1
|
Shu Y, Shen H, Yao M, Shen J, Yang G, Chen H, Tang Y, Ma M. Metal protoporphyrin-induced self-assembly nanoprobe enabling precise tracking and antioxidant protection of stem cells for ischemic stroke therapy. SMART MEDICINE 2023; 2:e20220037. [PMID: 39188561 PMCID: PMC11236039 DOI: 10.1002/smmd.20220037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/03/2023] [Indexed: 08/28/2024]
Abstract
Mesenchymal stem cell (MSC)-based therapy has provided a promising strategy for the treatment of ischemic stroke, which is still restricted by the lack of long-term cell tracking strategy as well as the poor survival rate of stem cells in ischemic region. Herein, a dual-functional nanoprobe, cobalt protoporphyrin-induced nano-self-assembly (CPSP), has been developed through a cobalt protoporphyrin IX (CoPP) aggregation-induced self-assembly strategy, which combines CoPP and superparamagnetic iron oxide (SPION) via a simple solvent evaporation-driven method. Without any additional carrier materials, the obtained CPSP is featured with good biocompatibility and high proportions of active ingredients. The SPIONs in CPSPs form a cluster-like structure, endowing this nano-self-assembly with excellent T2-weighted magnetic resonance (MR) imaging performance. Furthermore, the CoPP released from CPSPs could effectively protect MSCs by upregulating heme oxygenase 1 (HO-1) expression. The in vivo cell tracing capacity of CPSPs is confirmed by monitoring the migration of labeled MSCs with MR imaging in a middle cerebral artery occlusion mouse model. More importantly, the sustained release of CoPP from CPSPs improves the survival of transplanted MSCs and promotes neural repair and neurobehavioral recovery of ischemic mice. Overall, this work presents a novel dual-functional nanoagent with an ingenious design for advancing MSC-based therapy.
Collapse
Affiliation(s)
- Yimeng Shu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesShanghaiChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
| | - Hui Shen
- Med‐X Research Institute and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Minghua Yao
- Department of UltrasoundShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jie Shen
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesShanghaiChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
| | - Guo‐Yuan Yang
- Med‐X Research Institute and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Hangrong Chen
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesShanghaiChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
- School of Chemistry and Materials ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhouChina
| | - Yaohui Tang
- Med‐X Research Institute and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- Ankerui (Shanxi) Biological Cell Co., Ltd.Xiaohe Industrial Park Comprehensive Reform Demonstration ZoneTaiyuanChina
| | - Ming Ma
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesShanghaiChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
- School of Chemistry and Materials ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhouChina
| |
Collapse
|
2
|
Recent Progress on Heparin–Protamine Particles for Biomedical Application. Polymers (Basel) 2022; 14:polym14050932. [PMID: 35267754 PMCID: PMC8912589 DOI: 10.3390/polym14050932] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Biomolecules are attractive building blocks with self-assembly ability, structural diversity, and excellent functionality for creating artificial materials. Heparin and protamine, a clinically relevant pair of biomolecules used in cardiac and vascular surgery, have been shown to coassemble into particulate polyelectrolyte complexes in vitro. The resulting heparin–protamine particles exhibit adhesive properties that enable advantageous interactions with proteins, cells, and various other substances and have been employed as functional materials for biomedical applications. In this review article, we summarize recent progress in research on the use of heparin–protamine particles as drug carriers, cell adhesives, and cell labels. Studies have demonstrated that heparin–protamine particles are potentially versatile in biomedical fields from drug delivery and regenerative medicine to plastic surgery.
Collapse
|
3
|
Gabashvili AN, Chmelyuk NS, Efremova MV, Malinovskaya JA, Semkina AS, Abakumov MA. Encapsulins-Bacterial Protein Nanocompartments: Structure, Properties, and Application. Biomolecules 2020; 10:biom10060966. [PMID: 32604934 PMCID: PMC7355545 DOI: 10.3390/biom10060966] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
Recently, a new class of prokaryotic compartments, collectively called encapsulins or protein nanocompartments, has been discovered. The shell proteins of these structures self-organize to form icosahedral compartments with a diameter of 25-42 nm, while one or more cargo proteins with various functions can be encapsulated in the nanocompartment. Non-native cargo proteins can be loaded into nanocompartments and the surface of the shells can be further functionalized, which allows for developing targeted drug delivery systems or using encapsulins as contrast agents for magnetic resonance imaging. Since the genes encoding encapsulins can be integrated into the cell genome, encapsulins are attractive for investigation in various scientific fields, including biomedicine and nanotechnology.
Collapse
Affiliation(s)
- Anna N. Gabashvili
- Laboratory “Biomedical Nanomaterials”, National University of Science and Technology “MISiS”, Leninskiy Prospect, 4, 119049 Moscow, Russia; (A.N.G.); (N.S.C.)
- Department of Medical Nanobiotechnoilogy, Pirogov Russian National Research Medical University, Ostrovityanova st, 1, 117997 Moscow, Russia;
| | - Nelly S. Chmelyuk
- Laboratory “Biomedical Nanomaterials”, National University of Science and Technology “MISiS”, Leninskiy Prospect, 4, 119049 Moscow, Russia; (A.N.G.); (N.S.C.)
| | - Maria V. Efremova
- Department of Nuclear Medicine, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany;
- Institute of Biological and Medical Imaging and Institute of Developmental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | | | - Alevtina S. Semkina
- Department of Medical Nanobiotechnoilogy, Pirogov Russian National Research Medical University, Ostrovityanova st, 1, 117997 Moscow, Russia;
| | - Maxim A. Abakumov
- Laboratory “Biomedical Nanomaterials”, National University of Science and Technology “MISiS”, Leninskiy Prospect, 4, 119049 Moscow, Russia; (A.N.G.); (N.S.C.)
- Department of Medical Nanobiotechnoilogy, Pirogov Russian National Research Medical University, Ostrovityanova st, 1, 117997 Moscow, Russia;
- Correspondence: ; Tel.: +7-903-586-4777
| |
Collapse
|
4
|
Abstract
Regenerative medicine with the use of stem cells has appeared as a potential therapeutic alternative for many disease states. Despite initial enthusiasm, there has been relatively slow transition to clinical trials. In large part, numerous questions remain regarding the viability, biology and efficacy of transplanted stem cells in the living subject. The critical issues highlighted the importance of developing tools to assess these questions. Advances in molecular biology and imaging have allowed the successful non-invasive monitoring of transplanted stem cells in the living subject. Over the years these methodologies have been updated to assess not only the viability but also the biology of transplanted stem cells. In this review, different imaging strategies to study the viability and biology of transplanted stem cells are presented. Use of these strategies will be critical as the different regenerative therapies are being tested for clinical use.
Collapse
Affiliation(s)
- Fakhar Abbas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joseph C. Wu
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
- Department of Medicine (Cardiology), Stanford University, Stanford, CA, USA
| | - Sanjiv Sam Gambhir
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
- Department of Bio-Engineering, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
5
|
Mitochondrial fission protein 1 up-regulation ameliorates senescence-related endothelial dysfunction of human endothelial progenitor cells. Angiogenesis 2019; 22:569-582. [DOI: 10.1007/s10456-019-09680-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/26/2019] [Indexed: 12/15/2022]
|
6
|
The function and magnetic resonance imaging of immature dendritic cells under ultrasmall superparamagnetic iron oxide (USPIO)-labeling. Biotechnol Lett 2017; 39:1079-1089. [DOI: 10.1007/s10529-017-2332-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 03/29/2017] [Indexed: 10/19/2022]
|
7
|
Goodfellow F, Simchick GA, Mortensen LJ, Stice SL, Zhao Q. Tracking and Quantification of Magnetically Labeled Stem Cells using Magnetic Resonance Imaging. ADVANCED FUNCTIONAL MATERIALS 2016; 26:3899-3915. [PMID: 28751853 PMCID: PMC5526633 DOI: 10.1002/adfm.201504444] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Stem cell based therapies have critical impacts on treatments and cures of diseases such as neurodegenerative or cardiovascular disease. In vivo tracking of stem cells labeled with magnetic contrast agents is of particular interest and importance as it allows for monitoring of the cells' bio-distribution, viability, and physiological responses. Herein, recent advances are introduced in tracking and quantification of super-paramagnetic iron oxide (SPIO) nanoparticles-labeled cells with magnetic resonance imaging, a noninvasive approach that can longitudinally monitor transplanted cells. This is followed by recent translational research on human stem cells that are dual-labeled with green fluorescence protein (GFP) and SPIO nanoparticles, then transplanted and tracked in a chicken embryo model. Cell labeling efficiency, viability, and cell differentiation are also presented.
