1
|
Piao YJ, Kim HS, Moon WK. Noninvasive Photoacoustic Imaging of Dendritic Cell Stimulated with Tumor Cell-Derived Exosome. Mol Imaging Biol 2020; 22:612-622. [PMID: 31385127 DOI: 10.1007/s11307-019-01410-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE The tools to trigger dendritic cell (DC) activation and to verify DC migration in vivo are important for directing DC immunotherapy toward successful treatment. We evaluated whether tumor cell-derived exosome (TEX)-stimulated DC migration into lymph node (LN) in mouse could be tracked using gold nanoparticle (GN)-labeling and ultrasound (US)-guided photoacoustic imaging (PAI). PROCEDURES GFP-transduced DC2.4 cells were used. RFP-tagged TEXs were purified from a stable 4T1 cell line overexpressing the exosomal CD63-RFP fusion protein. The TEX uptake by DCs was visualized using confocal laser scanning microscopy. GNs with surface plasmon resonance at 808 nm were used for DC-labeling. DCs that migrated into axillary LN were longitudinally monitored by US-guided PAI and analyzed by silver staining and immunohistochemistry. RESULTS TEXs were easily internalized in DCs, increased proliferation and migration capacities, and upregulated co-stimulatory molecules, CCR7 and TNF-α without cytotoxicity. The GN-labeling exerted no adverse effects on the biological functions of DCs. US-guided PAI and DC-labeling allowed for sensitive and longitudinal monitoring of TEX-stimulated DC migration toaxillary LN. CONCLUSIONS TEXs efficiently activated DCs and GN-labeled DC migration into LN was successfully monitored using US-guided PAI, suggesting that TEXs are a good source for DC activation and US-guided PAI is a cost-effective and easy-to-use imaging modality for noninvasive tracking of DCs.
Collapse
Affiliation(s)
- Yin Ji Piao
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Hoe Suk Kim
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
2
|
Zhang C, Xu Z, Di H, Zeng E, Jiang Y, Liu D. Gadolinium-doped Au@prussian blue nanoparticles as MR/SERS bimodal agents for dendritic cell activating and tracking. Am J Cancer Res 2020; 10:6061-6071. [PMID: 32483438 PMCID: PMC7255006 DOI: 10.7150/thno.42114] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
In vivo tracking of dendritic cell (DC) migration to the lymphatic system is essential for evaluating the outcome of DC-based immunotherapies. Novel multimodal imaging strategies with high analytical performance are urgently needed to supply complementary information about the migration and colonization of DCs. In this study, we designed a bimodal imaging agent, namely Au@Prussian blue-Gd@ovalbumin nanoparticles (APG@OVA NPs), for activating DCs and real-time tracking of DC migration process by magnetic resonance imaging (MRI). Moreover, the distribution of the colonized DCs in the lymphatic system was profiled at the single-cell levels based on surface-enhanced Raman scattering (SERS) technique. Methods: In this strategy, PBs as cyanide (CN)-bridged coordination blocks were assembled onto the gold nanoparticles core to provide SERS signal in the Raman-silent region (1800 and 2800 cm-1), which could avoid background signal interference. The doping Gd3+ located in the lattice of PB enables the MRI ability with high relaxivity of the probe. Ovalbumin, an egg allergen, was used as an antigen to activate DCs due to its immunological properties. The prepared APG@OVA NP agents were used to activate DCs with high efficacy and to track their migration and distribution in vivo through SERS/MR bimodal imaging. Results: The APG@OVA NP agents could not only enable DC activating and labeling, but also achieve real-time monitoring of DC migration in vivo and accurate profiling of DC distribution in the lymphatic system. MR imaging indicated the time-dependent migration of the APG@OVA NP-labeled DCs from the footpad to the sentinel lymph node. The background-free Raman mapping of the lymph node tissue slice demonstrated that the activated DCs have successfully colonized to the sentinel lymph node. Conclusion: Concerning the high activating efficacy, dual complementary imaging readouts, and low biological toxicity, the APG@OVA NPs act as high-performance tracking agents for DC-based immunotherapies.
