1
|
Kahts M, Mellet J, Durandt C, Moodley K, Summers B, Ebenhan T, Zeevaart JR, Aras O, Pepper MS. A proof-of-concept study to investigate the radiolabelling of human mesenchymal and hematopoietic stem cells with [ 89Zr]Zr-Df-Bz-NCS. EJNMMI Radiopharm Chem 2024; 9:82. [PMID: 39611856 DOI: 10.1186/s41181-024-00311-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The transplantation of hematopoietic stem and progenitor cells (HSPCs) or mesenchymal stromal/stem cells (MSCs) for the treatment of a wide variety of diseases has been studied extensively. A challenge with cell-based therapies is that migration to and retention at the target site is often difficult to monitor and quantify. Zirconium-89 (89Zr) is a positron-emitting radionuclide with a half-life of 3.3 days, which allows long-term cell tracking. Para-isothiocyanatobenzyl-desferrioxamine B (Df-Bz-NCS) is the chelating agent of choice for 89Zr-cell surface labelling. We utilised a shortened labelling method, thereby avoiding a 30-60-min incubation step for [89Zr]Zr-Df-Bz-NCS chelation, to radiolabel HSPCs and MSCs with zirconium-89. RESULTS Three 89Zr-MSC labelling attempts were performed. High labelling efficiencies (81.30 and 87.30%) and relatively good labelling yields (59.59 and 67.00%) were achieved with the use of a relatively larger number of MSCs (4.425 and 3.855 million, respectively). There was no significant decrease in MSC viability after 89Zr-labeling (p = 0.31). This labelling method was also translatable to prepare 89Zr-HSPC; preliminary data from one preparation indicated high 89Zr-HSPC labelling efficiency (88.20%) and labelling yield (71.06%), as well as good HSPC viability after labelling (68.65%). CONCLUSIONS Successful 89Zr-MSC and 89Zr-HSPC labelling was achieved, which underlines the prospects for in vivo cell tracking studies with positron emission tomography. In vitro investigations with larger sample sizes and preclinical studies are recommended.
Collapse
Affiliation(s)
- Maryke Kahts
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University (SMU), Ga-Rankuwa, 0208, South Africa.
| | - Juanita Mellet
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Chrisna Durandt
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Kinosha Moodley
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Beverley Summers
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University (SMU), Ga-Rankuwa, 0208, South Africa
| | - Thomas Ebenhan
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Hartebeespoort, South Africa
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Hartebeespoort, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- DST/NWU, Preclinical Drug Development Platform, North-West University, Potchefstroom, South Africa
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Radiology Department, AMRIC Health, New York, NY, USA
| | - Michael S Pepper
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
2
|
Sočan A. Radiolabeling of red blood cells and platelets and quality controls. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
3
|
Kim EE, Youn H, Kang KW. Imaging in Tumor Immunology. Nucl Med Mol Imaging 2021; 55:225-236. [PMID: 34721715 PMCID: PMC8517056 DOI: 10.1007/s13139-021-00706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/31/2021] [Accepted: 06/15/2021] [Indexed: 10/20/2022] Open
Abstract
Recent advances in immune modulation have made impressive progress in cancer immunotherapy. Because dynamic nature of the immune response often makes it difficult to evaluate therapeutic outcomes, innovative imaging technologies have been developed to enable non-invasive visualization of immune cells and tumors in their microenvironment. This review summarizes the current tumor immunology and describes new innovative imaging methods with great potential to obtain non-invasive real-time insights into the complex functions of the immune system and into the management of cancer immunotherapy.
Collapse
Affiliation(s)
- Euishin Edmund Kim
- Department of Radiological Sciences, UCI Medical Center, Orange County, CA USA
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul, South Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
4
|
Kiraga Ł, Kucharzewska P, Paisey S, Cheda Ł, Domańska A, Rogulski Z, Rygiel TP, Boffi A, Król M. Nuclear imaging for immune cell tracking in vivo – Comparison of various cell labeling methods and their application. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
5
|
Kim IK, Park JH, Kim B, Hwang KC, Song BW. Recent advances in stem cell therapy for neurodegenerative disease: Three dimensional tracing and its emerging use. World J Stem Cells 2021; 13:1215-1230. [PMID: 34630859 PMCID: PMC8474717 DOI: 10.4252/wjsc.v13.i9.1215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/20/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative disease is a brain disorder caused by the loss of structure and function of neurons that lowers the quality of human life. Apart from the limited potential for endogenous regeneration, stem cell-based therapies hold considerable promise for maintaining homeostatic tissue regeneration and enhancing plasticity. Despite many studies, there remains insufficient evidence for stem cell tracing and its correlation with endogenous neural cells in brain tissue with three-dimensional structures. Recent advancements in tissue optical clearing techniques have been developed to overcome the existing shortcomings of cross-sectional tissue analysis in thick and complex tissues. This review focuses on recent progress of stem cell treatments to improve neurodegenerative disease, and introduces tissue optical clearing techniques that can implement a three-dimensional image as a proof of concept. This review provides a more comprehensive understanding of stem cell tracing that will play an important role in evaluating therapeutic efficacy and cellular interrelationship for regeneration in neurodegenerative diseases.