Collapse
Affiliation(s)
| | - Gregory A Simchick
- Bioimaging Research Center, Regenerative Bioscience Center, and Department of Physics University of Georgia, Athens, GA. 30602, USA
| | | | | | - Qun Zhao
- Bioimaging Research Center, Regenerative Bioscience Center, and Department of Physics University of Georgia, Athens, GA. 30602, USA
| |
Collapse
|
8
|
Chung T, Youn H, Yeom CJ, Kang KW, Chung JK. Glycosylation of Sodium/Iodide Symporter (NIS) Regulates Its Membrane Translocation and Radioiodine Uptake. PLoS One 2015; 10:e0142984. [PMID: 26599396 PMCID: PMC4658105 DOI: 10.1371/journal.pone.0142984] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/29/2015] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Human sodium/iodide symporter (hNIS) protein is a membrane glycoprotein that transports iodide ions into thyroid cells. The function of this membrane protein is closely regulated by post-translational glycosylation. In this study, we measured glycosylation-mediated changes in subcellular location of hNIS and its function of iodine uptake. METHODS HeLa cells were stably transfected with hNIS/tdTomato fusion gene in order to monitor the expression of hNIS. Cellular localization of hNIS was visualized by confocal microscopy of the red fluorescence of tdTomato. The expression of hNIS was evaluated by RT-PCR and immunoblot analysis. Functional activity of hNIS was estimated by radioiodine uptake. Cyclic AMP (cAMP) and tunicamycin were used to stimulate and inhibit glycosylation, respectively. In vivo images were obtained using a Maestro fluorescence imaging system. RESULTS cAMP-mediated Glycosylation of NIS resulted in increased expression of hNIS, stimulating membrane translocation, and enhanced radioiodine uptake. In contrast, inhibition of glycosylation by treatment with tunicamycin dramatically reduced membrane translocation of intracellular hNIS, resulting in reduced radioiodine uptake. In addition, our hNIS/tdTomato fusion reporter successfully visualized cAMP-induced hNIS expression in xenografted tumors from mouse model. CONCLUSIONS These findings clearly reveal that the membrane localization of hNIS and its function of iodine uptake are glycosylation-dependent, as our results highlight enhancement of NIS expression and glycosylation with subsequent membrane localization after cAMP treatment. Therefore, enhancing functional NIS by the increasing level of glycosylation may be suggested as a promising therapeutic strategy for cancer patients who show refractory response to conventional radioiodine treatment.
Collapse
Affiliation(s)
- Taemoon Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
- Cancer Imaging Center, Seoul National University Hospital, Seoul, Korea
- * E-mail: (HY); (JKC)
| | - Chan Joo Yeom
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
- * E-mail: (HY); (JKC)
| |
Collapse
|
9
|
MG G. Intracavernous Administration of Adipose Stem Cells: A New Technique of Treating Erectile Dysfunction in Diabetic Patient, Preliminary Report of 6 Cases. ACTA ACUST UNITED AC 2015. [DOI: 10.15406/mojcsr.2015.02.00018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
10
|
Insulin-producing cells from embryonic stem cells rescues hyperglycemia via intra-spleen migration. Sci Rep 2014; 4:7586. [PMID: 25533571 PMCID: PMC4274503 DOI: 10.1038/srep07586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 11/24/2014] [Indexed: 02/07/2023] Open
Abstract
Implantation of embryonic stem cells (ESC)-derived insulin-producing cells has been extensively investigated for treatment of diabetes in animal models. However, the in vivo behavior and migration of transplanted cells in diabetic models remains unclear. Here we investigated the location and migration of insulin-producing cells labeled with superparamagnetic iron oxide (SPIO) using a dynamic MRI tracking method. SPIO labeled cells showed hypointense signal under the kidney subcapsules of diabetic mice on MRI, and faded gradually over the visiting time. However, new hypointense signal appeared in the spleen 1 week after transplantation, and became obvious with the time prolongation. Further histological examination proved the immigrated cells were insulin and C-peptide positive cells which were evenly distributed throughout the spleen. These intra-spleen insulin-producing cells maintained their protective effects against hyperglycemia in vivo, and these effects were reversed upon spleen removal. Transplantation of insulin-producing cells through spleen acquired an earlier blood glucose control as compared with that through kidney subcapsules. In summary, our data demonstrate that insulin-producing cells transplanted through kidney subcapsules were not located in situ but migrated into spleen, and rescues hyperglycemia in diabetic models. MRI may provide a novel tracking method for preclinical cell transplantation therapy of diabetes continuously and non-invasively.
Collapse
|
11
|
Kim MH, Woo SK, Lee KC, An GI, Pandya D, Park NW, Nahm SS, Eom KD, Kim KI, Lee TS, Kim CW, Kang JH, Yoo J, Lee YJ. Longitudinal monitoring adipose-derived stem cell survival by PET imaging hexadecyl-4-¹²⁴I-iodobenzoate in rat myocardial infarction model. Biochem Biophys Res Commun 2014; 456:13-9. [PMID: 25446095 DOI: 10.1016/j.bbrc.2014.11.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 11/08/2014] [Indexed: 01/09/2023]
Abstract
This study aims to monitor how the change of cell survival of transplanted adipose-derived stem cells (ADSCs) responds to myocardial infarction (MI) via the hexadecyl-4-(124)I-iodobenzoate ((124)I-HIB) mediated direct labeling method in vivo. Stem cells have shown the potential to improve cardiac function after MI. However, monitoring of the fate of transplanted stem cells at target sites is still unclear. Rat ADSCs were labeled with (124)I-HIB, and radiolabeled ADSCs were transplanted into the myocardium of normal and MI model. In the group of (124)I-HIB-labeled ADSC transplantation, in vivo imaging was performed using small-animal positron emission tomography (PET)/computed tomography (CT) for 9 days. Twenty-one days post-transplantation, histopathological analysis and apoptosis assay were performed. ADSC viability and differentiation were not affected by (124)I-HIB labeling. In vivo tracking of the (124)I-HIB-labeled ADSCs was possible for 9 and 3 days in normal and MI model, respectively. Apoptosis of transplanted cells increased in the MI model compared than that in normal model. We developed a direct labeling agent, (124)I-HIB, and first tried to longitudinally monitor transplanted stem cell to MI. This approach may provide new insights on the roles of stem cell monitoring in living bodies for stem cell therapy from pre-clinical studies to clinical trials.
Collapse
Affiliation(s)
- Min Hwan Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Sang-Keun Woo
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Kyo Chul Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Gwang Il An
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Darpan Pandya
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Republic of Korea
| | - Noh Won Park
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sang-Soep Nahm
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ki Dong Eom
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Kwang Il Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Tae Sup Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Chan Wha Kim
- School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Joo Hyun Kang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jeongsoo Yoo
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Republic of Korea.
| | - Yong Jin Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Inoue K, Gibbs SL, Liu F, Lee JH, Xie Y, Ashitate Y, Fujii H, Frangioni JV, Choi HS. Microscopic validation of macroscopic in vivo images enabled by same-slide optical and nuclear fusion. J Nucl Med 2014; 55:1899-904. [PMID: 25324521 DOI: 10.2967/jnumed.114.141606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED It is currently difficult to determine the molecular and cellular basis for radioscintigraphic signals obtained during macroscopic in vivo imaging. The field is in need of technology that helps bridge the macroscopic and microscopic regimes. To solve this problem, we developed a fiducial marker (FM) simultaneously compatible with 2-color near-infrared (NIR) fluorescence (700 and 800 nm), autoradiography, and conventional hematoxylin-eosin (HE) histology. METHODS The FM was constructed from an optimized concentration of commercially available human serum albumin, 700- and 800-nm NIR fluorophores, (99m)Tc-pertechnetate, dimethyl sulfoxide, and glutaraldehyde. Lymphangioleiomyomatosis cells coexpressing the sodium iodide symporter and green fluorescent protein were labeled with 700-nm fluorophore and (99m)Tc-pertechnatate and then administered intratracheally into CD-1 mice. After in vivo SPECT imaging and ex vivo SPECT and NIR fluorescence imaging of the lungs, 30-μm frozen sections were prepared and processed for 800-nm NIR fluorophore costaining, autoradiography, and HE staining on the same slide using the FMs to coregister all datasets. RESULTS Optimized FMs, composed of 100 μM unlabeled human serum albumin, 1 μM NIR fluorescent human serum albumin, 15% dimethyl sulfoxide, and 3% glutaraldehyde in phosphate-buffered saline (pH 7.4), were prepared within 15 min, displayed homogeneity and stability, and were visible by all imaging modalities, including HE staining. Using these FMs, tissue displaying high signal by SPECT could be dissected and analyzed on the same slide and at the microscopic level for 700-nm NIR fluorescence, 800-nm NIR fluorescence, autoradiography, and HE histopathologic staining. CONCLUSION When multimodal FMs are combined with a new technique for simultaneous same-slide NIR fluorescence imaging, autoradiography, and HE staining, macroscopic in vivo images can now be studied unambiguously at the microscopic level.