Collapse
|
3
|
Perrin J, Capitao M, Mougin-Degraef M, Guérard F, Faivre-Chauvet A, Rbah-Vidal L, Gaschet J, Guilloux Y, Kraeber-Bodéré F, Chérel M, Barbet J. Cell Tracking in Cancer Immunotherapy. Front Med (Lausanne) 2020; 7:34. [PMID: 32118018 PMCID: PMC7033605 DOI: 10.3389/fmed.2020.00034] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
The impressive development of cancer immunotherapy in the last few years originates from a more precise understanding of control mechanisms in the immune system leading to the discovery of new targets and new therapeutic tools. Since different stages of disease progression elicit different local and systemic inflammatory responses, the ability to longitudinally interrogate the migration and expansion of immune cells throughout the whole body will greatly facilitate disease characterization and guide selection of appropriate treatment regiments. While using radiolabeled white blood cells to detect inflammatory lesions has been a classical nuclear medicine technique for years, new non-invasive methods for monitoring the distribution and migration of biologically active cells in living organisms have emerged. They are designed to improve detection sensitivity and allow for a better preservation of cell activity and integrity. These methods include the monitoring of therapeutic cells but also of all cells related to a specific disease or therapeutic approach. Labeling of therapeutic cells for imaging may be performed in vitro, with some limitations on sensitivity and duration of observation. Alternatively, in vivo cell tracking may be performed by genetically engineering cells or mice so that may be revealed through imaging. In addition, SPECT or PET imaging based on monoclonal antibodies has been used to detect tumors in the human body for years. They may be used to detect and quantify the presence of specific cells within cancer lesions. These methods have been the object of several recent reviews that have concentrated on technical aspects, stressing the differences between direct and indirect labeling. They are briefly described here by distinguishing ex vivo (labeling cells with paramagnetic, radioactive, or fluorescent tracers) and in vivo (in vivo capture of injected radioactive, fluorescent or luminescent tracers, or by using labeled antibodies, ligands, or pre-targeted clickable substrates) imaging methods. This review focuses on cell tracking in specific therapeutic applications, namely cell therapy, and particularly CAR (Chimeric Antigen Receptor) T-cell therapy, which is a fast-growing research field with various therapeutic indications. The potential impact of imaging on the progress of these new therapeutic modalities is discussed.
Collapse
Affiliation(s)
- Justine Perrin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marisa Capitao
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marie Mougin-Degraef
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - François Guérard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - Latifa Rbah-Vidal
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Joëlle Gaschet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yannick Guilloux
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | | |
Collapse
|
4
|
Sahu KK, Pandey RS. Development and characterization of HBsAg-loaded Eudragit nanoparticles for effective colonic immunization. Pharm Dev Technol 2018; 24:166-175. [DOI: 10.1080/10837450.2018.1444639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Kantrol Kumar Sahu
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
5
|
Tsukasaki Y, Komatsuzaki A, Mori Y, Ma Q, Yoshioka Y, Jin T. A short-wavelength infrared emitting multimodal probe for non-invasive visualization of phagocyte cell migration in living mice. Chem Commun (Camb) 2015; 50:14356-9. [PMID: 25296382 DOI: 10.1039/c4cc06542e] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
For the non-invasive visualization of cell migration in deep tissues, we synthesized a short-wavelength infrared (SWIR) emitting multimodal probe that contains PbS/CdS quantum dots, rhodamine 6G and iron oxide nanoparticles. This probe enables multimodal (SWIR fluorescence/magnetic resonance) imaging of phagocyte cell migration in living mice.
Collapse
Affiliation(s)
- Y Tsukasaki
- RIKEN Quantitative Biology Center, 6-2-3 Furuedai, Suita, Osaka 565-0874, Japan.
| | | | | | | | | | | |
Collapse
|
6
|
Young JS, Kim JW, Ahmed AU, Lesniak MS. Therapeutic cell carriers: a potential road to cure glioma. Expert Rev Neurother 2015; 14:651-60. [PMID: 24852229 DOI: 10.1586/14737175.2014.917964] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many different experimental molecular therapeutic approaches have been evaluated in an attempt to treat brain cancer. However, despite the success of these experimental molecular therapies, research has shown that the specific and efficient delivery of therapeutic agents to tumor cells is a limitation. In this regard, cell carrier systems have garnered significant attraction due to their capacity to be loaded with therapeutic agents and carry them specifically to tumor sites. Furthermore, cell carriers can be genetically modified to express therapeutic agents that can directly eradicate cancerous cells or can modulate tumor microenvironments. This review describes the current state of cell carriers, their use as vehicles for the delivery of therapeutic agents to brain tumors, and future directions that will help overcome the present obstacles to cell carrier mediated therapy for brain cancer.