Collapse
Affiliation(s)
- Il-Kwon Kim
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon Metropolitan City 22711, South Korea
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangwon-do 25601, South Korea
| | - Jun-Hee Park
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon Metropolitan City 22711, South Korea
| | - Bomi Kim
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon Metropolitan City 22711, South Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon Metropolitan City 22711, South Korea
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangwon-do 25601, South Korea
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon Metropolitan City 22711, South Korea
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangwon-do 25601, South Korea.
| |
Collapse
|
6
|
D’Hollander A, Van Roosbroeck R, Trekker J, Stakenborg T, Dresselaers T, Vande Velde G, Struys T, Lambrichts I, Lammertyn J, Lagae L, Himmelreich U. Synthetic Antiferromagnetic Gold Nanoparticles as Bimodal Contrast Agents in MRI and CT-An Experimental In Vitro and In Vivo Study. Pharmaceutics 2021; 13:pharmaceutics13091494. [PMID: 34575570 PMCID: PMC8472775 DOI: 10.3390/pharmaceutics13091494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 01/16/2023] Open
Abstract
The use of multimodal contrast agents can potentially overcome the intrinsic limitations of individual imaging methods. We have validated synthetic antiferromagnetic nanoparticles (SAF-NPs) as bimodal contrast agents for in vitro cell labeling and in vivo cell tracking using magnetic resonance imaging (MRI) and computed tomography (CT). SAF-NP-labeled cells showed high contrast in MRI phantom studies (r2* = 712 s−1 mM−1), while pelleted cells showed clear contrast enhancement in CT. After intravenous SAF-NP injection, nanoparticles accumulated in the liver and spleen, as visualized in vivo by significant MRI contrast enhancement. Intravenous injection of SAF-NP-labeled cells resulted in cell accumulation in the lungs, which was clearly detectable by using CT but not by using MRI. SAF-NPs proved to be very efficient cell labeling agents for complementary MRI- and CT-based cell tracking. Bimodal monitoring of SAF-NP labeled cells is in particular of interest for applications where the applied imaging methods are not able to visualize the particles and/or cells in all organs.
Collapse
Affiliation(s)
- Antoine D’Hollander
- Biomedical MRI Unit, Department of Imaging and Pathology, KU Leuven, O&N 1, Herestraat 49, 3000 Leuven, Belgium; (A.D.); (J.T.); (T.D.); (G.V.V.)
- Department of Life Science Technology, IMEC, Kapeldreef 75, 3001 Leuven, Belgium; (R.V.R.); (T.S.); (L.L.)
| | - Ruben Van Roosbroeck
- Department of Life Science Technology, IMEC, Kapeldreef 75, 3001 Leuven, Belgium; (R.V.R.); (T.S.); (L.L.)
- Division of Mechatronics, Department of Biosystems, Biostatistics and Sensors, KU Leuven, 3001 Leuven, Belgium;
| | - Jesse Trekker
- Biomedical MRI Unit, Department of Imaging and Pathology, KU Leuven, O&N 1, Herestraat 49, 3000 Leuven, Belgium; (A.D.); (J.T.); (T.D.); (G.V.V.)
- Department of Life Science Technology, IMEC, Kapeldreef 75, 3001 Leuven, Belgium; (R.V.R.); (T.S.); (L.L.)
| | - Tim Stakenborg
- Department of Life Science Technology, IMEC, Kapeldreef 75, 3001 Leuven, Belgium; (R.V.R.); (T.S.); (L.L.)
| | - Tom Dresselaers
- Biomedical MRI Unit, Department of Imaging and Pathology, KU Leuven, O&N 1, Herestraat 49, 3000 Leuven, Belgium; (A.D.); (J.T.); (T.D.); (G.V.V.)
| | - Greetje Vande Velde
- Biomedical MRI Unit, Department of Imaging and Pathology, KU Leuven, O&N 1, Herestraat 49, 3000 Leuven, Belgium; (A.D.); (J.T.); (T.D.); (G.V.V.)
| | - Tom Struys
- Lab of Histology, Biomedical Research Institute, Hasselt University, Agora Laan Gebouw C, 3590 Diepenbeek, Belgium; (T.S.); (I.L.)
| | - Ivo Lambrichts
- Lab of Histology, Biomedical Research Institute, Hasselt University, Agora Laan Gebouw C, 3590 Diepenbeek, Belgium; (T.S.); (I.L.)
| | - Jeroen Lammertyn
- Division of Mechatronics, Department of Biosystems, Biostatistics and Sensors, KU Leuven, 3001 Leuven, Belgium;
| | - Liesbet Lagae
- Department of Life Science Technology, IMEC, Kapeldreef 75, 3001 Leuven, Belgium; (R.V.R.); (T.S.); (L.L.)
