identified as the independent predictor of local recurrence and tumor response to chemotherapy. We investigated whether changes in uMUC-1 expression during tumor development and therapeutic intervention could be monitored non-invasively using molecular imaging approach with the uMUC-1-specific contrast agent (MN-EPPT) detectable by magnetic resonance and fluorescence optical imaging. This was done in mice that express human uMUC-1 tumor antigen (MMT mice) and develop spontaneous mammary carcinoma in a stage-wise fashion. After the injection of MN-EPPT there was a significant reduction in average T2 relaxation times of the mammary fat pad between pre-malignancy and cancer. In addition, T2 relaxation times were already altered at pre-malignant state in these mice compared to non-tumor bearing mice. This indicated that targeting uMUC-1 could be useful for detecting pre-malignant transformation in the mammary fat pad. We also probed changes in uMUC-1 expression with MN-EPPT during therapy with doxorubicin (Dox). We observed that tumor delta-T2s were significantly reduced by treatment with Dox indicating lower accumulation of MN-EPPT. This correlated with a lower level of MUC-1 expression in the Dox-treated tumors, as confirmed by immunoblotting. Our study could provide a very sensitive molecular imaging approach for monitoring tumor progression and therapeutic response.
Collapse
Affiliation(s)
- Subrata K Ghosh
- Molecular Imaging Laboratory, MGH/HST Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Yang FY, Yu MX, Zhou Q, Chen WL, Gao P, Huang Z. Effects of Iron Oxide Nanoparticle Labeling on Human Endothelial Cells. Cell Transplant 2012; 21:1805-20. [PMID: 22776829 DOI: 10.3727/096368912x652986] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Iron oxide nanoparticles (INOPS) are a potential contrast agent for magnetic resonance (MR) tracking of transplanted endothelial cells. The objective of this study was to examine the effect of INOPS labeling on endothelial cells. The mixture of INOPS and poly-l-lysine (PLL) was used to label human endothelial cells. Labeling efficiency was examined by Prussian blue staining, transmission electron microscopy, and atomic absorption spectrometry. The effect of iron oxide concentration on cell viability and proliferation were determined. The correlation of reactive oxygen species (ROS) and apoptosis was also examined. In vitro MRI scanning was carried out using a 1.5T MR system. INOPS-PLL could be readily taken up by endothelial cells and subsequently induce MRI signal intensity changes. However, higher labeling concentration (>50 μg/ml) and longer incubation (48 h) can affect cell viability and proliferation. Mitochondrial damage, apoptosis, and autolysosmes were observed under high INOPS-PLL concentrations, which were correlated to ROS production. INOPS-PLL nanoparticles can be used to label transplanted endothelial cells. However, high concentration of INOPS can impair cell viability, possibly through ROS-mediated apoptosis and autophagy.
Collapse
Affiliation(s)
- Fu-Yuan Yang
- MOE Key Laboratory of Laser Life Science, South China Normal University, Guangzhou, China
| | - Ming-Xi Yu
- MOE Key Laboratory of Laser Life Science, South China Normal University, Guangzhou, China
| | - Quan Zhou
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wen-Li Chen
- MOE Key Laboratory of Laser Life Science, South China Normal University, Guangzhou, China
| | - Peng Gao
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zheng Huang
- School of Medicine and School of Engineering and Applied Science, University of Colorado-Denver, Denver, CO, USA
| |
Collapse
|
24
|
Janic B, Arbab AS. Cord blood endothelial progenitor cells as therapeutic and imaging probes. ACTA ACUST UNITED AC 2012; 4:477-490. [PMID: 23227114 DOI: 10.2217/iim.12.35] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Numerous studies demonstrated that neovascularization processes associated with severe tissue ischemia commonly found in conditions such as cardiovascular disorders and tumor growth occur via angiogenic and vasculogenic mechanisms. Over the past decade, it has been demonstrated that endothelial progenitor cells (EPCs) play a significant role in neo-angiogenic and neovasculogenic processes. Due to their ability to self-renew, circulate, home to the ischemic sites and differentiate into mature endothelial cells, EPCs derived from various sources hold enormous potential to be used as therapeutic agents in pro- or anti-angiogenic strategies for the treatment of ischemic and tumor conditions, respectively. However, the development of EPC-based therapies requires accompanying, noninvasive imaging protocol for in vivo tracking of transplanted cells. Hence, this review focuses on cord blood-derived EPCs and their role in neovascularization with emphasis on the potential use of EPCs as a therapeutic and imaging probe.