Collapse
Affiliation(s)
- Jacob S Young
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
7
|
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
|
8
|
Chen YC, Wen S, Shang SA, Cui Y, Luo B, Teng GJ. Magnetic resonance and near-infrared imaging using a novel dual-modality nano-probe for dendritic cell tracking in vivo. Cytotherapy 2013; 16:699-710. [PMID: 24219906 DOI: 10.1016/j.jcyt.2013.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 09/14/2013] [Accepted: 09/28/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND AIMS The effect of cellular-based immunotherapy is highly correlated with the success of dendritic cells (DCs) homing to the draining lymph nodes (LNs) and interacting with antigen-specific CD4(+) T cells. In this study, a novel magneto-fluorescent nano-probe was used to track the in vivo migration of DCs to the draining LNs. METHODS A dual-modality nano-probe composed of superparamagnetic iron oxide (SPIO) and near-infrared fluorescent (NIRF) dye (NIR797) was developed, and its magnetic and optical contrasting properties were characterized. DCs generated from mouse bone marrow were co-cultured with the probe at a lower concentration of 10 μg/mL. The cell phenotype and function of DCs were also investigated by fluorescence-activated cell sorting analysis and mixed leukocyte reactivity assay. Labeled DCs were injected into the footpad of C57BL/6 mice. Afterward, magnetic resonance imaging, NIRF imaging, Perls staining and CD11c immunofluorescence were used to observe the migration of the labeled DCs into draining LNs. RESULTS The synthetic SPIO-NIR797 nano-probe had a desirable superparamagnetic and near-infrared behavior. Perls staining showed perfect labeling efficiency. The cell phenotypes, including CD11c, CD80, CD86 and major histocompatibility complex class II, as well as the T-cell activation potential of the mature DCs were insignificantly affected after incubation (P > 0.05). Labeled DCs migrating into LNs could be detected by both magnetic resonance imaging and NIRF imaging simultaneously, which was further confirmed by Perls staining and immunofluorescence. CONCLUSIONS The novel dual-modality SPIO-NIR797 nano-probe has highly biocompatible characteristics for labeling and tracking DCs, which can be used to evaluate cancer immunotherapy in clinical applications.
Collapse
Affiliation(s)
- Yu-Chen Chen
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Song Wen
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Song-An Shang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Ying Cui
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Bing Luo
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China.
| |
Collapse
|
9
|
Su H, Mou Y, An Y, Han W, Huang X, Xia G, Ni Y, Zhang Y, Ma J, Hu Q. The migration of synthetic magnetic nanoparticle labeled dendritic cells into lymph nodes with optical imaging. Int J Nanomedicine 2013; 8:3737-44. [PMID: 24124362 PMCID: PMC3795011 DOI: 10.2147/ijn.s52135] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background The successful biotherapy of carcinoma with dendritic cell (DC) vaccines pivotally relies on DCs’ migratory capability into lymph tissues and activation of T cells. Accurate imaging and evaluation of DC migration in vivo have great significance during antitumor treatment with DC vaccine. We herein examined the behavior of DCs influenced by synthetic superparamagnetic iron oxide (SPIO) nanoparticle labeling. Methods γ-Fe2O3 nanoparticles were prepared and DCs, which were induced from bone marrow monocytes of enhanced green fluorescent protein (EGFP) transgenic mice, were labeled. The endocytosis of the SPIO, surface molecules, cell apoptosis and fluorescence intensity of EGFP-DCs were displayed by Prussian blue staining and flow cytometry (FCM), respectively. After EGFP-DCs, labeled with SPIO, were injected into footpads (n = 5) for 24 hours, the mice were examined in vivo by optical imaging (OPI). Meanwhile, confocal imaging and FCM were applied, respectively, to detect the migration of labeled DCs into draining lymph nodes. Results Nearly 100% of cells were labeled by the SPIO, in which the intracellular blue color gradually deepened and the iron contents rose with the increase of labeling iron concentrations. In addition, cell apoptosis and the surface molecules on DCs were at similar levels after SPIO labeling. After confirming that the fluorescence intensity of EGFP on DCs was not influenced by SPIO, the homing ability of EGFP-DCs labeled with SPIO displayed that the fluorescence intensity and the ratios of EGFP-DCs in draining lymph nodes were gradually decreased with the increase of labeling iron concentrations. Conclusion The synthetic SPIO nanoparticles possess perfect labeling ability and biocompatibility. Moreover, DCs labeled with a low dose of SPIO showed stronger migratory capability in vivo.