Collapse
Affiliation(s)
- Kazumasa Inoue
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts Department of Radiological Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Summer L Gibbs
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Fangbing Liu
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Jeong Heon Lee
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Yang Xie
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Yoshitomo Ashitate
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Hirofumi Fujii
- Functional Imaging Division, Research Center for Innovative Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - John V Frangioni
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts; and Curadel, LLC, Worcester, Massachusetts
| | - Hak Soo Choi
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
13
|
Katsikis A, Koutelou M. Cardiac Stem Cell Imaging by SPECT and PET. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014. [DOI: 10.1007/s12410-014-9265-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Abstract
Advances in noninvasive imaging technologies that allow for in vivo dynamic monitoring of cells and cellular function in living research subjects have revealed new insights into cell biology in the context of intact organs and their native environment. In the field of hematopoiesis and stem cell research, studies of cell trafficking involved in injury repair and hematopoietic engraftment have made great progress using these new tools. Stem cells present unique challenges for imaging since after transplantation, they proliferate dramatically and differentiate. Therefore, the imaging modality used needs to have a large dynamic range, and the genetic regulatory elements used need to be stably expressed during differentiation. Multiple imaging technologies using different modalities are available, and each varies in sensitivity, ease of data acquisition, signal to noise ratios (SNR), substrate availability, and other parameters that affect utility for monitoring cell fates and function. For a given application, there may be several different approaches that can be used. For mouse models, clinically validated technologies such as magnetic resonance imaging (MRI) and positron emission tomography (PET) have been joined by optical imaging techniques such as in vivo bioluminescence imaging (BLI) and fluorescence imaging (FLI), and all have been used to monitor bone marrow and stem cells after transplantation into mice. Photoacoustic imaging that utilizes the sound created by the thermal expansion of absorbed light to generate an image best represents hybrid technologies. Each modality requires that the cells of interest be marked with a genetic reporter that acts as a label making them uniquely visible using that technology. For each modality, there are several labels to choose from. Multiple methods for applying these different labels are available. This chapter provides an overview of the imaging technologies and commonly used labels for each, as well as detailed protocols for gene delivery into hematopoietic cells for the purposes of applying these specific labels to cell trafficking. The goal of this chapter is to provide adequate background information to allow the design and implementation of an experimental system for in vivo imaging in mice.
Collapse
|
15
|
Zhou J, Yang F, Zhou Q, Yang K, Chen W. Magnetic resonance imaging tracking of ultra small superparamagnetic iron oxide labeled rabbit dendritic cells. Exp Biol Med (Maywood) 2013; 239:13-23. [PMID: 24227632 DOI: 10.1177/1535370213508712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) play an important role in atherosclerosis plaque formation, but the mechanism has not been elucidated clearly. This study is designed to establish a method for tracing DCs in vivo facilitating the investigation of the DCs' specific roles in atherosclerosis. Rabbit DCs labeled by different concentrations of ultra small superparamagnetic iron oxide (USPIO) were injected into atherosclerosis rabbit model and traced with magnetic resonance imaging (MRI). Results showed that USPIO labeling nearly have no cytotoxicity to DCs in low concentrations (<500 µg/mL) but induced some decrease of cell viability at high concentrations (>500 µg/mL). Moreover, USPIO labeling, from 200 to 2000 µg/mL, caused a dose-dependent decrease of the mitochondrial membrane potential in DCs. The high labeling concentration (2000 µg/mL) triggered necrosis instead of apoptosis in DCs. By T2WI and fs T2WI sequence imaging comparison, DCs were found to exist in rabbit abdominal artery plaques after 24 h of transplantation and in spleen after one week detected by Prussian blue staining of tissue sections. We concluded that about 200 µg/mL USPIO is ideal to effectively label DCs for MRI tracing in vivo without a threat to cell viability. Combining USPIO labeling and MRI to track the movement of injected DCs in vivo is a feasible method.
Collapse
Affiliation(s)
- Jing Zhou
- College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | | | | | | | | |
Collapse
|
16
|
Lacroix S, Egrise D, Van Simaeys G, Doumont G, Monclus M, Sherer F, Herbaux T, Leroy D, Goldman S. [18F]-FBEM, a tracer targeting cell-surface protein thiols for cell trafficking imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:409-16. [DOI: 10.1002/cmmi.1540] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 03/02/2013] [Accepted: 03/10/2013] [Indexed: 11/06/2022]
|
17
|
Roura S, Gálvez-Montón C, Bayes-Genis A. Bioluminescence imaging: a shining future for cardiac regeneration. J Cell Mol Med 2013; 17:693-703. [PMID: 23402217 PMCID: PMC3823173 DOI: 10.1111/jcmm.12018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/28/2012] [Indexed: 12/28/2022] Open
Abstract
Advances in bioanalytical techniques have become crucial for both basic research and medical practice. One example, bioluminescence imaging (BLI), is based on the application of natural reactants with light-emitting capabilities (photoproteins and luciferases) isolated from a widespread group of organisms. The main challenges in cardiac regeneration remain unresolved, but a vast number of studies have harnessed BLI with the discovery of aequorin and green fluorescent proteins. First described in the luminous hydromedusan Aequorea victoria in the early 1960s, bioluminescent proteins have greatly contributed to the design and initiation of ongoing cell-based clinical trials on cardiovascular diseases. In conjunction with advances in reporter gene technology, BLI provides valuable information about the location and functional status of regenerative cells implanted into numerous animal models of disease. The purpose of this review was to present the great potential of BLI, among other existing imaging modalities, to refine effectiveness and underlying mechanisms of cardiac cell therapy. We recount the first discovery of natural primary compounds with light-emitting capabilities, and follow their applications to bioanalysis. We also illustrate insights and perspectives on BLI to illuminate current efforts in cardiac regeneration, where the future is bright.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Spain
| | | | | |
Collapse
|
18
|
Reduction of connexin43 in human endothelial progenitor cells impairs the angiogenic potential. Angiogenesis 2013; 16:553-60. [PMID: 23354732 DOI: 10.1007/s10456-013-9335-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 01/15/2013] [Indexed: 10/27/2022]
Abstract
Our previous work showed that arsenic trioxide down-regulated Cx43 and attenuated the angiogenic potential of human late endothelial progenitor cells (EPC). However, the relation between Cx43 and angiogenic activity of the EPC remained unclear. In the study, human late EPC were treated with siRNA specific to Cx43 (Cx43siRNA). The expression profiles as well as activity of the treated cells were examined. In parallel, the angiogenic potential of human EPC treated with Cx43siRNA was evaluated using murine hind limb ischemic model. The results showed that, in the EPC treated with Cx43siRNA, the activity of migration, proliferation, and angiogenic potential were attenuated, accompanied by reduction in vascular endothelial growth factor (VEGF) expression. In hind limb ischemia mice, EPC treated with Cx43siRNA lost the therapeutic angiogenic potential. VEGF supplementation partially recovered the activity impaired by Cx43 down-regulation. In conclusion, reduced Cx43 expression per se in the EPC causes decreased expression of VEGF and impaired angiogenic potential of the cells. Prevention of Cx43 reduction is a potential target to maintain the angiogenic potential of the EPC.
Collapse
|
19
|
Rodriguez-Porcel M, Kronenberg MW, Henry TD, Traverse JH, Pepine CJ, Ellis SG, Willerson JT, Moyé LA, Simari RD. Cell tracking and the development of cell-based therapies: a view from the Cardiovascular Cell Therapy Research Network. JACC Cardiovasc Imaging 2012; 5:559-65. [PMID: 22595165 DOI: 10.1016/j.jcmg.2011.12.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 12/13/2011] [Accepted: 12/15/2011] [Indexed: 12/12/2022]
Abstract
Cell-based therapies are being developed for myocardial infarction (MI) and its consequences (e.g., heart failure) as well as refractory angina and critical limb ischemia. The promising results obtained in preclinical studies led to the translation of this strategy to clinical studies. To date, the initial results have been mixed: some studies showed benefit, whereas in others, no benefit was observed. There is a growing consensus among the scientific community that a better understanding of the fate of transplanted cells (e.g., cell homing and viability over time) will be critical for the long-term success of these strategies and that future studies should include an assessment of cell homing, engraftment, and fate as an integral part of the trial design. In this review, different imaging methods and technologies are discussed within the framework of the physiological answers that the imaging strategies can provide, with a special focus on the inherent regulatory issues.
Collapse
|
20
|
Templin C, Zweigerdt R, Schwanke K, Olmer R, Ghadri JR, Emmert MY, Müller E, Küest SM, Cohrs S, Schibli R, Kronen P, Hilbe M, Reinisch A, Strunk D, Haverich A, Hoerstrup S, Lüscher TF, Kaufmann PA, Landmesser U, Martin U. Transplantation and tracking of human-induced pluripotent stem cells in a pig model of myocardial infarction: assessment of cell survival, engraftment, and distribution by hybrid single photon emission computed tomography/computed tomography of sodium iodide symporter transgene expression. Circulation 2012; 126:430-9. [PMID: 22767659 DOI: 10.1161/circulationaha.111.087684] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Evaluation of novel cellular therapies in large-animal models and patients is currently hampered by the lack of imaging approaches that allow for long-term monitoring of viable transplanted cells. In this study, sodium iodide symporter (NIS) transgene imaging was evaluated as an approach to follow in vivo survival, engraftment, and distribution of human-induced pluripotent stem cell (hiPSC) derivatives in a pig model of myocardial infarction. METHODS AND RESULTS Transgenic hiPSC lines stably expressing a fluorescent reporter and NIS (NIS(pos)-hiPSCs) were established. Iodide uptake, efflux, and viability of NIS(pos)-hiPSCs were assessed in vitro. Ten (±2) days after induction of myocardial infarction by transient occlusion of the left anterior descending artery, catheter-based intramyocardial injection of NIS(pos)-hiPSCs guided by 3-dimensional NOGA mapping was performed. Dual-isotope single photon emission computed tomographic/computed tomographic imaging was applied with the use of (123)I to follow donor cell survival and distribution and with the use of (99m)TC-tetrofosmin for perfusion imaging. In vitro, iodide uptake in NIS(pos)-hiPSCs was increased 100-fold above that of nontransgenic controls. In vivo, viable NIS(pos)-hiPSCs could be visualized for up to 15 weeks. Immunohistochemistry demonstrated that hiPSC-derived endothelial cells contributed to vascularization. Up to 12 to 15 weeks after transplantation, no teratomas were detected. CONCLUSIONS This study describes for the first time the feasibility of repeated long-term in vivo imaging of viability and tissue distribution of cellular grafts in large animals. Moreover, this is the first report demonstrating vascular differentiation and long-term engraftment of hiPSCs in a large-animal model of myocardial infarction. NIS(pos)-hiPSCs represent a valuable tool to monitor and improve current cellular treatment strategies in clinically relevant animal models.