- Department of Physics, Faculty of Sciences, Laboratory of Soft Matter and Biophysics, KU Leuven, Celestijnenlaan 200D, 3001 Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI Unit, Department of Imaging and Pathology, KU Leuven, O&N 1, Herestraat 49, 3000 Leuven, Belgium; (A.D.); (J.T.); (T.D.); (G.V.V.)
- Department of Life Science Technology, IMEC, Kapeldreef 75, 3001 Leuven, Belgium; (R.V.R.); (T.S.); (L.L.)
- Correspondence: ; Tel.: +32-16-330-925
| |
Collapse
|
7
|
Van Nguyen P, Fan W, Zhu T, Qian W, Li Y, Liu B, Zhang W, Henry J, Yuan S, Wang X, Paulus YM. Long-Term, Noninvasive In Vivo Tracking of Progenitor Cells Using Multimodality Photoacoustic, Optical Coherence Tomography, and Fluorescence Imaging. ACS NANO 2021; 15:13289-13306. [PMID: 34378374 PMCID: PMC8984873 DOI: 10.1021/acsnano.1c03035] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Stem cell regenerative medicine therapies have emerged as promising treatments for currently incurable diseases. A remaining challenge for cell therapies is the ability to track the migration and distribution of the transplanted cells in a long-term, noninvasive manner in vivo to assess their efficacy. This study develops a noninvasive, and high spatial resolution photoacoustic microscopy (PAM) and optical coherence tomography (OCT) imaging system for in vivo tracking of subretinally injected progenitor human retinal pigment epithelium cells (ARPE-19) labeled with chainlike gold nanoparticle (CGNP) clusters in RPE damage. CGNP provided significant PAM, OCT, and fluorescence signals to selectively track the migration of ARPE-19 cells in living rabbit eyes for 3 months. PAM and OCT imaging allow accurate anatomical information to determine the exact retinal layer in which the transplanted ARPE-19 cells are located which was confirmed by histology. This presents an efficient and advanced technology to visualize fundamental biological processes of cell therapies in complex in vivo environments in real time.
Collapse
Affiliation(s)
- Phuc Van Nguyen
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Wen Fan
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, 210029, China
| | - Tianye Zhu
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, 210029, China
| | - Wei Qian
- IMRA America Inc., Ann Arbor, MI 48105, USA
| | - Yanxiu Li
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Bing Liu
- IMRA America Inc., Ann Arbor, MI 48105, USA
| | - Wei Zhang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jessica Henry
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, 210029, China
| | - Xueding Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Yannis M. Paulus
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
8
|
Zhang S, Lachance BB, Moiz B, Jia X. Optimizing Stem Cell Therapy after Ischemic Brain Injury. J Stroke 2020; 22:286-305. [PMID: 33053945 PMCID: PMC7568970 DOI: 10.5853/jos.2019.03048] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Stem cells have been used for regenerative and therapeutic purposes in a variety of diseases. In ischemic brain injury, preclinical studies have been promising, but have failed to translate results to clinical trials. We aimed to explore the application of stem cells after ischemic brain injury by focusing on topics such as delivery routes, regeneration efficacy, adverse effects, and in vivo potential optimization. PUBMED and Web of Science were searched for the latest studies examining stem cell therapy applications in ischemic brain injury, particularly after stroke or cardiac arrest, with a focus on studies addressing delivery optimization, stem cell type comparison, or translational aspects. Other studies providing further understanding or potential contributions to ischemic brain injury treatment were also included. Multiple stem cell types have been investigated in ischemic brain injury treatment, with a strong literature base in the treatment of stroke. Studies have suggested that stem cell administration after ischemic brain injury exerts paracrine effects via growth factor release, blood-brain barrier integrity protection, and allows for exosome release for ischemic injury mitigation. To date, limited studies have investigated these therapeutic mechanisms in the setting of cardiac arrest or therapeutic hypothermia. Several delivery modalities are available, each with limitations regarding invasiveness and safety outcomes. Intranasal delivery presents a potentially improved mechanism, and hypoxic conditioning offers a potential stem cell therapy optimization strategy for ischemic brain injury. The use of stem cells to treat ischemic brain injury in clinical trials is in its early phase; however, increasing preclinical evidence suggests that stem cells can contribute to the down-regulation of inflammatory phenotypes and regeneration following injury. The safety and the tolerability profile of stem cells have been confirmed, and their potent therapeutic effects make them powerful therapeutic agents for ischemic brain injury patients.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brittany Bolduc Lachance
- Program in Trauma, Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bilal Moiz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Galli F, Aguilera JV, Palermo B, Markovic SN, Nisticò P, Signore A. Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. J Exp Clin Cancer Res 2020; 39:89. [PMID: 32423420 PMCID: PMC7236372 DOI: 10.1186/s13046-020-01586-y] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-infiltrating immune cells play a key role against cancer. However, malignant cells are able to evade the immune response and establish a very complex balance in which different immune subtypes may drive tumor progression, metastatization and resistance to therapy. New immunotherapeutic approaches aim at restoring the natural balance and increase immune response against cancer by different mechanisms. The complexity of these interactions and the heterogeneity of immune cell subpopulations are a real challenge when trying to develop new immunotherapeutics and evaluate or predict their efficacy in vivo. To this purpose, molecular imaging can offer non-invasive diagnostic tools like radiopharmaceuticals, contrast agents or fluorescent dyes. These agents can be useful for preclinical and clinical purposes and can overcome [18F]FDG limitations in discriminating between true-progression and pseudo-progression. This review provides a comprehensive overview of immune cells involved in microenvironment, available immunotherapies and imaging agents to highlight the importance of new therapeutic biomarkers and their in vivo evaluation to improve the management of cancer patients.