Collapse
Affiliation(s)
- Branislava Janic
- Cellular & Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, 1 Ford Place, 2F, Box 82, Detroit, MI 48202, USA
| | | |
Collapse
|
25
|
Kosaka N, Bernardo M, Mitsunaga M, Choyke PL, Kobayashi H. MR and optical imaging of early micrometastases in lymph nodes: triple labeling with nano-sized agents yielding distinct signals. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 7:247-53. [PMID: 22434638 DOI: 10.1002/cmmi.489] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Few imaging methods are available for depicting in vivo cancer cell migration within the lymphatic system. Detection of such early micrometastases requires extremely high target to background. In this study, we dual-labeled human breast cancer cells (MDA-MB468) with a small particle of iron oxide (SPIO) and a quantum dot (QD), and tracked these cells in the lymphatic system in mice using in vivo MRI and optical imaging. A generation-6 gadolinium-dendrimer-based MRI contrast agent (Gd-G6) was employed for visualizing regional lymphatic channels and nodes. Since Gd-G6 shortened T(1) leading to high signal, whereas SPIO-labeled cancer cells greatly lowered signal, a small number of cells were simultaneously visualized within the draining lymphatic basins. One million dual-labeled cancer cells were subcutaneously injected into the paws of mice 24 h prior to imaging. Then whole body images were acquired pre- and post-intracutaneous injection of Gd-G6 with 3D-T(1) w-FFE and balanced-FFE sequences for cancer cell tracking and MR lymphangiography. In vivo MRI clearly visualized labeled cancer cells migrating from the paw to the axillary lymph nodes using draining lymphatics. In vivo optical imaging using a fluorescence surgical microscope demonstrated tiny cancer cell clusters in the axillary lymph node with high spatial resolution. Thus, using a combination of MRI and optical imaging, it is possible to depict macro- and early micrometastases within the lymphatic system. This platform offers a versatile research tool for investigating and treating lymphatic metastases in animal models.
Collapse
Affiliation(s)
- Nobuyuki Kosaka
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1088, USA
| | | | | | | | | |
Collapse
|
26
|
Arbab AS. Activation of alternative pathways of angiogenesis and involvement of stem cells following anti-angiogenesis treatment in glioma. Histol Histopathol 2012; 27:549-57. [PMID: 22419019 DOI: 10.14670/hh-27.549] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Malignant gliomas are hypervascular tumors that are highly resistant to all the currently available multimodal treatments. Therefore, anti-angiogenic therapies targeting VEGF or VEGF receptors (VEGFRs) were designed and thought to be an effective tool for controlling the growth of malignant gliomas. However, recent results of early clinical trials using humanized monoclonal antibodies against VEGF (Bevacizumab), as well as small-molecule tyrosine kinase inhibitors that target different VEGF receptors (VEGFRs) (Vatalanib, Vandetanib, Sunitinib, Sorafenib, etc) alone or in combination with other therapeutic agents demonstrated differing outcomes, with the majority of reports indicating that glioma developed resistance to the employed anti-angiogenic treatments. It has been noted that continued anti-angiogenic therapy targeting only the VEGF-VEGFR system might affect pro-angiogenic factors other than VEGF, such as basic fibroblast growth factor (bFGF), stromal derived factor 1 (SDF-1) and Tie-2. These factors may in turn stimulate angiogenesis by mobilizing bone marrow derived precursor cells, such as endothelial progenitor cells (EPCs), which are known to promote angiogenesis and vasculogenesis. In this short review, the current antiangiogenic treatments, possible mechanisms of activation of alternative pathways of angiogenesis, and possible involvement of bone marrow derived progenitor cells in the failure of anti-angiogenic treatments are discussed.
Collapse
Affiliation(s)
- Ali S Arbab
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, MI 48202, USA.
| |
Collapse
|
27
|
Chaudeurge A, Wilhelm C, Chen-Tournoux A, Farahmand P, Bellamy V, Autret G, Ménager C, Hagège A, Larghéro J, Gazeau F, Clément O, Menasché P. Can Magnetic Targeting of Magnetically Labeled Circulating Cells Optimize Intramyocardial Cell Retention? Cell Transplant 2012; 21:679-91. [DOI: 10.3727/096368911x612440] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Therapeutic intracavitary stem cell infusion currently suffers from poor myocardial homing. We examined whether cardiac cell retention could be enhanced by magnetic targeting of endothelial progenitor cells (EPCs) loaded with iron oxide nanoparticles. EPCs were magnetically labeled with citrate-coated iron oxide nanoparticles. Cell proliferation, migration, and CXCR4 chemokine receptor expression were assessed in different labeling conditions and no adverse effects of the magnetic label were observed. The magnetophoretic mobility of labeled EPCs was determined in vitro, with the same magnet as that subsequently used in vivo. Coronary artery occlusion was induced for 30 min in 36 rats (31 survivors), followed by 20 min of reperfusion. The rats were randomized to receive, during brief aortic cross-clamping, direct intraventricular injection of culture medium ( n = 7) or magnetically labeled EPCs ( n = 24), with ( n = 14) or without ( n = 10) subcutaneous insertion of a magnet over the chest cavity ( n = 14). The hearts were explanted 24 h later and engrafted cells were visualized by magnetic resonance imaging (MRI) of the heart at 1.5 T. Their abundance in the myocardium was also analyzed semiquantitatively by immunofluorescence, and quantitatively by real-time polymerase chain reaction (RT-PCR). Although differences in cell retention between groups failed to be statistically significant using RT-PCR quantification, due to the variability of the animal model, immunostaining showed that the average number of engrafted EPCs was significantly ten times higher with than without magnetic targeting. There was thus a consistent trend favoring the magnet-treated hearts, thereby suggesting magnetic targeting as a potentially new mean of enhancing myocardial homing of intravascularly delivered stem cells. Magnetic targeting has the potential to enhance myocardial retention of intravascularly delivered endothelial progenitor cells.