Collapse
Affiliation(s)
- Christian Templin
- Department of Cardiology, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wu C, Ma G, Li J, Zheng K, Dang Y, Shi X, Sun Y, Li F, Zhu Z. In vivo cell tracking via ¹⁸F-fluorodeoxyglucose labeling: a review of the preclinical and clinical applications in cell-based diagnosis and therapy. Clin Imaging 2012. [PMID: 23206605 DOI: 10.1016/j.clinimag.2012.02.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The rising interest in using functional cells for diagnosis and treatment has created an urgent need for in vivo cell-tracking techniques. Certain advanced techniques, such as those involving reporter genes or nanoparticles, are still awaiting confirmation of their safety and feasibility in human patients. Tracking cells by labeling them with (18)F-fluorodeoxyglucose, a tracer clinically used in positron emission tomography (PET), may be one way to rapidly translate some of these principles from bench to bedside. The preliminary results are exciting, although further development, optimization, and validation are required. Here, several applications of the technique are surveyed: finding inflammatory foci, targeting cancer immunotherapies, tracking transplanted islet cells, and monitoring cardiac stem cells. Advantages, limitations, and prospects of the technique are discussed. These early experiences only highlight the existing need to improve cell-labeling techniques using PET tracers. This method may finally lead to the development of effective and convenient methods for clinical cell-tracking techniques involving PET/computed tomography.
Collapse
Affiliation(s)
- Chenxi Wu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Park BN, Shim W, Ahn YH, Lee JH, An YS, Yoon JK. High-dose (111)in induces g1 cell cycle arrest and cell death in rat bone marrow mesenchymal stem cells. Nucl Med Mol Imaging 2012; 46:81-8. [PMID: 24900039 DOI: 10.1007/s13139-011-0124-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 11/28/2011] [Accepted: 12/14/2011] [Indexed: 01/17/2023] Open
Abstract
PURPOSE This study was performed to evaluate the effect of (111)In-labeling on the cell growth, cycle and viability of bone marrow mesenchymal stem cells (BMSCs). METHODS Rat BMSCs were labeled with various doses of (111)In (0.4-11.1 Bq/cell). The growth curve of (111)In-BMSCs was obtained up to 14th day of labeling. The cell cycle was evaluated by 5-bromo-2-deoxyuridine (BrdU) labeling or propidium iodide (PI) staining. Senescent cells were counted under a light microscope after staining with 5-bromo-4-chloro-3-indolyl-D-galactopyranoside. Flow cytometry was performed to measure apoptotic and necrotic fractions after staining with annexin V-FITC and PI. RESULTS The growth of BMSCs labeled with higher doses of (111)In (4.4 or 11.1 Bq/cell) was significantly inhibited from the 3rd day of labeling. Flow cytometry revealed less BrdU-positive BMSCs at 11.1 Bq (111)In/cell during all measurement days and G1 arrest at 4.4 and 11.1 Bq (111)In/cell. Significant increases in apoptosis and necrosis were also observed at 4.4 (3.04%/1.35%) and 11.1 Bq (111)In/cell (9.07%/3.18%) on the 14th day (control = 1.60%/0.39%). However, no cellular senescence was visualized up to the 14th day. CONCLUSION A high dose of (111)In-labeling induced cell cycle arrest and death in BMSCs; therefore, it should be used with a careful dosimetry in case of applying it to humans.
Collapse
Affiliation(s)
- Bok-Nam Park
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, 442-749 Republic of Korea
| | - Wooyoung Shim
- Institute for Neuroregeneration and Stem Cell Research, Ajou University School of Medicine, Suwon, Republic of Korea ; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Young Hwan Ahn
- Institute for Neuroregeneration and Stem Cell Research, Ajou University School of Medicine, Suwon, Republic of Korea ; Department of Neurosurgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jae-Ho Lee
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Young-Sil An
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, 442-749 Republic of Korea
| | - Joon-Kee Yoon
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, 442-749 Republic of Korea
| |
Collapse
|
23
|
Non-invasive bioluminescence imaging of myoblast-mediated hypoxia-inducible factor-1 alpha gene transfer. Mol Imaging Biol 2012; 13:1124-32. [PMID: 21267661 DOI: 10.1007/s11307-011-0471-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE We tested a novel imaging strategy, in which both the survival of transplanted myoblasts and their therapeutic transgene expression, a recombinant hypoxia-inducible factor-1α (HIF-1α-VP2), can be monitored using firefly luciferase (fluc) and Renilla luciferase (hrl) bioluminescence reporter genes, respectively. PROCEDURES The plasmid pUbi-hrl-pUbi-HIF-1α-VP2, which expresses both hrl and HIF-1α-VP2 using two ubiquitin promoters, was characterized in vitro. C2c12 myoblasts stably expressing fluc and transiently transfected with pUbi-hrl-pUbi-HIF-1α-VP2 were injected into the mouse hindlimb. Both hrl and fluc expression were monitored using bioluminescence imaging (BLI). RESULTS Strong correlations existed between the expression of hRL and each of HIF-1α-VP2, VEGF, and PlGF (r(2) > 0.83, r(2) > 0.82, and r(2) > 0.97, respectively). In vivo, both transplanted cells and HIF-1α-VP2 transgene expression were successfully imaged using BLI. CONCLUSIONS An objective evaluation of myoblast-mediated gene transfer in living mice can be performed by monitoring both the survival and the transgene expression of transplanted myoblasts using the techniques developed herein.
Collapse
|
24
|
Park BN, Shim W, Lee G, Bang OY, An YS, Yoon JK, Ahn YH. Early distribution of intravenously injected mesenchymal stem cells in rats with acute brain trauma evaluated by 99mTc-HMPAO labeling. Nucl Med Biol 2011; 38:1175-82. [DOI: 10.1016/j.nucmedbio.2011.05.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 05/17/2011] [Indexed: 02/01/2023]
|
25
|
Affiliation(s)
- Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
26
|
Dimayuga VM, Rodriguez-Porcel M. Molecular imaging of cell therapy for gastroenterologic applications. Pancreatology 2011; 11:414-27. [PMID: 21912197 DOI: 10.1159/000327395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stem cell therapy has appeared as a possible therapeutic alternative for numerous diseases. Furthermore, cancer stem cells are a focus of significant interest as they may allow for a better understanding of the genesis of different malignancies. The ultimate goal of stem cell therapeutics is to ensure the viability and functionality of the transplanted cells. Similarly, the ultimate goal of understanding cancer stem cells is to understand how they behave in the living subject. Until recently, the efficacy of stem cell therapies has been assessed by overall organ function recovery. Understanding the behavior and biology of stem cells directly in the living subject can also lead to therapy optimization. Thus, there is a critical need for reliable and accurate methods to understand stem cell biology in vivo. Recent advances in both imaging and molecular biology have enabled transplanted stem cells to be successfully monitored in the living subject. The use of molecular imaging modalities has the capability to answer these questions and may one day be translated to patients. In this review, we will discuss the potential imaging strategies and how they can be utilized, depending on the questions that need to be answered.
Collapse
|
27
|
Perfusion defect size predicts engraftment but not early retention of intra-myocardially injected cardiosphere-derived cells after acute myocardial infarction. Basic Res Cardiol 2011; 106:1379-86. [PMID: 21706191 PMCID: PMC3228962 DOI: 10.1007/s00395-011-0197-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 05/13/2011] [Accepted: 06/13/2011] [Indexed: 11/29/2022]
Abstract
Therapeutic cell retention and engraftment are critical for myocardial regeneration. Underlying mechanisms, including the role of tissue perfusion, are not well understood. In Wistar Kyoto rats, syngeneic cardiosphere-derived cells (CDCs) were injected intramyocardially, after experimental myocardial infarction. CDCs were labeled with [18F]-FDG (n = 7), for quantification of 1-h retention, or with sodium-iodide-symporter gene (NIS; n = 8), for detection of 24-h engraftment by reporter imaging. Perfusion was imaged simultaneously. Infarct size was 37 ± 9 and 38 ± 9% of LV in FDG and NIS groups. Cell signal was located in the infarct border zone in all animals. No significant relationship was observed between infarct size and 1-h CDC retention (r = −0.65; P = 0.11). However, infarct size correlated significantly with 24-h engraftment (r = 0.75; P = 0.03). Residual perfusion at the injection site was not related to cell retention/engraftment. Larger infarcts are associated with improved CDC engraftment. This observation encourages further investigation of microenvironmental conditions after ischemic damage and their role in therapeutic cell survival.
Collapse
|
28
|
Abstract
Regenerative medicine using stem cells has appeared as a potential therapeutic alternative for coronary artery disease, and stem cell clinical studies are currently on their way. However, initial results of these studies have provided mixed information, in part because of the inability to correlate organ functional information with the presence/absence of transplanted stem cells. Recent advances in molecular biology and imaging have allowed the successful noninvasive monitoring of transplanted stem cells in the living subject. In this article, different imaging strategies (direct labeling, indirect labeling with reporter genes) to study the viability and biology of stem cells are discussed. In addition, the limitations of each approach and imaging modality (eg, single photon emission computed tomography, positron emission tomography, and MRI) and their requirements for clinical use are addressed. Use of these strategies will be critical as the different regenerative therapies are being tested for clinical use.