Collapse
Affiliation(s)
- Hang Su
- Center Laboratory of Stomatology, Stomatological Hospital Affiliated Medical School, Nanjing University, Nanjing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Sagar D, Foss C, El Baz R, Pomper MG, Khan ZK, Jain P. Mechanisms of dendritic cell trafficking across the blood-brain barrier. J Neuroimmune Pharmacol 2012; 7:74-94. [PMID: 21822588 PMCID: PMC3276728 DOI: 10.1007/s11481-011-9302-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 07/25/2011] [Indexed: 12/14/2022]
Abstract
Although the central nervous system (CNS) is considered to be an immunoprivileged site, it is susceptible to a host of autoimmune as well as neuroinflammatory disorders owing to recruitment of immune cells across the blood-brain barrier into perivascular and parenchymal spaces. Dendritic cells (DCs), which are involved in both primary and secondary immune responses, are the most potent immune cells in terms of antigen uptake and processing as well as presentation to T cells. In light of the emerging importance of DC traficking into the CNS, these cells represent good candidates for targeted immunotherapy against various neuroinflammatory diseases. This review focuses on potential physiological events and receptor interactions between DCs and the microvascular endothelial cells of the brain as they transmigrate into the CNS during degeneration and injury. A clear understanding of the underlying mechanisms involved in DC migration may advance the development of new therapies that manipulate these mechanistic properties via pharmacologic intervention. Furthermore, therapeutic validation should be in concurrence with the molecular imaging techniques that can detect migration of these cells in vivo. Since the use of noninvasive methods to image migration of DCs into CNS has barely been explored, we highlighted potential molecular imaging techniques to achieve this goal. Overall, information provided will bring this important leukocyte population to the forefront as key players in the immune cascade in the light of the emerging contribution of DCs to CNS health and disease.
Collapse
Affiliation(s)
- Divya Sagar
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Catherine Foss
- Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Rasha El Baz
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Martin G. Pomper
- Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Zafar K. Khan
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Pooja Jain
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
- Department of Microbiology & Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, 3805 Old Easton Road, Doylestown, PA 18902, USA
| |
Collapse
|
11
|
Mou Y, Hou Y, Chen B, Hua Z, Zhang Y, Xie H, Xia G, Wang Z, Huang X, Han W, Ni Y, Hu Q. In vivo migration of dendritic cells labeled with synthetic superparamagnetic iron oxide. Int J Nanomedicine 2011; 6:2633-40. [PMID: 22114494 PMCID: PMC3218577 DOI: 10.2147/ijn.s24307] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Successful treatment of cancer with dendritic cell tumor vaccine is highly dependent on how effectively the vaccine migrates into lymph nodes and activates T cells. In this study, a simple method was developed to trace migration of dendritic cells to lymph nodes. METHODS Superparamagnetic iron oxide (SPIO) of γ-Fe(2)O(3) nanoparticles were prepared to label dendritic cells generated from bone marrow of enhanced green fluorescent protein (EGFP) transgenic mice, to explore the fluorescence intensity of EGFP influenced by the SPIO, and to make images of labeled dendritic cells with the help of magnetic resonance imaging in vitro. The SPIO-EGFP-labeled dendritic cells were injected into the footpads of five mice. After 48 hours, magnetic resonance imaging, optical imaging, confocal imaging, and Prussian blue staining were used to confirm migration of the SPIO-EGFP-labeled dendritic cells into draining lymph nodes. RESULTS The synthetic SPIO nanoparticles had a spherical shape and desirable superparamagnetism, and confocal imaging and Prussian blue staining showed perfect labeling efficiency as well. Furthermore, the dendritic cells dual-labeled by SPIO and EGFP could migrate into lymph nodes after footpad injection, and could be detected by both magnetic resonance imaging and optical imaging simultaneously, which was further confirmed by immunohistochemistry and Prussian blue staining. The percentage of dendritic cells migrated to the draining lymph nodes was about 4%. CONCLUSION Synthetic SPIO nanoparticles are strong contrast agents with good biocompatibility, and EGFP transgenic dendritic cells can be labeled efficiently by SPIO, which are suitable for further study of the migratory behavior and biodistribution of dendritic cells in vivo.