Collapse
Affiliation(s)
- Filippo Galli
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, Roma, Italy.
| | - Jesus Vera Aguilera
- Department of oncology and Department of Immunology, Mayo Clinic, (MN), Rochester, USA
| | - Belinda Palermo
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Svetomir N Markovic
- Department of oncology and Department of Immunology, Mayo Clinic, (MN), Rochester, USA
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, Roma, Italy
| |
Collapse
|
10
|
Iking J, Klose J, Staniszewska M, Fendler WP, Herrmann K, Rischpler C. Imaging inflammation after myocardial infarction: implications for prognosis and therapeutic guidance. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:35-50. [PMID: 32077669 DOI: 10.23736/s1824-4785.20.03232-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammation after myocardial infarction (MI) has been in the focus of cardiovascular research for several years as it influences the remodeling process of the ischemic heart and thereby critically determines the clinical outcome of the patient. Today, it is well appreciated that inflammation is a crucial necessity for the initiation of the natural wound healing process; however, excessive inflammation can have detrimental effects and might result in adverse ventricular remodeling which is associated with an increased risk of heart failure. Newly emerged imaging techniques facilitate the non-invasive assessment of immune cell infiltration into the ischemic myocardium and can provide greater insight into the underlying complex and dynamic repair mechanisms. Molecular imaging of inflammation in the context of MI may help with stratification of patients at high risk of adverse ventricular remodeling post-MI which may be of diagnostic, therapeutic, and prognostic value. Novel radiopharmaceuticals may additionally provide a way to combine patient monitoring and therapy. In spite of great advances in recent years in the field of imaging sciences, clinicians still need to overcome some obstacles to a wider implementation of inflammation imaging post-MI. This review focuses on inflammation as a molecular imaging target and its potential implication in prognosis and therapeutic guidance.
Collapse
Affiliation(s)
- Janette Iking
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany.,Department of Cardiology I for Coronary and Peripheral Vascular Disease, and Heart Failure, University Hospital Münster, Münster, Germany
| | - Jasmin Klose
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | | | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | | |
Collapse
|
11
|
Masterson CH, Curley GF, Laffey JG. Modulating the distribution and fate of exogenously delivered MSCs to enhance therapeutic potential: knowns and unknowns. Intensive Care Med Exp 2019; 7:41. [PMID: 31346794 PMCID: PMC6658643 DOI: 10.1186/s40635-019-0235-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are undergoing intensive translational research for several debilitating conditions, including critical illnesses such as ARDS and sepsis. MSCs exert diverse biologic effects via their interaction with host tissues, via mechanisms that require the MSC to be in close proximity to the area of injury. Fully harnessing the therapeutic potential of advanced medicinal therapeutic products such as MSCs and their successful translation to clinical use requires a detailed understanding of MSC distribution and persistence in the injured tissues. Key aspects include understanding MSC distribution within the body, the response of the host to MSC administration, and the ultimate fate of exogenously administered MSCs within the host. Factors affecting this interaction include the MSC tissue source, the in vitro MSC culture conditions, the route of MSC administration and the specific issues relating to the target disease state, each of which remains to be fully characterised. Understanding these factors may generate strategies to modify MSC distribution and fate that may enhance their therapeutic effect. This review will examine our understanding of the mechanisms of action of MSCs, the early and late phase distribution kinetics of MSCs following in vivo administration, the ultimate fate of MSCs following administration and the potential importance of these MSC properties to their therapeutic effects. We will critique current cellular imaging and tracking methodologies used to track exogenous MSCs and their suitability for use in patients, discuss the insights they provide into the distribution and fate of MSCs after administration, and suggest strategies by which MSC biodistribution and fate may be modulated for therapeutic effect and clinical use. In conclusion, a better understanding of patterns of biodistribution and of the fate of MSCs will add important additional safety data regarding MSCs, address regulatory requirements, and may uncover strategies to increase the distribution and/or persistence of MSC at the sites of injury, potentially increasing their therapeutic potential for multiple disorders.