Collapse
|
29
|
Jeon YH, Ahn SJ, Lee YJ, Lee YL, Lee SW, Park SY, Kim IS, Ahn BC, Ha JH, Lee J. Human sodium iodide symporter added to multidrug resistance 1 small hairpin RNA in a single gene construct enhances the therapeutic effects of radioiodine in a nude mouse model of multidrug resistant colon cancer. Cancer Biother Radiopharm 2011; 25:671-9. [PMID: 21204761 DOI: 10.1089/cbr.2010.0837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The objective of this study was to investigate the therapeutic potential of ¹³¹I added to doxorubicin therapy in multidrug resistance (MDR) mouse colon cancer coexpressing the MDR1 small hairpin RNA (shRNA) and human sodium iodide symporter (hNIS) gene in a single gene construct and to visualize the antitumor effects using molecular nuclear imaging. HCT-15 coexpressing shRNA for MDR1 gene (MDR1 shRNA) and hNIS gene with a single construct was established (referred to as MN61 cell). Inhibition of P-gp function by MDR1 shRNA and functional activity of hNIS gene was assessed using a ⁹⁹(m)Tc sestamibi uptake and ¹²⁵I uptake, respectively. Cytotoxic effects by a combination of doxorubicin and ¹³¹I were determined in parental (HCT-15) or MN61 cells using an in vitro clonogenic assay. Therapeutic effect of either combination therapy (doxorubicin and ¹³¹I) or single therapy (doxorubicin or ¹³¹I alone) was evaluated by tumor volume measurement. ⁹⁹(m)Tc-sestamibi, ¹²³I, and ⁹⁹(m)Tc-pertechnetate images of mice were acquired to evaluate functional assessment in vivo. Cellular uptake of ⁹⁹(m)Tc-sestamibi and ¹²⁵I was approximately 2-fold and 100-fold higher in MN61 cells than in parental cells, respectively. Combination of ¹³¹I and doxorubicin resulted in higher cytotoxcity in MN61 cells as compared with parental cells. Scintigraphic imaging showed higher uptake of ⁹⁹(m)Tc-sestamibi and ¹²³I in MN61 tumor as compared with parental tumor. In mice treated with doxorubicin, there was a slight delay in tumor growth in the MN61 tumor but not in the parental tumor. Cancer treatment with ¹³¹I or doxorubicin induced a rapid reduction of tumor volume in the MN61 tumor but not in the parental tumor. Combination therapy further generated a rapid reduction of tumor volume as compared with ¹³¹I therapy alone (p < 0.05). A combination hNIS mediated radioiodine gene therapy added to MDR1 shRNA treatment improved the effects of cancer treatment in a MDR cancer model and could enable visualization of the antitumor effects with nuclear imaging.
Collapse
Affiliation(s)
- Yong Hyun Jeon
- Department of Nuclear Medicine, Kyungpook National University, Daegu, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Comparison of in leakage from labeled endocardial and epicardial cells: impact on modeling viability of cells to be transplanted into myocardium. INTERNATIONAL JOURNAL OF MOLECULAR IMAGING 2011; 2011:472375. [PMID: 21603238 PMCID: PMC3094859 DOI: 10.1155/2011/472375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 11/18/2010] [Accepted: 02/09/2011] [Indexed: 02/02/2023]
Abstract
Introduction. Previously we proposed a cellular imaging technique to determine the surviving fraction of transplanted cells in vivo. Epicardial kinetics using Indium-111 determined the Debris Impulse Response Function (DIRF) and leakage coefficient parameters. Convolution-based modeling which corrected for these signal contributions indicated that 111In activity was quantitative of cell viability with half-lives within 20 hrs to 37 days. We determine if the 37-day upper limit remains valid for endocardial injections by comparing previous epicardial cell leakage parameter estimates to those for endocardial cells. Methods. Normal canine myocardium was injected (111In-tropolone) epicardially (9 injections) or endocardially (10 injections). Continuous whole body and SPECT scans for 5 hours were acquired with three weekly follow-up imaging sessions up to 20–26 days. Time-activity curves evaluated each injection type. Results. The epicardial and endocardial kinetics were not significantly different (Epi: 1286 ± 253; Endo: 1567 ± 470 hours P = .62).
Conclusion. The original epicardial estimate of leakage kinetics has been validated for use in endocardial injections.
Collapse
|
31
|
Abstract
INTRODUCTION Cell transplants to replace cells lost due to injury or degenerative diseases, for which there are currently no cures, are being pursued in a wide range of experimental models. Thus, the application of stem cell-based therapies to treat neurodegenerative and traumatic injuries is now a clinical reality. However, the monitoring of cellular grafts, non-invasively, is an important aspect of the ongoing efficacy and safety assessment of cell-based therapies. Hence, there is a need for non-invasive imaging techniques to ensure that transplants are not only administered to the relevant site, but also allow the monitoring of inappropriate cellular migration to improve our understanding of stem cell migration in the context of the whole organism. AREAS COVERED This review provides an up to date overview of molecular imaging approaches that have been used for visualizing and tracking transplanted stem cells, in vivo. EXPERT OPINION It's important to emphasize that the application of molecular imaging to interrogate transplanted cells may require one or even two imaging modalities to provide a reasonable assessment of transplanted cells in specific organs.
Collapse
Affiliation(s)
- Kishore Bhakoo
- Singapore Bioimaging Consortium - A*Star - Translational Molecular Imaging, Singapore 138667, Singapore.
| |
Collapse
|
32
|
Powerski M, Henrich D, Sander A, Teiler A, Marzi I. In vitro manipulation of endothelial progenitor cell adhesion to vascular endothelium and extracellular matrix by the phorbol ester PMA. ARTIFICIAL CELLS, BLOOD SUBSTITUTES, AND IMMOBILIZATION BIOTECHNOLOGY 2011; 39:214-22. [PMID: 21314293 DOI: 10.3109/10731199.2011.555838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Injection of endothelial progenitor cells (EPCs) into arteries for cell therapy is a promising field in regenerative medicine. However, adhesion of EPCs during capillary passage is restricted, and non-adhering cells are lost into circulation. Here we demonstrate that it is possible to achieve a three- to sevenfold higher rate of EPC adhesion to endothelium and extracellular matrix molecules after short-term activation with phorbol myristate acetate (PMA). In addition, differentiation and toxicity analyses of PMA activated EPCs showed no impact on cell differentiation and negligible impact on cell survival.
Collapse
Affiliation(s)
- Maciej Powerski
- Department of Trauma Surgery, Johann Wolfgang Goethe University, Frankfurt/Main, Germany.
| | | | | | | | | |
Collapse
|
33
|
Musialek P, Tekieli L, Kostkiewicz M, Majka M, Szot W, Walter Z, Zebzda A, Pieniazek P, Kadzielski A, Banys RP, Olszowska M, Pasowicz M, Zmudka K, Tracz W. Randomized transcoronary delivery of CD34(+) cells with perfusion versus stop-flow method in patients with recent myocardial infarction: Early cardiac retention of ⁹⁹(m)Tc-labeled cells activity. J Nucl Cardiol 2011; 18:104-16. [PMID: 21161463 PMCID: PMC3032199 DOI: 10.1007/s12350-010-9326-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 09/20/2010] [Indexed: 12/21/2022]
Abstract
BACKGROUND For transcoronary progenitor cells' administration, injections under flow arrest (over-the-wire balloon technique, OTW) are used universally despite lack of evidence for being required for cell delivery or being effective in stimulating myocardial engraftment. Flow-mediated endothelial rolling is mandatory for subsequent cell adhesion and extravasation. METHODS To optimize cell directing toward the coronary endothelium under maintained flow, the authors developed a cell-delivery side-holed perfusion catheter (PC). Thirty-four patients (36-69 years, 30 men) with primary stent-assisted angioplasty-treated anterior MI (peak TnI 151 [53-356]ng/dL, mean[range]) were randomly assigned to OTW or PC autologous ⁹⁹Tc-extametazime-labeled bone marrow CD34(+) cells (4.34 [0.92-7.54] × 10⁶) administration at 6-14 days after pPCI (LVEF 37.1 [24-44]%). Myocardial perfusion (⁹⁹(m)Tc-MIBI) and labeled cells' activity were evaluated (SPECT) at, respectively, 36-48 h prior to and 60 min after delivery. RESULTS In contrast to OTW coronary occlusions, no intolerance or ventricular arrhythmia occurred with PC cells' administration (P < .001). One hour after delivery, 4.86 [1.7-7.6]% and 5.05 [2.2-9.9]% activity was detected in the myocardium (OTW and PC, respectively, P = .84). Labeled cell activity was clearly limited to the (viable) peri-infarct zone in 88% patients, indicating that the infarct core zone may be largely inaccessible to transcoronary-administered cells. CONCLUSIONS Irrespective of the transcoronary delivery method, only ≈ 5% of native (i.e., non-engineered) CD34(+) cells spontaneously home to the injured myocardium, and cell retention occurs preferentially in the viable peri-infarct zone. Although the efficacy of cell delivery is not increased with the perfusion method, by avoiding provoking ischemic episodes PC offers a rational alternative to the OTW delivery.