Collapse
Affiliation(s)
- Yongbin Mou
- Central Laboratory of Stomatology, Stomatological Hospital Affiliated Medical School, Nanjing University, Nanjing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ju S, Qiu Y, Li C, Teng GJ, Ni Y. Multimodality Imaging of Endothelial Progenitor Cells with a Novel Multifunctional Probe Featuring Positive Magnetic Resonance Contrast and Near-Infrared Fluorescence. Mol Imaging 2011; 10:359-69. [PMID: 21521551 DOI: 10.2310/7290.2010.00055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Accepted: 08/09/2010] [Indexed: 11/18/2022] Open
Affiliation(s)
- Shenghong Ju
- From the Laboratory of Molecular Imaging, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China; School of Pharmacy, Fudan University, Shanghai, China; and Department of Radiology, University Hospitals, Catholic University of Leuven, Leuven, Belgium
| | - Yiru Qiu
- From the Laboratory of Molecular Imaging, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China; School of Pharmacy, Fudan University, Shanghai, China; and Department of Radiology, University Hospitals, Catholic University of Leuven, Leuven, Belgium
| | - Cong Li
- From the Laboratory of Molecular Imaging, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China; School of Pharmacy, Fudan University, Shanghai, China; and Department of Radiology, University Hospitals, Catholic University of Leuven, Leuven, Belgium
| | - Gao-Jun Teng
- From the Laboratory of Molecular Imaging, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China; School of Pharmacy, Fudan University, Shanghai, China; and Department of Radiology, University Hospitals, Catholic University of Leuven, Leuven, Belgium
| | - Yicheng Ni
- From the Laboratory of Molecular Imaging, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China; School of Pharmacy, Fudan University, Shanghai, China; and Department of Radiology, University Hospitals, Catholic University of Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Nolting DD, Gore JC, Pham W. NEAR-INFRARED DYES: Probe Development and Applications in Optical Molecular Imaging. Curr Org Synth 2011; 8:521-534. [PMID: 21822405 DOI: 10.2174/157017911796117223] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The recent emergence of optical imaging has brought forth a unique challenge for chemists: development of new biocompatible dyes that fluoresce in the near-infrared (NIR) region for optimal use in biomedical applications. This review describes the synthesis of NIR dyes and the design of probes capable of noninvasively imaging molecular events in small animal models.
Collapse
Affiliation(s)
- Donald D Nolting
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, 1161 21 Avenue South, Medical Center North, AA-1105, Nashville, TN 37232-2310
| | | | | |
Collapse
|
14
|
Induction of antitumor immunity by dendritic cells loaded with membrane-translocating mucin 1 Peptide antigen. Transl Oncol 2011; 4:1-8. [PMID: 21286372 DOI: 10.1593/tlo.10166] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 10/14/2010] [Accepted: 10/18/2010] [Indexed: 01/20/2023] Open
Abstract
To investigate the role of enhanced antigen presentation in dendritic cell (DC)-based immunotherapy. Here, we describe the development of a cell-penetrating mucin 1 (MUC1) antigen and its immunotherapeutic potential against tumors. After animal groups received two immunizations of MUC1-MPA(11)P-pulsed DCs, we observed a marked tumor regression compared with the mice treated with DCs alone or DCs pulsed with MUC1 peptide. We confirmed the migration and homing of DCs in the popliteal lymph node using magnetic resonance imaging during the study. In summary, enhanced antigen uptake using an MPA(11)P delivery molecule improves cell therapy.