Collapse
Affiliation(s)
- Aurélie Chaudeurge
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Ecole de Chirurgie, Paris, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes MSC, CNRS UMR 7057, Paris, France
- Université Paris-Diderot, Paris, France
| | - Annabel Chen-Tournoux
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Ecole de Chirurgie, Paris, France
| | - Patrick Farahmand
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Valérie Bellamy
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Ecole de Chirurgie, Paris, France
| | - Gwennhael Autret
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- INSERM, U970, Paris Cardiovascular Research Center-PARCC, Paris, France
| | | | - Albert Hagège
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jerome Larghéro
- Univ Paris 06-CNRS-ESPCI Laboratoire PECSA UMR7195, Paris, France
- University Paris Diderot, Paris, France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes MSC, CNRS UMR 7057, Paris, France
- Université Paris-Diderot, Paris, France
| | - Olivier Clément
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- INSERM, U970, Paris Cardiovascular Research Center-PARCC, Paris, France
| | - Philippe Menasché
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
28
|
Abarrategi A, Fernandez-Valle ME, Desmet T, Castejón D, Civantos A, Moreno-Vicente C, Ramos V, Sanz-Casado JV, Martínez-Vázquez FJ, Dubruel P, Miranda P, López-Lacomba JL. Label-free magnetic resonance imaging to locate live cells in three-dimensional porous scaffolds. J R Soc Interface 2012; 9:2321-31. [PMID: 22442095 DOI: 10.1098/rsif.2012.0068] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Porous scaffolds are widely tested materials used for various purposes in tissue engineering. A critical feature of a porous scaffold is its ability to allow cell migration and growth on its inner surface. Up to now, there has not been a method to locate live cells deep inside a material, or in an entire structure, using real-time imaging and a non-destructive technique. Herein, we seek to demonstrate the feasibility of the magnetic resonance imaging (MRI) technique as a method to detect and locate in vitro non-labelled live cells in an entire porous material. Our results show that the use of optimized MRI parameters (4.7 T; repetition time = 3000 ms; echo time = 20 ms; resolution 39 × 39 µm) makes it possible to obtain images of the scaffold structure and to locate live non-labelled cells in the entire material, with a signal intensity higher than that obtained in the culture medium. In the current study, cells are visualized and located in different kinds of porous scaffolds. Moreover, further development of this MRI method might be useful in several three-dimensional biomaterial tests such as cell distribution studies, routine qualitative testing methods and in situ monitoring of cells inside scaffolds.
Collapse
Affiliation(s)
- A Abarrategi
- Institute of Biofunctional Studies, Nuclear Magnetic Resonance, Complutense University, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gianolio E, Stefania R, Di Gregorio E, Aime S. MRI Paramagnetic Probes for Cellular Labeling. Eur J Inorg Chem 2012. [DOI: 10.1002/ejic.201101399] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
30
|
Ramaswamy S, Schornack PA, Smelko AG, Boronyak SM, Ivanova J, Mayer JE, Sacks MS. Superparamagnetic iron oxide (SPIO) labeling efficiency and subsequent MRI tracking of native cell populations pertinent to pulmonary heart valve tissue engineering studies. NMR IN BIOMEDICINE 2012; 25:410-417. [PMID: 22351640 DOI: 10.1002/nbm.1642] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 10/07/2010] [Accepted: 10/14/2010] [Indexed: 05/31/2023]
Abstract
The intimal and medial linings of the pulmonary artery consist largely of vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs), respectively. The migration of these cell types to a potential tissue-engineered pulmonary valve (TEPV) implant process is therefore of interest in understanding the valve remodeling process. Visualization and cell tracking by MRI, which employs hypointense contrast achievable through the use of superparamagnetic iron oxide (SPIO) microparticles to label cells, provides a method in which this can be studied. We investigated the SPIO labeling efficiency of human VECs and VSMCs, and used two- and three-dimensional gradient echo sequences to track the migration of these cells in agar gel constructs. Protamine sulfate (4.5 µg/mL) was used to enhance SPIO uptake and was found to have no influence on cell viability or proliferation. MRI experiments were initially performed using a 9.4-T scanner. The results demonstrated that the spatial positions of hypointense spots were relatively unchanged over 12 days. Subsequent MR experiments performed at 7 T demonstrated that three-dimensional imaging provided the best spatial resolution to assess cell fate. R(2)* maps were bright in SPIO cell-encapsulated gels in comparison with unlabeled counterparts. Signal voids were ruled out as hypointense regions owing to the smooth exponential decay of T(2)* in these voxels. As a next step, we intend to use the SPIO cell labeling and MR protocols established in this study to assess whether hemodynamic stresses will alter the vascular cell migratory patterns. These studies will shed light on the mechanisms of vascular remodeling after TEPV implantation.