Collapse
Affiliation(s)
- Claire H Masterson
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland.,School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Gerard F Curley
- Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland Education and Research Centre Smurfit Building, Beaumont Hospital, Dublin, 9, Ireland
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland. .,School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland. .,Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, SAOLTA Hospital Group, Galway, Ireland.
| |
Collapse
|
12
|
Dhada KS, Hernandez DS, Suggs LJ. In Vivo Photoacoustic Tracking of Mesenchymal Stem Cell Viability. ACS NANO 2019; 13:7791-7799. [PMID: 31250647 PMCID: PMC7155740 DOI: 10.1021/acsnano.9b01802] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Adult stem cell therapy has demonstrated improved outcomes for treating cardiovascular diseases in preclinical trials. The development of imaging tools may increase our understanding of the mechanisms of stem cell therapy, and a variety of imaging tools have been developed to image transplanted stem cells in vivo; however, they lack the ability to interrogate stem cell function longitudinally. Here, we report the use of a nanoparticle-based contrast agent that can track stem cell viability using photoacoustic imaging. The contrast agent consists of inert gold nanorods coated with IR775c, a reactive oxygen species (ROS) sensitive near-infrared dye. Upon cell death, stem cells produce ROS to degrade the cell. Using this feature of stem cells, the viability can be measured by comparing the IR775c signal to the ROS insensitive gold nanorod signal, which can also be used to track stem cell location. The nanoprobe was successfully loaded into mesenchymal stem cells (MSCs), and then, MSCs were transplanted into the lower limb of a mouse and imaged using combined ultrasound and photoacoustic imaging. MSC viability was assessed using the nanoprobe and displayed significant cell death within 24 h and an estimated 5% viability after 10 days. This nanoparticle system allows for longitudinal tracking of MSC viability in vivo with high spatial and temporal resolution which other imaging modalities currently cannot achieve.
Collapse
Affiliation(s)
- Kabir S. Dhada
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Derek S. Hernandez
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | | |
Collapse
|
13
|
Breuls N, Giacomazzi G, Sampaolesi M. (Epi)genetic Modifications in Myogenic Stem Cells: From Novel Insights to Therapeutic Perspectives. Cells 2019; 8:cells8050429. [PMID: 31075875 PMCID: PMC6562881 DOI: 10.3390/cells8050429] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/17/2022] Open
Abstract
The skeletal muscle is considered to be an ideal target for stem cell therapy as it has an inherent regenerative capacity. Upon injury, the satellite cells, muscle stem cells that reside under the basal lamina of the myofibres, start to differentiate in order to reconstitute the myofibres while maintaining the initial stem cell pool. In recent years, it has become more and more evident that epigenetic mechanisms such as histon modifications, DNA methylations and microRNA modulations play a pivatol role in this differentiation process. By understanding the mechanisms behind myogenesis, researchers are able to use this knowledge to enhance the differentiation and engraftment potential of different muscle stem cells. Besides manipulation on an epigenetic level, recent advances in the field of genome-engineering allow site-specific modifications in the genome of these stem cells. Combining epigenetic control of the stem cell fate with the ability to site-specifically correct mutations or add genes for further cell control, can increase the use of stem cells as treatment of muscular dystrophies drastically. In this review, we will discuss the advances that have been made in genome-engineering and the epigenetic regulation of muscle stem cells and how this knowledge can help to get stem cell therapy to its full potential.
Collapse
Affiliation(s)
- Natacha Breuls
- Translational Cardiomyology Lab, Department of Development and Regeneration, Stem Cell Institute Leuven, 3000 KU Leuven, Belgium.
| | - Giorgia Giacomazzi
- Translational Cardiomyology Lab, Department of Development and Regeneration, Stem Cell Institute Leuven, 3000 KU Leuven, Belgium.
| | - Maurilio Sampaolesi
- Translational Cardiomyology Lab, Department of Development and Regeneration, Stem Cell Institute Leuven, 3000 KU Leuven, Belgium.
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, and Interuniversity Institute of Myology, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
14
|
Socan A, Petrik M, Kolenc Peitl P, Krošelj M, Rangger C, Novy Z, Svajger U, Gmeiner T, Decristoforo C. On-cartridge preparation and evaluation of 68Ga-, 89Zr- and 64Cu-precursors for cell radiolabelling. Nucl Med Biol 2019; 71:23-31. [PMID: 31128475 DOI: 10.1016/j.nucmedbio.2019.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/17/2019] [Accepted: 04/05/2019] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Indium-111 when formulated as indium-111 oxine remains the gold standard for long term cell tracking, whereas radiometals for improved PET applications still have to be established. We here describe the on-cartridge formation of gallium-68, zirconium-89 and copper-64 complexes in small volumes suitable for cell labelling, including labelling of red blood cells (RBC) and white blood cells (WBC) and their biological evaluation in vivo. METHODS Small volumes (1-2 mL) of tracers (oxine, tropolone) were directly prepared on an anion exchange cartridge (Sep-Pak QMA). Cells were radiolabelled and the labelling efficiency and efflux were evaluated. The in vivo biodistribution of copper-64-labelled WBC using [64Cu][Cu(oxinate)2] and [64Cu][Cu(tropolonate)2] was monitored in an infection and inflammation animal model using BALB/c mice. RESULTS On-cartridge concentration of gallium-68, zirconium-89 and copper-64 enabled formation of oxine and tropolone tracers in small volumes with good yields (≥50%) and quality (extraction ≥90%). Prepared tracers radiolabelled the RBC comparable to indium-111 tracers and in vivo biodistribution of copper-64 labelled WBC showed clear accumulation of cells at the site of infection and inflammation. CONCLUSIONS This on-cartridge preparation method enables simple formation of various PET tracers for cell radiolabelling. Zirconium-89 and copper-64 tracers radiolabelled cells with sufficient stability. Due to their longer half-life this approach could be promising for routine applications where longer evaluation periods for cell tracking are needed. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE This novel approach for on-cartridge concentration and preparation of oxine and tropolone precursors with different positron emitters, in small volume and suitable pH, offers a versatile tool towards cell labelling for preclinical and clinical PET applications.