Collapse
|
15
|
Mackay PS, Kremers GJ, Kobukai S, Cobb JG, Kuley A, Rosenthal SJ, Koktysh DS, Gore JC, Pham W. Multimodal imaging of dendritic cells using a novel hybrid magneto-optical nanoprobe. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:489-96. [PMID: 21215329 DOI: 10.1016/j.nano.2010.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 12/14/2010] [Accepted: 12/19/2010] [Indexed: 11/29/2022]
Abstract
UNLABELLED A transfecting agent-coated hybrid imaging nanoprobe (HINP) composed of visible and near-infrared (NIR) light emitting quantum dots (QDs) tethered to superparamagnetic iron oxide (SPIO) nanoparticles was developed. The surface modification of QDs and SPIO particles and incorporation of dual QDs within the SPIO were characterized by dynamic light scattering (DLS), quartz crystal microbalance (QCM) analysis and atomic force microscopy (AFM). The optical contrasting properties of HINP were characterized by absorption and photoluminescence spectroscopy and fluorescence imaging. Multicolor HINP was used in imaging the migration of dendritic cells (DCs) by optical, two-photon and magnetic resonance imaging techniques. FROM THE CLINICAL EDITOR The development of a transfecting agent-coated hybrid imaging nanoprobe (HINP) composed of visible and near-infrared light emitting quantum dots (QDs) tethered to superparamagnetic iron oxide nanoparticles is reported in this paper. Multicolor HINP was used in imaging the migration of dendritic cells by optical, two-photon and magnetic resonance imaging techniques.
Collapse
Affiliation(s)
- Patrick S Mackay
- Institute of Imaging Science, Vanderbilt University, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Montes-Worboys A, Brown S, Regev D, Bellew BF, Mohammed KA, Faruqi I, Sharma P, Moudgil B, Antony VB. Targeted delivery of amikacin into granuloma. Am J Respir Crit Care Med 2010; 182:1546-53. [PMID: 20693382 PMCID: PMC3029940 DOI: 10.1164/rccm.200912-1888oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 08/06/2010] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Nontuberculous mycobacterial (NTM) infection is a growing problem in the United States and remains underrecognized in the developing world. The management of NTM infections is further complicated by several factors, including the need to use high systemic doses of toxic agents, the length of therapy, and the development of drug resistance. OBJECTIVES We have evaluated the use of monocyte-derived dendritic cells (DCs) as a delivery vehicle for a luminescent derivative of amikacin prepared by conjugation to fluorescein isothiocyanate (FITC) (amikacin-FITC) into granulomas formed in the tissues of mice infected with Mycobacterium avium. METHODS Amikacin-FITC was prepared and quantitative fluorescence was used to track the intracellular uptake of this modified antibiotic. The antibiotic activity of amikacin-FITC was also determined to be comparable to unmodified amikacin against M. avium. Amikacin-FITC-loaded DCs were first primed with M. avium, and then the cells were injected into the tail vein of infected mice. After 24 hours, the mice were sacrificed and the tissues were analyzed under fluorescence microscope. MEASUREMENTS AND MAIN RESULTS We found that we were able to deliver amikacin into granulomas in a mouse model of disseminated mycobacterial infection. No increase in levels of monocyte chemoattractant protein-1 and its CCR2 as markers of inflammation were found when DCs were treated with amikacin-FITC. CONCLUSIONS DC-based drug delivery may be an adjunct and useful method of delivering high local concentrations of antibiotics into mycobacterial granulomas.