Collapse
Affiliation(s)
- Sharan Ramaswamy
- Department of Biomedical Engineering, Florida International University, College of Engineering and Computing, Miami, FL 33174, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Plank C, Zelphati O, Mykhaylyk O. Magnetically enhanced nucleic acid delivery. Ten years of magnetofection-progress and prospects. Adv Drug Deliv Rev 2011; 63:1300-31. [PMID: 21893135 PMCID: PMC7103316 DOI: 10.1016/j.addr.2011.08.002] [Citation(s) in RCA: 205] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 12/28/2022]
Abstract
Nucleic acids carry the building plans of living systems. As such, they can be exploited to make cells produce a desired protein, or to shut down the expression of endogenous genes or even to repair defective genes. Hence, nucleic acids are unique substances for research and therapy. To exploit their potential, they need to be delivered into cells which can be a challenging task in many respects. During the last decade, nanomagnetic methods for delivering and targeting nucleic acids have been developed, methods which are often referred to as magnetofection. In this review we summarize the progress and achievements in this field of research. We discuss magnetic formulations of vectors for nucleic acid delivery and their characterization, mechanisms of magnetofection, and the application of magnetofection in viral and nonviral nucleic acid delivery in cell culture and in animal models. We summarize results that have been obtained with using magnetofection in basic research and in preclinical animal models. Finally, we describe some of our recent work and end with some conclusions and perspectives.
Collapse
|
32
|
Sandu N, Momen-Heravi F, Sadr-Eshkevari P, Schaller B. Molecular imaging for stem cell transplantation in neuroregenerative medicine. NEURODEGENER DIS 2011; 9:60-7. [PMID: 22042219 DOI: 10.1159/000330713] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 06/03/2011] [Indexed: 12/12/2022] Open
Abstract
Stem cell transplantation is a promising new therapeutic option in different neurological diseases. However, it was not yet possible to translate its potential from animal models to clinical application. One of the main problems of applying stem cell transplantation in clinical medium is the difficulty of detection, localization, and examination of the stem cells in vivo at both cellular and molecular levels. State-of-the-art molecular imaging techniques provide new and better means for noninvasive, repeated, and quantitative tracking of stem cell implant or transplant. From initial deposition to the survival, migration, and differentiation of the transplant/implanted stem cells, current molecular imaging methods allow monitoring of the infused cells in the same live recipient over time. The present review briefly summarizes and compares these molecular imaging methods for cell labeling and imaging in animal models as well as in clinical application and sheds light on consecutive new therapeutic options if appropriate.
Collapse
Affiliation(s)
- Nora Sandu
- Department of Neurosurgery, University of Lausanne, Lausanne, Switzerland
| | | | | | | |
Collapse
|
33
|
Levy M, Wilhelm C, Luciani N, Deveaux V, Gendron F, Luciani A, Devaud M, Gazeau F. Nanomagnetism reveals the intracellular clustering of iron oxide nanoparticles in the organism. NANOSCALE 2011; 3:4402-4410. [PMID: 21931920 DOI: 10.1039/c1nr10778j] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
There are very few methods to investigate how nanoparticles (NPs) are taken up and processed by cells in the organism in the short and long terms. We propose a nanomagnetism approach, in combination with electron microscopy, to document the magnetic outcome of iron oxide-based P904 NPs injected intravenously into mice. The NP superparamagnetic properties are shown to be modified by cell internalization, due to magnetic interactions between NPs sequestered within intracellular organelles. These modifications of magnetic behaviour are observed in vivo after NP uptake by resident macrophages in spleen and liver or by inflammatory macrophages in adipose tissue as well as in vitro in monocyte-derived macrophages. The dynamical magnetic response of cell-internalized NPs is theoretically and experimentally evidenced as a global signature of their local organization in the intracellular compartments. The clustering of NPs and their magnetism become dependent on the targeted organ, on the dose administrated and on the time elapsed since their injection. Nanomagnetism probes the intracellular clustering of iron-oxide NPs and sheds light on the impact of cellular metabolism on their magnetic responsivity.