Collapse
Affiliation(s)
- Piotr Musialek
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | - Lukasz Tekieli
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | - Magdalena Kostkiewicz
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | - Marcin Majka
- Department of Transplantation, Jagiellonian University, Krakow, Poland
| | | | - Zbigniew Walter
- Department of Hematology, Jagiellonian University, Krakow, Poland
| | - Anna Zebzda
- Department of Transplantation, Jagiellonian University, Krakow, Poland
| | - Piotr Pieniazek
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | | | | | - Maria Olszowska
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | | | - Krzysztof Zmudka
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | - Wieslawa Tracz
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| |
Collapse
|
34
|
Seo JH, Jeon YH, Lee YJ, Yoon GS, Won DI, Ha JH, Jeong SY, Lee SW, Ahn BC, Lee J. Trafficking macrophage migration using reporter gene imaging with human sodium iodide symporter in animal models of inflammation. J Nucl Med 2010; 51:1637-43. [PMID: 20847173 DOI: 10.2967/jnumed.110.077891] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
UNLABELLED The aim of this study was to investigate the feasibility of nuclear molecular imaging using the human sodium iodide symporter (hNIS) as a reporter gene to monitor macrophage migration toward the inflammatory foci. METHODS A stable macrophage cell line coexpressing hNIS and green fluorescent protein (GFP) genes (RAW264.7/hNIS-GFP and R(NIS) cell) was established from an immortalized macrophage cell line (RAW264.7 cells). (125)I uptake was determined (for hNIS protein functional activity), and flow cytometry analysis (to examine GFP gene expression), a cell proliferation assay, a cytokine assay, and a phagocytic activity assay were performed. (99m)Tc-pertechnetate images were acquired at 1 d after subcutaneous inoculation of R(NIS) cells in nude mice. Chemical inflammation was induced for in vivo imaging in the thigh of nude mice by turpentine oil injection. Small-animal PET with (18)F-FDG and (124)I was performed with an intravenous administration of RAW264.7 or R(NIS) cells in inflammation-induced animals. RESULTS The expression of hNIS and GFP genes was confirmed in R(NIS) cells by flow cytometry and immunofluorescent staining. (125)I uptake was about 67 times higher in R(NIS) cells than in RAW264.7 cells. No significant difference was observed in cell proliferation, cytokine production, and phagocytic activity between RAW264.7 and R(NIS) cells. (99m)Tc-pertechnetate imaging revealed increased tracer uptake at the inoculation site. PET with (124)I demonstrated a donut-shaped uptake, correlating with uptake shown by the (18)F-FDG PET images, at the inflammation site of mice administered R(NIS) cells. (124)I uptake (percentage injected dose per gram) was about 2.12 times higher at the inflammation site in the R(NIS) mice than in RAW264.7 mice. By immunohistochemistry, the migration of macrophages was further confirmed by positive staining for GFP and hNIS at the inflammation site of R(NIS) mice. CONCLUSION These data support the feasibility of hNIS reporter gene imaging to monitor the macrophage migration toward an inflammatory lesion. Macrophages expressing hNIS may provide a new strategy to investigate the cellular behavior seen with inflammatory response in a preclinical model.
Collapse
Affiliation(s)
- Ji Hyoung Seo
- Department of Nuclear Medicine, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Donor and recipient contribution to transplant vasculopathy in chronic renal transplant dysfunction. Transplantation 2010; 88:1386-92. [PMID: 20029335 DOI: 10.1097/tp.0b013e3181bca1e4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Chronic transplant dysfunction is the leading cause of long-term renal allograft loss. One of the histologic hallmarks of chronic transplant dysfunction is transplant vasculopathy characterized by accumulation of smooth muscle cells (SMCs) in the arterial subendothelial space, leading to ischemic graft failure. Currently, no therapy is available for transplant vasculopathy, and knowledge of the origin (donor vs. recipient) of neointimal cells may contribute to develop adequate strategies. METHODS Origin of neointimal SMCs, endothelial, and tubular cells was determined in four nephrectomy samples from male recipients transplanted with a female kidney. Recipient-derived cells were detected using X- and Y-chromosome-specific fluorescent in situ hybridization combined with immunofluorescent staining. Specificity and sensitivity of fluorescent in situ hybridization were determined with corresponding controls. RESULTS No Y-chromosome-positive cells were detected in the female to female graft, whereas approximately 31% of nucleated cells in male to male grafts had a detectable Y-chromosome. In female to male grafts, a recipient-derived population of neointimal alpha-smooth muscle actin-positive SMCs were detected (6%, range 3%-11%). Percentages of recipient-derived arterial endothelial cells, glomerular endothelial cells, and tubular epithelial cells were 14% (range 4%-32%), 19% (range 7%-31%) and 3% (range 2%-5%), respectively. CONCLUSIONS Both donor- and recipient-derived cells contribute to vascular remodeling in clinical renal transplantation. The presence of alpha-smooth muscle actin in donor- and recipient-derived cells supports a constructive role for these cells in neointimal formation. However, the predominance of donor-derived cells in the neointima points to these cells as the likely therapeutic target.
Collapse
|
36
|
Cao AH, Shi HJ, Zhang Y, Teng GJ. In vivo tracking of dual-labeled mesenchymal stem cells homing into the injured common carotid artery. Anat Rec (Hoboken) 2009; 292:1677-83. [PMID: 19685506 DOI: 10.1002/ar.20951] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The aim of this study is to conduct in vivo, noninvasive magnetic resonance imaging of labeled rat bone mesenchymal stem cells (BMSCs) as they home into the site of injured common carotid artery following allograft transplantation. Our study was approved by the Institutional Committee on Animal Research. Purified rat BMSCs were dual labeled with superparamagnetic iron oxide (SPIO) particle and fluorescent DiI dye, and subsequently transplanted into recipient rats injured in the left common carotid arteries. Immediately before and 3 hr, 3, 7 and 12 days after transplantation, the labeled cells were monitored in vivo using a 7T micromagnetic resonance imaging (7T micro-MRI) scanner. The signal-to-noise ratios (SNRs) at the injured sites were corroborated with histological examination using Prussian blue staining and fluorescent imaging. Rat BMSCs were labeled with SPIO and DiI at 100% efficiency. When compared with the baseline level before transplantation, the SNR decreased significantly on Days 3 and 7 after injection in the experimental group (Dunnet t test, P < 0.05), whereas insignificant differences were observed after 3 hr and 12 days (Dunnet t test, P > 0.05). In the control group, no significant differences in SNR were found among different time points (ANOVA, P > 0.05). Histological analyses illustrated that red fluorescence and Prussian blue-positive cells were mainly distributed around the lesion areas of injured common carotid arteries. Rat BMSCs can be efficiently labeled with SPIO and DiI, and the directional homing of labeled cells to the site of injured common carotid arteries after intravascular transplantation could be tracked in vivo with 7T micro-MRI.
Collapse
Affiliation(s)
- Ai Hong Cao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | | | | | | |
Collapse
|
37
|
Terrovitis J, Lautamäki R, Bonios M, Fox J, Engles JM, Yu J, Leppo MK, Pomper MG, Wahl RL, Seidel J, Tsui BM, Bengel FM, Abraham MR, Marbán E. Noninvasive quantification and optimization of acute cell retention by in vivo positron emission tomography after intramyocardial cardiac-derived stem cell delivery. J Am Coll Cardiol 2009; 54:1619-26. [PMID: 19833262 DOI: 10.1016/j.jacc.2009.04.097] [Citation(s) in RCA: 198] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Revised: 03/16/2009] [Accepted: 04/13/2009] [Indexed: 02/08/2023]
Abstract
OBJECTIVES The aim of this study was to quantify acute myocardial retention of cardiac-derived stem cells (CDCs) and evaluate different delivery methods with positron emission tomography (PET). BACKGROUND Success of stem cell transplantation for cardiac regeneration is partially limited by low retention/engraftment of the delivered cells. A clinically applicable method for accurate quantification of cell retention would enable optimization of cell delivery. METHODS The CDCs were derived from syngeneic, male Wistar Kyoto (WK) rats labeled with [(18)F]-fluoro-deoxy-glucose ((18)FDG) and injected intramyocardially into the ischemic region of female WK rats after permanent left coronary artery ligation. The effects of fibrin glue (FG), bradycardia (adenosine), and cardiac arrest were examined. Imaging with (18)FDG PET was performed for quantification of cell retention. Quantitative polymerase chain reaction (PCR) for the male-specific SRY gene was performed to validate the PET results. RESULTS Myocardial retention of cells suspended in phosphate-buffered saline 1 h after delivery was 17.6 +/- 11.5% by PCR and 17.8 +/- 7.3% by PET. When CDCs were injected immediately after induction of cardiac arrest, retention was increased to 75.6 +/- 18.6%. Adenosine slowed the ventricular rate and doubled CDC retention (35.4 +/- 5.3%). A similar increase in CDC retention was observed after epicardial application of FG at the injection site (37.5 +/- 8.2%). The PCR revealed a significant increase in 3-week cell engraftment in the FG animals (22.1 +/- 18.6% and 5.3 +/- 3.1%, for FG and phosphate-buffered saline, respectively). CONCLUSIONS In vivo PET permits accurate measurement of CDC retention early after intramyocardial delivery. Sealing injection sites with FG or lowering ventricular rate by adenosine might be clinically translatable methods for improving stem cell engraftment in a beating heart.