Collapse
Affiliation(s)
- Ana Montes-Worboys
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Scott Brown
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Doron Regev
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Brendan F. Bellew
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Kamal A. Mohammed
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Ibrahim Faruqi
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Parvesh Sharma
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Brij Moudgil
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Veena B. Antony
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, Particle Engineering Research Center, Center for Nano-Bio Sensors, and Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| |
Collapse
|
17
|
Lee JS, Tung CH. Lipo-oligoarginines as effective delivery vectors to promote cellular uptake. MOLECULAR BIOSYSTEMS 2010; 6:2049-55. [PMID: 20694264 DOI: 10.1039/c004684a] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
An effective cellular delivery vector with enhanced intracellular retention was developed by conjugating a cell-penetrating peptide (CPP) with a fatty acid chain. The optimized lipopeptide (LP), myristoylated hendecaarginine (C14R11), penetrated the cell membrane with high efficiency, and achieved superior metabolic stability and versatility as compared with unmodified oligoarginine CPPs, offering no adverse effect on cell viability and function. Cellular uptake, intracellular localization, cytotoxicity, and release kinetics of oligoarginines and LPs were investigated using flow cytometry analysis, cytotoxicity assay, and confocal microscopy. The cellular uptake efficiency and intracellular metabolic stability of C14R11 LP was further enhanced by replacing the L-arginine residues with D-arginine isomers. The cellular uptake and intracellular metabolic stability of D-form C14R11 (C14dR11) was significantly increased without any noticeable cytotoxicity compared to the unmodified parent hepta-arginine CPP or L-arginine LPs.
Collapse
Affiliation(s)
- Jae Sam Lee
- Department of Radiology, The Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | | |
Collapse
|
18
|
Kobukai S, Baheza R, Cobb JG, Virostko J, Xie J, Gillman A, Koktysh D, Kerns D, Does M, Gore JC, Pham W. Magnetic nanoparticles for imaging dendritic cells. Magn Reson Med 2010; 63:1383-90. [PMID: 20432309 DOI: 10.1002/mrm.22313] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We report the development of superparamagnetic iron oxide (SPIOs) nanoparticles and investigate the migration of SPIO-labeled dendritic cells (DCs) in a syngeneic mouse model using magnetic resonance (MR) imaging. The size of the dextran-coated SPIO is roughly 30 nm, and the DCs are capable of independent uptake of these particles, although not at levels comparable to particle uptake in the presence of a transfecting reagent. On average, with the assistance of polylysine, the particles were efficiently delivered inside DCs within one hour of incubation. The SPIO particles occupy approximately 0.35% of cell surface and are equivalent to 34.6 pg of iron per cell. In vivo imaging demonstrated that the labeled cells migrated from the injection site in the footpad to the corresponding popliteal lymph node. The homing of labeled cells in the lymph nodes resulted in a signal drop of up to 79%. Furthermore, labeling DCs with SPIO particles did not compromise cell function, we demonstrated that SPIO-enhanced MR imaging can be used to track the migration of DCs effectively in vivo.
Collapse
Affiliation(s)
- Saho Kobukai
- Vanderbilt University, Institute of Imaging Science, Nashville, Tennessee 37232-2310, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
The War on Cancer rages on. Neoplasia 2010; 11:1252-63. [PMID: 20019833 DOI: 10.1593/neo.91866] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 11/03/2009] [Accepted: 11/03/2009] [Indexed: 02/08/2023] Open
Abstract
In 1971, the "War on Cancer" was launched by the US government to cure cancer by the 200-year anniversary of the founding of the United States of America, 1976. This article briefly looks back at the progress that has been made in cancer research and compares progress made in other areas of human affliction. While progress has indeed been made, the battle continues to rage on.
Collapse
|
20
|
Wang Q, Ornstein M, Kaufman HL. Imaging the immune response to monitor tumor immunotherapy. Expert Rev Vaccines 2009; 8:1427-37. [PMID: 19803763 DOI: 10.1586/erv.09.100] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The goal of cancer immunotherapy is to promote antitumor immunity, and novel approaches include vaccination, adoptive transfer of tumor-reactive T cells, and administration of monoclonal antibodies and small molecules that target immune regulatory pathways. The molecular and cellular events responsible for tumor rejection are not completely defined and correlative studies have been used to help understand the mechanisms and extent of immune activation and tumor regression with these approaches. The real-time monitoring of immune responses to immunotherapy has been challenging as specific cell subsets may be difficult to define, and molecular pathways have evolved functionally diverse outcomes in different cells and in different tissues. Recently, improvements in optics and digital imaging have led to novel imaging techniques that make it possible to track the migration of individual immune cells ex vivo and in vivo, and to detect the dynamic interactions between T cells and antigen-presenting cells or tumor cells within complex microenvironments, including lymphoid tissue and established tumors. This review will explain some of the more established imaging techniques and discuss their role in monitoring the immune response in patients treated with various tumor immunotherapy approaches.