Collapse
Affiliation(s)
- Michael Levy
- Laboratoire Matière et Systèmes Complexes, UMR 7057 CNRS/Université Paris - Diderot, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Rago G, Langer CM, Brackman C, Day JP, Domke KF, Raschzok N, Schmidt C, Sauer IM, Enejder A, Mogl MT, Bonn M. CARS microscopy for the visualization of micrometer-sized iron oxide MRI contrast agents in living cells. BIOMEDICAL OPTICS EXPRESS 2011; 2:2470-2483. [PMID: 21991541 PMCID: PMC3184857 DOI: 10.1364/boe.2.002470] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 07/25/2011] [Accepted: 07/28/2011] [Indexed: 05/31/2023]
Abstract
Micrometer-sized iron oxide particles (MPIOs) attract increasing interest as contrast agents for cellular tracking by clinical Magnetic Resonance Imaging (MRI). Despite the great potential of MPIOs for in vivo imaging of labeled cells, little is known on the intracellular localization of these particles following uptake due to the lack of techniques with the ability to monitor the particle uptake in vivo at single-cell level. Here, we show that coherent anti-Stokes Raman scattering (CARS) microscopy enables non-invasive, label-free imaging of MPIOs in living cells with sub-micron resolution in three dimensions. CARS allows simultaneous visualization of the cell framework and the MPIOs, where the particles can be readily distinguished from other cellular components of comparable dimensions, and localized inside the cell.
Collapse
Affiliation(s)
- Gianluca Rago
- FOM Institute AMOLF, Science Park 104, 1098XG Amsterdam, The Netherlands
| | - Carolin M. Langer
- General, Visceral, and Transplantation Surgery and Regenerative Medicine, Charité-Campus Virchow, Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Christian Brackman
- Department of Chemical and Biological Engineering, Chalmers University of Technology, Kemivagen 4, Göteborg, Sweden
| | - James P.R. Day
- FOM Institute AMOLF, Science Park 104, 1098XG Amsterdam, The Netherlands
| | - Katrin F. Domke
- FOM Institute AMOLF, Science Park 104, 1098XG Amsterdam, The Netherlands
| | - Nathanael Raschzok
- General, Visceral, and Transplantation Surgery and Regenerative Medicine, Charité-Campus Virchow, Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | | | - Igor M. Sauer
- General, Visceral, and Transplantation Surgery and Regenerative Medicine, Charité-Campus Virchow, Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Annika Enejder
- Department of Chemical and Biological Engineering, Chalmers University of Technology, Kemivagen 4, Göteborg, Sweden
- These authors contributed equally to the manuscript
| | - Martina T. Mogl
- General, Visceral, and Transplantation Surgery and Regenerative Medicine, Charité-Campus Virchow, Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
- These authors contributed equally to the manuscript
| | - Mischa Bonn
- FOM Institute AMOLF, Science Park 104, 1098XG Amsterdam, The Netherlands
- These authors contributed equally to the manuscript
| |
Collapse
|
35
|
Harrington JK, Chahboune H, Criscione JM, Li AY, Hibino N, Yi T, Villalona GA, Kobsa S, Meijas D, Duncan DR, Devine L, Papademetri X, Shin'oka T, Fahmy TM, Breuer CK. Determining the fate of seeded cells in venous tissue-engineered vascular grafts using serial MRI. FASEB J 2011; 25:4150-61. [PMID: 21846838 DOI: 10.1096/fj.11-185140] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A major limitation of tissue engineering research is the lack of noninvasive monitoring techniques for observations of dynamic changes in single tissue-engineered constructs. We use cellular magnetic resonance imaging (MRI) to track the fate of cells seeded onto functional tissue-engineered vascular grafts (TEVGs) through serial imaging. After in vitro optimization, murine macrophages were labeled with ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles and seeded onto scaffolds that were surgically implanted as inferior vena cava interposition grafts in SCID/bg mice. Serial MRI showed the transverse relaxation times (T(2)) were significantly lower immediately following implantation of USPIO-labeled scaffolds (T(2) = 44 ± 6.8 vs. 71 ± 10.2 ms) but increased rapidly at 2 h to values identical to control implants seeded with unlabeled macrophages (T(2) = 63 ± 12 vs. 63 ± 14 ms). This strongly indicates the rapid loss of seeded cells from the scaffolds, a finding verified using Prussian blue staining for iron containing macrophages on explanted TEVGs. Our results support a novel paradigm where seeded cells are rapidly lost from implanted scaffolds instead of developing into cells of the neovessel, as traditionally thought. Our findings confirm and validate this paradigm shift while demonstrating the first successful application of noninvasive MRI for serial study of cellular-level processes in tissue engineering.