Collapse
Affiliation(s)
- A Socan
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - M Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - P Kolenc Peitl
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - M Krošelj
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - C Rangger
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Z Novy
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - U Svajger
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - T Gmeiner
- Institue of Pharmacy, Faculty of Pharmacy, Ljubljana, Slovenia
| | - C Decristoforo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
15
|
Neyrinck K, Breuls N, Holvoet B, Oosterlinck W, Wolfs E, Vanbilloen H, Gheysens O, Duelen R, Gsell W, Lambrichts I, Himmelreich U, Verfaillie CM, Sampaolesi M, Deroose CM. The human somatostatin receptor type 2 as an imaging and suicide reporter gene for pluripotent stem cell-derived therapy of myocardial infarction. Am J Cancer Res 2018; 8:2799-2813. [PMID: 29774076 PMCID: PMC5957010 DOI: 10.7150/thno.22980] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 02/28/2018] [Indexed: 12/12/2022] Open
Abstract
Rationale: Pluripotent stem cells (PSCs) are being investigated as a cell source for regenerative medicine since they provide an infinitive pool of cells that are able to differentiate towards every cell type of the body. One possible therapeutic application involves the use of these cells to treat myocardial infarction (MI), a condition where billions of cardiomyocytes (CMs) are lost. Although several protocols have been developed to differentiate PSCs towards CMs, none of these provide a completely pure population, thereby still posing a risk for neoplastic teratoma formation. Therefore, we developed a strategy to (i) monitor cell behavior noninvasively via site-specific integration of firefly luciferase (Fluc) and the human positron emission tomography (PET) imaging reporter genes, sodium iodide symporter (hNIS) and somatostatin receptor type 2 (hSSTr2), and (ii) perform hSSTr2-mediated suicide gene therapy via the clinically used radiopharmacon 177Lu-DOTATATE. Methods: Human embryonic stem cells (ESCs) were gene-edited via zinc finger nucleases to express Fluc and either hNIS or hSSTr2 in the safe harbor locus, adeno-associated virus integration site 1. Firstly, these cells were exposed to 4.8 MBq 177Lu-DOTATATE in vitro and cell survival was monitored via bioluminescence imaging (BLI). Afterwards, hNIS+ and hSSTr2+ ESCs were transplanted subcutaneously and teratomas were allowed to form. At day 59, baseline 124I and 68Ga-DOTATATE PET and BLI scans were performed. The day after, animals received either saline or 55 MBq 177Lu-DOTATATE. Weekly BLI scans were performed, accompanied by 124I and 68Ga-DOTATATE PET scans at days 87 and 88, respectively. Finally, hSSTr2+ ESCs were differentiated towards CMs and transplanted intramyocardially in the border zone of an infarct that was induced by left anterior descending coronary artery ligation. After transplantation, the animals were monitored via BLI and PET, while global cardiac function was evaluated using cardiac magnetic resonance imaging. Results: Teratoma growth of both hNIS+ and hSSTr2+ ESCs could be followed noninvasively over time by both PET and BLI. After 177Lu-DOTATATE administration, successful cell killing of the hSSTr2+ ESCs was achieved both in vitro and in vivo, indicated by reductions in total tracer lesion uptake, BLI signal and teratoma volume. As undifferentiated hSSTr2+ ESCs are not therapeutically relevant, they were differentiated towards CMs and injected in immune-deficient mice with a MI. Long-term cell survival could be monitored without uncontrolled cell proliferation. However, no improvement in the left ventricular ejection fraction was observed. Conclusion: We developed isogenic hSSTr2-expressing ESCs that allow noninvasive cell monitoring in the context of PSC-derived regenerative therapy. Furthermore, we are the first to use the hSSTr2 not only as an imaging reporter gene, but also as a suicide mechanism for radionuclide therapy in the setting of PSC-derived cell treatment.