Collapse
Affiliation(s)
- John Terrovitis
- The Heart Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Higuchi T, Anton M, Saraste A, Dumler K, Pelisek J, Nekolla SG, Bengel FM, Schwaiger M. Reporter gene PET for monitoring survival of transplanted endothelial progenitor cells in the rat heart after pretreatment with VEGF and atorvastatin. J Nucl Med 2009; 50:1881-6. [PMID: 19837770 DOI: 10.2967/jnumed.109.067801] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED It has been suggested that vascular endothelial growth factor (VEGF) and statins enhance the survival, proliferation, and function of endothelial progenitor cells (EPCs). We investigated whether reporter gene PET can be used to detect the effects of atorvastatin and VEGF on survival of EPCs after transplantation in the rat heart. METHODS Healthy nude rats received an intramyocardial injection of 4 million human EPCs retrovirally transduced with the sodium/iodide symporter gene for reporter gene imaging. Reporter gene expression was imaged at days 1 and 3 after injection on a small-animal PET scanner with (124)I, and the presence of EPCs was confirmed by immunohistochemistry with human CD31 antibodies. The control group received EPCs transduced only with the reporter gene, whereas treatment groups received oral atorvastatin (10 mg/kg/d) and EPCs cotransduced with adenoviral vectors encoding VEGF in addition to sodium/iodide symporter. RESULTS Immunohistochemistry showed more EPCs at the site of injection after atorvastatin treatment and in the presence of VEGF expression in EPCs than in controls. PET successfully visualized EPCs as focal (124)I accumulation at the site of injection. The quantitative amount of (124)I accumulation assessed by PET was significantly higher in the pretreatment than control group. Autoradiography confirmed (124)I accumulation in the myocardium that correlated with the number of EPCs. CONCLUSION Early survival of transplanted EPCs in the rat myocardium is prolonged by pretreatment with a combination of atorvastatin and VEGF. Reporter gene PET, by successfully quantifying the effect, is an attractive tool for monitoring stem cell survival in vivo.
Collapse
Affiliation(s)
- Takahiro Higuchi
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Bleiziffer O, Horch RE, Hammon M, Arkudas A, Naschberger E, Rath S, Pryymachuk G, Beier JP, Hatzopoulos AK, Stürzl M, Kneser U. T17b murine embryonal endothelial progenitor cells can be induced towards both proliferation and differentiation in a fibrin matrix. J Cell Mol Med 2009; 13:926-35. [PMID: 19538255 PMCID: PMC2827195 DOI: 10.1111/j.1582-4934.2008.00527.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Endothelial progenitor cells (EPC) may enhance blood vessel formation in a variety of clinical settings such as ischaemia and tumour angiogenesis as well as in tissue-engineered matrices. In the present study, we cultured a murine endothelial progenitor cell line, T17b, in vitro in cell culture as well as in an FDA-approved fibrin matrix and investigated cell proliferation, differentiation and secretion patterns of the angiogenic growth factor VEGF under hypoxia and differentiation. We show that T17b EPC remain viable for at least 8 days in the fibrin matrix where they proliferate and form clusters including lumen-like structures. Proliferation in fibrin clots overlayed with basal medium (BM) was confirmed morphologically and immunohistochemically by positive Ki67 staining, indicating mitotic activity. Significant cell proliferation and Ki-67 expression were absent when cells were incubated with dibutyryl-cAMP and retinoic acid (RA). Incubation with dibutyryl-cAMP and RA stimulated the expression of the EPC differentiation markers von Willebrand Factor (vWF) and VEGF receptor 2 (VEGFR-2), indicating successful differentiation in the fibrin clot. EPC differentiation induced by dibutyryl-cAMP and RA was confirmed in 2-D chamber slide cultures by positive vWF immunostaining, which was absent in BM controls. EPC chamber slides also displayed positive vWF staining when exposed to hypoxia under BM conditions, indicating EPC activation and differentiation could also be induced by hypoxia. Taken together, T17b EPC secrete increased levels of VEGF when submitted to either hypoxia or differentiation and can be differentiated into mature endothelial cells not only in cell and matrigel cultures but also in a fibrin matrix that is FDA approved for clinical application.
Collapse
Affiliation(s)
- Oliver Bleiziffer
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University of Erlangen Medical Center, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ly HQ, Hoshino K, Pomerantseva I, Kawase Y, Yoneyama R, Takewa Y, Fortier A, Gibbs-Strauss SL, Vooght C, Frangioni JV, Hajjar RJ. In vivo myocardial distribution of multipotent progenitor cells following intracoronary delivery in a swine model of myocardial infarction. Eur Heart J 2009; 30:2861-8. [PMID: 19687154 DOI: 10.1093/eurheartj/ehp322] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
AIMS There are few data comparing the fate of multipotent progenitor cells (MPCs) used in cardiac cell therapy after myocardial infarction (MI). To document in vivo distribution of MPCs delivered by intracoronary (IC) injection. METHODS AND RESULTS Using an anterior MI swine model, near-infrared (NIR) fluorescence was used for in vivo tracking of labelled MPCs [mesenchymal stromal (MSCs), bone marrow mononuclear (BMMNCs), and peripheral blood mononuclear (PBMNCs)] cells early after IC injection. Signal intensity ratios (SIRs) of injected over non-injected (reference) zones were used to report NIR fluorescence emission. Following IC injection, significant differences in mean SIR were documented when MSCs were compared with BMMNCs [1.28 +/- 0.10 vs. 0.77 +/- 0.11, P < 0.001; 95% CI (0.219, 0.805), respectively] or PBMNCs [1.28 +/- 0.10 vs. 0.80 +/- 0.14, P = 0.005; 95% CI (0.148, 0.813), respectively]. Differences were maintained during the 60 min tracking period, with only the MSC-injected groups continuously emitting NIR fluorescence (SIR>1). This is correlated with greater cell retention for MSCs relative to mononuclear cells. However, there was evidence of MSC-related vessel plugging in some swine. CONCLUSION Our in vivo NIR fluorescence findings suggest that MPC distribution and retention immediately after intracoronary delivery vary depending on cell population and could potentially impact the clinical efficacy of cardiac cell therapy.
Collapse
Affiliation(s)
- Hung Q Ly
- Mount Sinai School of Medicine, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Técnicas no invasivas de imagen cardiovascular en investigación básica: aplicación en la terapia celular. Rev Esp Cardiol 2009. [DOI: 10.1016/s0300-8932(09)72074-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
42
|
Higuchi T, Anton M, Dumler K, Seidl S, Pelisek J, Saraste A, Welling A, Hofmann F, Oostendorp RAJ, Gansbacher B, Nekolla SG, Bengel FM, Botnar RM, Schwaiger M. Combined reporter gene PET and iron oxide MRI for monitoring survival and localization of transplanted cells in the rat heart. J Nucl Med 2009; 50:1088-94. [PMID: 19525455 DOI: 10.2967/jnumed.108.060665] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED There is a need for in vivo monitoring of cell engraftment and survival after cardiac cell transplantation therapy. This study assessed the feasibility and usefulness of combined PET and MRI for monitoring cell engraftment and survival after cell transplantation. METHODS Human endothelial progenitor cells (HEPCs), derived from CD34+ mononuclear cells of umbilical cord blood, were retrovirally transduced with the sodium iodide symporter (NIS) gene for reporter gene imaging by (124)I-PET and labeled with iron oxides for visualization by MRI. Imaging and histologic analysis were performed on 3 groups of nude rats on days 1, 3, and 7 after intramyocardial injection of 4 million HEPCs. RESULTS In vitro studies demonstrated stable expression of functional NIS protein and normal viability of HEPCs after transduction. On day 1, after intramyocardial transplantation, iron- and NIS-labeled HEPCs were visualized successfully on MRI as a regional signal void in the healthy myocardium and on PET as (124)I accumulation. The (124)I uptake decreased on day 3 and was undetectable on day 7, and the MRI signal remained unchanged throughout the follow-up period. Histologic analysis with CD31 and CD68 antibodies confirmed the presence of either labeled or nonlabeled control transplanted HEPCs at the site of injection on day 1 but not on day 7, when only iron-loaded macrophages were seen. Furthermore, deoxyuride-5'-triphosphate biotin nick end labeling showed extensive apoptosis at the site of transplantation. CONCLUSION The combination of MRI and PET allows imaging of localization and survival of transplanted HEPCs together with morphologic information about the heart. Although iron labeling rapidly loses specificity for cell viability because of phagocytosis of iron particles released from dead cells, reporter gene expression provided specific information on the number of surviving cells. This multimodality approach allows complementary analysis of cell localization and viability.
Collapse
Affiliation(s)
- Takahiro Higuchi
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Advances in noninvasive imaging techniques may aid in the understanding of cardiac stem cell therapy. Nuclear imaging enables in vivo evaluation of myocardial perfusion, metabolism, and function, in addition to the stem cell fate. This article summarizes recent clinical and experimental nuclear imaging studies in cardiac stem cell therapy.