Collapse
Affiliation(s)
- Jamie K Harrington
- Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Řehoř I, Vilímová V, Jendelová P, Kubíček V, Jirák D, Herynek V, Kapcalová M, Kotek J, Černý J, Hermann P, Lukeš I. Phosphonate–Titanium Dioxide Assemblies: Platform for Multimodal Diagnostic–Therapeutic Nanoprobes. J Med Chem 2011; 54:5185-94. [DOI: 10.1021/jm200449y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
37
|
Robert D, Pamme N, Conjeaud H, Gazeau F, Iles A, Wilhelm C. Cell sorting by endocytotic capacity in a microfluidic magnetophoresis device. LAB ON A CHIP 2011; 11:1902-10. [PMID: 21512692 DOI: 10.1039/c0lc00656d] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Magnetically labelled cells are finding a wealth of applications for in vitro analysis as well as in vivo treatments. Sorting of cells into subpopulations based on their magnetite loading is an important step in such procedures. Here, we study the sorting of monocytes and macrophages which internalise nanoparticles to different extents based on their endocytotic capacity. Macrophages featured a high endocytotic activity and were found to internalise between 4 and 60 pg of iron per cell. They were successfully sorted into five subpopulations of narrow iron loading distributions via on-chip free-flow magnetophoresis, thus demonstrating the potential of sorting of relatively similarly loaded cells. Monocytes featured a low endocytotic capacity and took on 1 to 4 pg of iron per cell. Mixtures of monocytes and macrophages were successfully sorted within the free-flow magnetophoresis chip and good purity (>88%), efficacy (>60%) and throughput (from 10 to 100 cells s(-1)) could be achieved. The introduced method constitutes a viable tool for studies of endocytotic capacity and sorting/selection of cells based on this functionality.
Collapse
Affiliation(s)
- Damien Robert
- Laboratoire Matière et Systèmes Complexes, UMR CNRS et Université Paris Diderot, France
| | | | | | | | | | | |
Collapse
|
38
|
Wu S, Zhang L, Zhong J, Zhang Z. Dual contrast magnetic resonance imaging tracking of iron-labeled cells in vivo. Cytotherapy 2011; 12:859-69. [PMID: 20184501 DOI: 10.3109/14653241003587652] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Negative-contrast magnetic resonance imaging (MRI) methods utilizing magnetic susceptibility contrast agents have become one of the most widely used approaches in cellular imaging research. However, visualizing and tracking super-paramagnetic iron oxide nanoparticle (SPIO)-labeled cells on the basis of negative-contrast can limit specificity and sensitivity. Therefore, there has been a strong motivation to explore MRI methods for cellular imaging with either positive or dual contrast (both positive and negative) for identifying labeled cells; these methods offer the potential to improve significantly the sensitivity and specificity of MRI-based cell-tracking approaches. In this review, current state-of-the-art positive- and dual-contrast MRI techniques and contrast agents are described specifically for applications involving in vivo cellular tracking and imaging.
Collapse
Affiliation(s)
- Shengyong Wu
- Medical Imaging Institute of Tianjin, Tianjin, China
| | | | | | | |
Collapse
|
39
|
Enhancing Stroke Recovery with Cellular Therapies. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
40
|
Vats N, Wilhelm C, Rautou PE, Poirier-Quinot M, Péchoux C, Devue C, Boulanger CM, Gazeau F. Magnetic tagging of cell-derived microparticles: new prospects for imaging and manipulation of these mediators of biological information. Nanomedicine (Lond) 2010; 5:727-38. [DOI: 10.2217/nnm.10.44] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: Submicron membrane fragments termed microparticles (MPs), which are released by apoptotic or activated cells, are newly considered as vectors of biological information and actors of pathology development. We propose the tagging of MPs with magnetic nanoparticles as a new approach allowing imaging, manipulation and targeting of cell-derived MPs. Materials & methods: MPs generated in vitro from human endothelial cells or isolated from atherosclerotic plaques were labeled using citrate-coated 8 nm iron-oxide nanoparticles. MPs were tagged with magnetic nanoparticles on their surface and detected as Annexin-V positive by flow cytometry. Results: Labeled MPs could be mobilized, isolated and manipulated at a distance in a magnetic field gradient. Magnetic mobility of labeled MPs was quantified by micromagnetophoresis. Interactions of labeled MPs with endothelial cells could be triggered and modulated by magnetic guidance. Nanoparticles served as tracers at different scales: at the subcellular level by electron microscopy, at the cellular level by histology and at the macroscopic level by MRI. Conclusion: Magnetic labeling of biogenic MPs opens new prospects for noninvasive monitoring and distal manipulations of these biological effectors.