Collapse
Affiliation(s)
- Qin Wang
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY, USA.
| | | | | |
Collapse
|
21
|
Neoplasia: the second decade. Neoplasia 2009; 10:1314-24. [PMID: 19048110 DOI: 10.1593/neo.81372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 10/27/2008] [Accepted: 10/27/2008] [Indexed: 12/30/2022] Open
Abstract
This issue marks the end of the 10-year anniversary of Neoplasia where we have seen exciting growth in both number of submitted and published articles in Neoplasia. Neoplasia was first published in 1999. During the past 10 years, Neoplasia has dynamically adapted to the needs of the cancer research community as technologies have advanced. Neoplasia is currently providing access to articles through PubMed Central to continue to facilitate rapid broad-based dissemination of published findings to the scientific community through an Open Access model. This has in part helped Neoplasia to achieve an improved impact factor this past year, demonstrating that the manuscripts published by Neoplasia are of great interest to the overall cancer research community. This past year, Neoplasia received a record number of articles for review and has had a 21% increase in the number of published articles.
Collapse
|
22
|
Pham W, Kobukai S, Hotta C, Gore JC. Dendritic cells: therapy and imaging. Expert Opin Biol Ther 2009; 9:539-64. [DOI: 10.1517/14712590902867739] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Wellington Pham
- Vanderbilt University, Institute of Imaging Science, 1161 21st Avenue South, AA. 1105 MCN, Nashville, TN 37232-2310, USA
| | - Saho Kobukai
- Vanderbilt University, Institute of Imaging Science, 1161 21st Avenue South, AA. 1105 MCN, Nashville, TN 37232-2310, USA
- *These individuals contributed equally to this work
| | - Chie Hotta
- Brigham and Women's Hospital, Harvard Medical School, Center for Neurologic Diseases, 77 Avenue Louis Pasteur, HIM 780, Boston, MA 02115, USA
- *These individuals contributed equally to this work
| | - John C Gore
- Vanderbilt University, Institute of Imaging Science, 1161 21st Avenue South, AA. 1105 MCN, Nashville, TN 37232-2310, USA
| |
Collapse
|
23
|
Glucosamine-bound near-infrared fluorescent probes with lysosomal specificity for breast tumor imaging. Neoplasia 2008; 10:389-98. [PMID: 18392136 DOI: 10.1593/neo.07856] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 02/15/2008] [Accepted: 02/15/2008] [Indexed: 01/18/2023] Open
Abstract
Noninvasive imaging of lysosomes will be useful 1) to elucidate the role of lysosomal parameters in cancer, 2) to diagnose malignant lesions, and 3) to evaluate future lysosome-targeted anticancer therapies. Lysosome-specific labeling of glucosamine-bound near-infrared (NIR) fluorescent probes, IR-1 and IR-2, but not control probe IR-15 without the glucosamine moiety, was observed by fluorescence microscopy in human breast epithelial cell lines. Lysosome labeling and tumor specificity of these NIR probes were investigated by dynamic optical imaging and immunofluorescence staining in human breast tumor xenografts. IR-1 and IR-2 demonstrated faster lysosome labeling rates in highly aggressive MDA-MB-231 and MDA-MB-435 cells compared with less aggressive MCF-7 and nontumorigenic MCF-12A cells. IR-1 and IR-2, but not IR-15, accumulated in human MDA-MB-231, MDA-MB-435, and MCF-7 breast tumor xenografts in vivo. IR-2 demonstrated the highest maximum fluorescence and tumor/normal tissue ratios in all tumor models. Specific lysosome labeling from IR-2 in vivo was validated by colocalization of the NIR fluorescence with CD63 immunofluorescence in tumor sections. IR-1 and IR-2 demonstrated high lysosome-labeling ability and breast tumor-targeting specificity in vitro and in vivo. They are promising for diagnosing malignant lesions and may provide a means for evaluating and monitoring future lysosome-targeted anticancer therapies.
Collapse
|
24
|
Neoplasia: An Anniversary of Progress. Neoplasia 2007. [DOI: 10.1593/neo.07968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|