Collapse
|
44
|
Stastna M, Abraham MR, Van Eyk JE. Cardiac stem/progenitor cells, secreted proteins, and proteomics. FEBS Lett 2009; 583:1800-7. [PMID: 19303873 PMCID: PMC4340703 DOI: 10.1016/j.febslet.2009.03.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 03/02/2009] [Accepted: 03/13/2009] [Indexed: 01/06/2023]
Abstract
Stem cell-based therapy is emerging as a novel approach for myocardial repair over conventional cardiovascular therapies. In addition to embryonic stem cells and adult stem cells from noncardiac sources, there is a small population of resident stem cells in the heart from which new cardiac cells (myocytes, vascular endothelial cells and smooth muscle cells) can be derived and used for cardiac repair in case of heart injury. It has been proposed that the clinical benefit of stem cells may arise from secreted proteins that mediate regeneration in a paracrine/autocrine manner. To be able to track the regulatory pathway on a molecular basis, utilization of proteomics in stem cell research is essential. Proteomics offers a tool that can address questions regarding stem cell response to disease/injury.
Collapse
Affiliation(s)
- Miroslava Stastna
- Institute of Analytical Chemistry of the ASCR, v.v.i., Veveri 97, 602 00 Brno, Czech Republic.
| | | | | |
Collapse
|
45
|
Molecular Imaging in Cardiology. Mol Imaging 2009. [DOI: 10.1007/978-3-540-76735-0_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
46
|
Terrovitis J, Kwok KF, Lautamäki R, Engles JM, Barth AS, Kizana E, Miake J, Leppo MK, Fox J, Seidel J, Pomper M, Wahl RL, Tsui B, Bengel F, Marbán E, Abraham MR. Ectopic expression of the sodium-iodide symporter enables imaging of transplanted cardiac stem cells in vivo by single-photon emission computed tomography or positron emission tomography. J Am Coll Cardiol 2008; 52:1652-60. [PMID: 18992656 DOI: 10.1016/j.jacc.2008.06.051] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 05/07/2008] [Accepted: 06/19/2008] [Indexed: 11/19/2022]
Abstract
OBJECTIVES We examined the sodium-iodide symporter (NIS), which promotes in vivo cellular uptake of technetium 99m ((99m)Tc) or iodine 124 ((124)I), as a reporter gene for cell tracking by single-photon emission computed tomography (SPECT) or positron emission tomography (PET) imaging. BACKGROUND Stem cells offer the promise of cardiac repair. Stem cell labeling is a prerequisite to tracking cell fate in vivo. METHODS The human NIS complementary deoxyribonucleic acid was transduced into rat cardiac-derived stem cells (rCDCs) using lentiviral vectors. Rats were injected intramyocardially with up to 4 million NIS(+)-rCDCs immediately after left anterior descending coronary artery ligation. Dual isotope SPECT (or PET) imaging was performed, using (99m)Tc (or (124)I) for cell detection and thallium 201 (or ammonia 13) for myocardial delineation. In a subset of animals, high resolution ex vivo SPECT scans of explanted hearts were obtained to confirm that in vivo signals were derived from the cell injection site. RESULTS NIS expression in rCDCs did not affect cell viability and proliferation. NIS activity was verified in isolated transduced cells by measuring (99m)Tc uptake. NIS(+) rCDCs were visualized in vivo as regions of (99m)Tc or (124)I uptake within a perfusion deficit in the SPECT and PET images, respectively. Cells could be visualized by SPECT up to 6 days post-injection. Ex vivo SPECT confirmed that in vivo (99m)Tc signals were localized to the cell injection sites. CONCLUSIONS Ectopic NIS expression allows noninvasive in vivo stem cell tracking in the myocardium, using either SPECT or PET. The general approach shows significant promise in tracking the fate of transplanted cells participating in cardiac regeneration, given its ability to observe living cells using clinically applicable imaging modalities.
Collapse
Affiliation(s)
- John Terrovitis
- Department of Cardiology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chen IY, Greve JM, Gheysens O, Willmann JK, Rodriguez-Porcel M, Chu P, Sheikh AY, Faranesh AZ, Paulmurugan R, Yang PC, Wu JC, Gambhir SS. Comparison of optical bioluminescence reporter gene and superparamagnetic iron oxide MR contrast agent as cell markers for noninvasive imaging of cardiac cell transplantation. Mol Imaging Biol 2008; 11:178-87. [PMID: 19034584 DOI: 10.1007/s11307-008-0182-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 05/31/2008] [Accepted: 07/22/2008] [Indexed: 10/21/2022]
Abstract
PURPOSE In this study, we compared firefly luciferase (Fluc) reporter gene and superparamagnetic iron oxide (Feridex) as cell markers for longitudinal monitoring of cardiomyoblast graft survival using optical bioluminescence imaging (BLI) and magnetic resonance imaging (MRI), respectively. PROCEDURES Rats (n = 31) underwent an intramyocardial injection of cardiomyoblasts (2 x 10(6)) labeled with Fluc, Feridex, or no marker (control) or an injection of Feridex alone (75 microg). Afterward, rats were serially imaged with BLI or MRI and killed at different time points for histological analysis. RESULTS BLI revealed a drastically different cell survival kinetics (half-life = 2.65 days over 6 days) than that revealed by MRI (half-life = 16.8 days over 80 days). Injection of Feridex alone led to prolonged tissue retention of Feridex (> or =16 days) and persistent MR signal (> or =42 days). CONCLUSIONS Fluc BLI reporter gene imaging is a more accurate gauge of transplanted cell survival as compared to MRI of Feridex-labeled cells.
Collapse
Affiliation(s)
- Ian Y Chen
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mani V, Adler E, Briley-Saebo KC, Bystrup A, Fuster V, Keller G, Fayad ZA. Serial in vivo positive contrast MRI of iron oxide-labeled embryonic stem cell-derived cardiac precursor cells in a mouse model of myocardial infarction. Magn Reson Med 2008; 60:73-81. [PMID: 18581415 DOI: 10.1002/mrm.21642] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Myocardial regeneration with stem-cell transplantation is a possible treatment option to reverse deleterious effects that occur after myocardial infarction. Since little is known about stem cell survival after transplantation, developing techniques for "tracking" cells would be desirable. Iron-oxide-labeled stem cells have been used for in vivo tracking using MRI but produce negative contrast images that are difficult to interpret. The aim of the current study was to test a positive contrast MR technique using reduced z-gradient rephasing (GRASP) to aid in dynamically tracking stem cells in an in vivo model of mouse myocardial infraction. Ferumoxides and protamine sulfate were complexed and used to magnetically label embryonic stem cell-derived cardiac-precursor-cells (ES-CPCs). A total of 500,000 ES-CPCs were injected in the border zone of infarcted mice and MR imaging was performed on a 9.4T scanner using T(2)*-GRE sequences (negative contrast) and positive contrast GRASP technique before, 24 hours, and 1 week after ES-CPC implantation. Following imaging, mice were sacrificed for histology and Perl's staining was used to confirm iron within myocardium. Good correlation was observed between signal loss seen on conventional T(2)* images, bright areas on GRASP, and the presence of iron on histology. This demonstrated the feasibility of in vivo stem cell imaging with positive contrast MRI.
Collapse
Affiliation(s)
- Venkatesh Mani
- Imaging Science Laboratories, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Bahk JY, Han H, Lee YS. Stem cell treatment for complicated diabetes. Int J Stem Cells 2008; 1:91-5. [PMID: 24855513 DOI: 10.15283/ijsc.2008.1.1.91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2008] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Self renewal, homing or migration and multipotent differentiation are characteristics of stem cell. We studied the effect of stem cell treatments on diabetes complicated with impotencies using human umbilical cord blood stem cells (hUCBSCs). METHODS AND RESULTS The patients who had no erection more than 6 months, were not responded to any medication more than 6 months and were waiting penile prosthesis due to type 2 diabetics were participated and number was 5. All had normal laboratory findings except diabetes mellitus related one. Prepared hUCBSCs were ABO, HLA-AB, DR and sex identical to each patient. Total 1.5×10(7) hUCBSCs were infused into both corpus cavernosa. Immune suppression was not done. The blood glucose, medication dose and erection diary were recorded and followed for 9 months. Mean age of participants were 61 (57∼66). The blood glucose dropped from second week, and insulin or hypoglycemic agent doses were reduced in all patients for 6∼7 months. The level of glycosylated hemoglobin was improved from post-treatment for 3∼4 months. The libido was improved and morning erection was regained from 3 weeks. During the follow-up, one patient turned out for prosthesis, two patients were returned to non-erection state at 8 and 9 months and two patients maintained erections with medication. CONCLUSIONS The hUCBSCs has positive effect on blood glucose and erectile dysfunction, although it is not sufficient. We suppose that the stem cell effects might be caused by combination of unknown humoral factors from hUCBSCs and hUCBSCs themselves.
Collapse
Affiliation(s)
- Jong Yoon Bahk
- Department of Urology, Gyeongsang National University Medical Graduate School, Jinju
| | | | | |
Collapse
|
50
|
Musialek P, Kostkiewicz M, Banys RP, Tekieli L, Majka M, Pasowicz M, Tracz W. Early myocardial engraftment of autologous CD34+ cells administered transcoronary via a physiological cell-delivery system. Eur J Nucl Med Mol Imaging 2008; 35:1929-30. [PMID: 18682941 DOI: 10.1007/s00259-008-0865-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2008] [Accepted: 06/01/2008] [Indexed: 10/21/2022]
Affiliation(s)
- Piotr Musialek
- Department of Cardiac and Vascular Diseases, Jagiellonian University, Kraków, Poland.
| | | | | | | | | | | | | |
Collapse
|