Collapse
Affiliation(s)
- Nidhi Vats
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS & Université Paris Diderot, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris cedex 13, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS & Université Paris Diderot, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris cedex 13, France
| | - Pierre-Emmanuel Rautou
- Paris Centre de Recherche Cardiovasculaire, INSERM U970, 56 Rue Leblanc, 75737, Paris cedex 15, France
| | - Marie Poirier-Quinot
- Laboratoire U2R2M, UMR8081, CNRS & Université Paris-Sud, Centre d’Orsay, 91405, Orsay cedex, France
| | - Christine Péchoux
- Centre de Microscopie Électronique, Plateforme MIMA2, INRA, UR1196 Génomique et Physiologie de la Lactation, Domaine de Vilvert, F-78352 Jouy-en-Josas, France
| | - Cécile Devue
- Paris Centre de Recherche Cardiovasculaire, INSERM U970, 56 Rue Leblanc, 75737, Paris cedex 15, France
| | - Chantal M Boulanger
- Paris Centre de Recherche Cardiovasculaire, INSERM U970, 56 Rue Leblanc, 75737, Paris cedex 15, France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS & Université Paris Diderot, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris cedex 13, France
| |
Collapse
|
41
|
Kotková Z, Kotek J, Jirák D, Jendelová P, Herynek V, Berková Z, Hermann P, Lukeš I. Cyclodextrin-Based Bimodal Fluorescence/MRI Contrast Agents: An Efficient Approach to Cellular Imaging. Chemistry 2010; 16:10094-102. [DOI: 10.1002/chem.200903519] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
42
|
Poirier-Quinot M, Frasca G, Wilhelm C, Luciani N, Ginefri JC, Darrasse L, Letourneur D, Le Visage C, Gazeau F. High-Resolution 1.5-Tesla Magnetic Resonance Imaging for Tissue-Engineered Constructs: A Noninvasive Tool to Assess Three-Dimensional Scaffold Architecture and Cell Seeding. Tissue Eng Part C Methods 2010; 16:185-200. [DOI: 10.1089/ten.tec.2009.0015] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Marie Poirier-Quinot
- Unité de Recherche en Résonance Magnétique Médicale, (U2R2M) UMR 8081 CNRS, Université Paris Sud, Orsay, France
| | - Guillaume Frasca
- Laboratoire Matière et Systèmes Complexes, UMR 7057 CNRS, Université Paris–Diderot, Paris, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes, UMR 7057 CNRS, Université Paris–Diderot, Paris, France
| | - Nathalie Luciani
- Laboratoire Matière et Systèmes Complexes, UMR 7057 CNRS, Université Paris–Diderot, Paris, France
| | - Jean-Christophe Ginefri
- Unité de Recherche en Résonance Magnétique Médicale, (U2R2M) UMR 8081 CNRS, Université Paris Sud, Orsay, France
| | - Luc Darrasse
- Unité de Recherche en Résonance Magnétique Médicale, (U2R2M) UMR 8081 CNRS, Université Paris Sud, Orsay, France
| | - Didier Letourneur
- Inserm U698, Bio-ingénierie Cardiovasculaire, CHU X. Bichat, Paris, France
| | | | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes, UMR 7057 CNRS, Université Paris–Diderot, Paris, France
| |
Collapse
|
43
|
Magnetic labeling, imaging and manipulation of endothelial progenitor cells using iron oxide nanoparticles. Future Med Chem 2010; 2:397-408. [DOI: 10.4155/fmc.09.165] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Endothelial progenitor cells (EPCs), originating from bone marrow, play a significant role in the repair of ischemic tissue and injured blood vessels. They are also involved in tumor angiogenesis. The therapeutic potential of EPCs for regenerative medicine and cancer treatment calls for new methods for monitoring and controlling cell migration. This review focuses on promising magnetic methods based on the internalization of magnetic nanoparticles by EPCs. We first describe the cellular uptake of iron oxide nanoparticles depending on their surface properties. We thus review the use of MRI for the detection of labeled cells and for noninvasive follow-up of EPCs homing in sites of endothelium regeneration. Finally, we show that remotely applied magnetic forces may enable intracellular manipulation and may optimize cell-delivery strategies for localizing cell therapy to target sites.
Collapse
|
44
|
Abstract
The development of new methods for noninvasive imaging is an area of biotechnology that is of great relevance for the diagnosis and characterization of diabetes mellitus. Noninvasive imaging can be used to study the dynamics of beta-cell mass and function; beta-cell death; vascularity, innervation and autoimmune attack of pancreatic islets; and the efficacy of islet transplantation to remedy beta-cell loss in patients with diabetes mellitus. In this Review, we focus on the application of MRI for monitoring islet transplantation and on the potential causes of islet graft failure, which are still poorly understood. Questions that have been addressed by MRI studies encompass graft longevity, and the effects of immune rejection, glucose toxic effects, and the transplanted islets' purity on graft fate. We also highlight novel technologies for simultaneous imaging and delivery of experimental therapies that aim to extend the lifespan and functionality of islet grafts. On the basis of this evidence, MRI represents a valuable platform for a thorough investigation of beta-cell function in the context of islet transplantation. State-of-the-art multimodality approaches, such as PET-MRI, can extend our current capabilities and help answer the critical questions that currently inhibit the prevention and cure of diabetes mellitus.