Collapse
|
16
|
Zini C, Venneri MA, Miglietta S, Caruso D, Porta N, Isidori AM, Fiore D, Gianfrilli D, Petrozza V, Laghi A. USPIO‐labeling in M1 and M2‐polarized macrophages: An in vitro study using a clinical magnetic resonance scanner. J Cell Physiol 2018; 233:5823-5828. [DOI: 10.1002/jcp.26360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/29/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Chiara Zini
- Faculty of Medicine and DentistryDepartment of Radiological SciencesOncology and PathologyUniversity of Rome “Sapienza”LatinaItaly
| | - Mary A. Venneri
- Faculty of Medicine and DentistryDepartment of Experimental MedicineUniversity of Rome “Sapienza”RomeItaly
| | - Selenia Miglietta
- Department of Anatomy, Histology, Forensic Medicine and OrthopaedicsElectron Microscopy Unit, Laboratory “Pietro M. Motta”University of Rome “Sapienza”RomeItaly
| | - Damiano Caruso
- Faculty of Medicine and DentistryDepartment of Radiological SciencesOncology and PathologyUniversity of Rome “Sapienza”LatinaItaly
| | - Natale Porta
- Faculty of Pharmacy and MedicineDepartment of Medico‐Surgical Sciences and BiotechnologiesUniversity of Rome “Sapienza”LatinaItaly
| | - Andrea M. Isidori
- Faculty of Medicine and DentistryDepartment of Experimental MedicineUniversity of Rome “Sapienza”RomeItaly
| | - Daniela Fiore
- Faculty of Medicine and DentistryDepartment of Experimental MedicineUniversity of Rome “Sapienza”RomeItaly
| | - Daniele Gianfrilli
- Faculty of Medicine and DentistryDepartment of Experimental MedicineUniversity of Rome “Sapienza”RomeItaly
| | - Vincenzo Petrozza
- Faculty of Pharmacy and MedicineDepartment of Medico‐Surgical Sciences and BiotechnologiesUniversity of Rome “Sapienza”LatinaItaly
| | - Andrea Laghi
- Faculty of Medicine and DentistryDepartment of Radiological SciencesOncology and PathologyUniversity of Rome “Sapienza”LatinaItaly
| |
Collapse
|
17
|
Zheng Y, Qin J, Wang X, Peng Z, Hou P, Lu X. Dynamic imaging of allogeneic adipose-derived regenerative cells transplanted in ischemic hind limb of apolipoprotein E mouse model. Int J Nanomedicine 2016; 12:61-71. [PMID: 28053524 PMCID: PMC5191626 DOI: 10.2147/ijn.s118328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Transplantation of allogeneic adipose-derived regenerative cells (ADRCs) is a promising treatment modality for severe ischemic diseases. However, minimal information is available on the in vivo effects, fate, and migration of ADRCs, as well as the mechanisms of their therapeutic angiogenesis. Materials and methods In this study, green fluorescent protein-expressing ADRCs (GFP-ADRCs) were obtained, labeled with acetylated 3-aminopropyltrimethoxysilane (APTS)-coated iron oxide nanoparticles (APTS NPs), and injected into an old apolipoprotein E knockout (ApoE-KO) mouse model with hind limb ischemia. Then, 3.0 T magnetic resonance imaging (MRI) was performed to dynamically trace the role of ADRCs targeting hind limb ischemia in the ApoE-KO mice model. Results Labeled cells were visualized as large hypointense spots in ischemic muscles by serial 3.0 T MRI scans during a 4-week follow-up. The presence of labeled GFP-ADRCs was confirmed by Prussian blue staining and fluorescence microscopy on postmortem specimens. Conclusion This study showed that allogeneic ADRCs offer great potential application for therapeutic angiogenesis in severe ischemic disease based on the efficacy and feasibility of ADRC transplantation and on the available amounts of tissue.