Collapse
Affiliation(s)
- Zdravka Medarova
- Molecular Imaging Laboratory, Massachusetts General Hospital-Massachusetts Institute of Technology-Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, 13th Street, Charlestown, MA 02129, USA
| | | |
Collapse
|
45
|
Zhang Z, Mascheri N, Dharmakumar R, Fan Z, Paunesku T, Woloschak G, Li D. Superparamagnetic iron oxide nanoparticle-labeled cells as an effective vehicle for tracking the GFP gene marker using magnetic resonance imaging. Cytotherapy 2009; 11:43-51. [PMID: 18956269 DOI: 10.1080/14653240802420243] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Detection of a gene using magnetic resonance imaging (MRI) is hindered by the magnetic resonance (MR) targeting gene technique. Therefore it may be advantageous to image gene-expressing cells labeled with superparamagnetic iron oxide (SPIO) nanoparticles by MRI. METHODS The GFP-R3230Ac (GFP) cell line was incubated for 24 h using SPIO nanoparticles at a concentration of 20 microg Fe/mL. Cell samples were prepared for iron content analysis and cell function evaluation. The labeled cells were imaged using fluorescent microscopy and MRI. RESULTS SPIO was used to label GFP cells effectively, with no effects on cell function and GFP expression. Iron-loaded GFP cells were successfully imaged with both fluorescent microscopy and T2*-weighted MRI. Prussian blue staining showed intracellular iron accumulation in the cells. All cells were labeled (100% labeling efficiency). The average iron content per cell was 4.75+/-0.11 pg Fe/cell (P<0.05 versus control). DISCUSSION This study demonstrates that the GFP expression of cells is not altered by the SPIO labeling process. SPIO-labeled GFP cells can be visualized by MRI; therefore, GFP, a gene marker, was tracked indirectly with the SPIO-loaded cells using MRI. The technique holds promise for monitoring the temporal and spatial migration of cells with a gene marker and enhancing the understanding of cell- and gene-based therapeutic strategies.
Collapse
Affiliation(s)
- Z Zhang
- Department of Radiology, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Cell labeling and tracking for experimental models using magnetic resonance imaging. Methods 2009; 48:112-24. [PMID: 19362150 DOI: 10.1016/j.ymeth.2009.03.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Accepted: 03/28/2009] [Indexed: 01/05/2023] Open
Abstract
Magnetic Resonance Imaging (MRI), as one of the most powerful methods in clinical diagnosis, has emerged as an additional method in the field of molecular and cellular imaging. Compared to established molecular imaging methods, MRI provides in vivo images with high resolution. In particularly in the field of cell-based therapy, non-invasively acquired information on temporal changes of cell location linked to high-resolution anatomical information is of great interest. Relatively new approaches like responsive contrast agents or MR imaging reporter gene expression are MRI applications beyond temporal and spatial information on labeled cells towards investigations on functional changes of cells in vivo. MRI-based cell monitoring and tracking studies require prior labeling of the cells under investigation for excellent contrast against the background of host tissue. Here, an overview is provided on contrast generation strategies for MRI of cells. This includes MR contrast agents, various approaches of cell labeling and MRI as well as MR spectroscopic methods used for cell tracking in vivo. Advantages and disadvantages of the particular labeling approaches and methods are discussed. In addition to description of the methods, the emphasis is on the potential but also challenges and shortcomings of this imaging technique for applications that aim to visualize cellular processes in vivo.
Collapse
|
47
|
Frasca G, Gazeau F, Wilhelm C. Formation of a three-dimensional multicellular assembly using magnetic patterning. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2009; 25:2348-2354. [PMID: 19166275 DOI: 10.1021/la8030792] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
We demonstrate a facile approach to design three-dimensional cellular assembly of tunable size and controlled geometry with applications for tissue engineering. Three-dimensional cell patterning was performed using external magnetic forces, without the need for substrate chemical or physical modifications. Human endothelial progenitor cells and mouse macrophages were magnetically labeled using anionic citrate-coated iron oxide nanoparticles. Two magnetic tips were designed, and their magnetic field cartographies were calibrated. The focalized magnetic force generated ensured an efficient entrapment of the cells at the tips vicinity. By tuning the magnetic field gradient geometry and intensity, the magnetic cellular load, and the number of cells, we fully described the formation of the three-dimensional multicellular assemblies, and estimated the corresponding packing factor for a large range of experimental conditions.
Collapse
Affiliation(s)
- Guillaume Frasca
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & Université Paris-Diderot, Paris Cedex 13, F-75205, France
| | | | | |
Collapse
|
48
|
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: 3.9] [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
|
49
|
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
|
50
|
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: 8.9] [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
|