Collapse
Affiliation(s)
- Yi Zheng
- Department of General Surgery, The Fourth Affiliated Hospital of Guangxi Medical University, Guangxi
| | - Jinbao Qin
- Department of Vascular Surgery, School of Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University
| | - Xin Wang
- Department of Vascular Surgery, School of Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University
| | - Zhiyou Peng
- Department of Vascular Surgery, School of Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University
| | - Peiyong Hou
- Department of General Surgery, The Fourth Affiliated Hospital of Guangxi Medical University, Guangxi
| | - Xinwu Lu
- Department of Vascular Surgery, School of Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University; Vascular Center of Shanghai JiaoTong University, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Schmuck EG, Koch JM, Centanni JM, Hacker TA, Braun RK, Eldridge M, Hei DJ, Hematti P, Raval AN. Biodistribution and Clearance of Human Mesenchymal Stem Cells by Quantitative Three-Dimensional Cryo-Imaging After Intravenous Infusion in a Rat Lung Injury Model. Stem Cells Transl Med 2016; 5:1668-1675. [PMID: 27460855 PMCID: PMC5189648 DOI: 10.5966/sctm.2015-0379] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 05/13/2016] [Indexed: 12/30/2022] Open
Abstract
To study three-dimensional (3D) cryo-imaging to measure cell biodistribution and clearance after intravenous infusion, the authors established a lung injury model in rats. Human mesenchymal stem cells (hMSCs) labeled with QTracker were infused via jugular vein. Organs were cryopreserved, followed by 3D cryo-imaging. At 60 minutes, 82 ± 9.7% of cells were detected, and at day 2, 0.06% of cells were detected. hMSCs were retained primarily in the liver, with fewer detected in lungs and spleen. Cell tracking is a critical component of the safety and efficacy evaluation of therapeutic cell products. To date, cell-tracking modalities have been hampered by poor resolution, low sensitivity, and inability to track cells beyond the shortterm. Three-dimensional (3D) cryo-imaging coregisters fluorescent and bright-field microcopy images and allows for single-cell quantification within a 3D organ volume. We hypothesized that 3D cryo-imaging could be used to measure cell biodistribution and clearance after intravenous infusion in a rat lung injury model compared with normal rats. A bleomycin lung injury model was established in Sprague-Dawley rats (n = 12). Human mesenchymal stem cells (hMSCs) labeled with QTracker655 were infused via jugular vein. After 2, 4, or 8 days, a second dose of hMSCs labeled with QTracker605 was infused, and animals were euthanized after 60, 120, or 240 minutes. Lungs, liver, spleen, heart, kidney, testis, and intestine were cryopreserved, followed by 3D cryo-imaging of each organ. At 60 minutes, 82% ± 9.7% of cells were detected; detection decreased to 60% ± 17% and 66% ± 22% at 120 and 240 minutes, respectively. At day 2, 0.06% of cells were detected, and this level remained constant at days 4 and 8 postinfusion. At 60, 120, and 240 minutes, 99.7% of detected cells were found in the liver, lungs, and spleen, with cells primarily retained in the liver. This is the first study using 3D cryo-imaging to track hMSCs in a rat lung injury model. hMSCs were retained primarily in the liver, with fewer detected in lungs and spleen. Significance Effective bench-to-bedside clinical translation of cellular therapies requires careful understanding of cell fate through tracking. Tracking cells is important to measure cell retention so that delivery methods and cell dose can be optimized and so that biodistribution and clearance can be defined to better understand potential off-target toxicity and redosing strategies. This article demonstrates, for the first time, the use of three-dimensional cryo-imaging for single-cell quantitative tracking of intravenous infused clinical-grade mesenchymal stem cells in a clinically relevant model of lung injury. The important information learned in this study will help guide future clinical and translational stem cell therapies for lung injuries.
Collapse
Affiliation(s)
- Eric G Schmuck
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Jill M Koch
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - John M Centanni
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Timothy A Hacker
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Rudolf K Braun
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA
| | - Marlowe Eldridge
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA
| | - Derek J Hei
- Waisman Biomanufacturing, Madison, Wisconsin, USA
| | - Peiman Hematti
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Amish N Raval
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
19
|
Gervois P, Wolfs E, Ratajczak J, Dillen Y, Vangansewinkel T, Hilkens P, Bronckaers A, Lambrichts I, Struys T. Stem Cell-Based Therapies for Ischemic Stroke: Preclinical Results and the Potential of Imaging-Assisted Evaluation of Donor Cell Fate and Mechanisms of Brain Regeneration. Med Res Rev 2016; 36:1080-1126. [DOI: 10.1002/med.21400] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/27/2016] [Accepted: 06/17/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Pascal Gervois
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Esther Wolfs
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Jessica Ratajczak
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Yörg Dillen
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Tim Vangansewinkel
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Petra Hilkens
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Annelies Bronckaers
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Ivo Lambrichts
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Tom Struys
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| |
Collapse
|
20
|
Increased Understanding of Stem Cell Behavior in Neurodegenerative and Neuromuscular Disorders by Use of Noninvasive Cell Imaging. Stem Cells Int 2016; 2016:6235687. [PMID: 26997958 PMCID: PMC4779824 DOI: 10.1155/2016/6235687] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/13/2022] Open
Abstract
Numerous neurodegenerative and neuromuscular disorders are associated with cell-specific depletion in the human body. This imbalance in tissue homeostasis is in healthy individuals repaired by the presence of endogenous stem cells that can replace the lost cell type. However, in most disorders, a genetic origin or limited presence or exhaustion of stem cells impairs correct cell replacement. During the last 30 years, methods to readily isolate and expand stem cells have been developed and this resulted in a major change in the regenerative medicine field as it generates sufficient amount of cells for human transplantation applications. Furthermore, stem cells have been shown to release cytokines with beneficial effects for several diseases. At present however, clinical stem cell transplantations studies are struggling to demonstrate clinical efficacy despite promising preclinical results. Therefore, to allow stem cell therapy to achieve its full potential, more insight in their in vivo behavior has to be achieved. Different methods to noninvasively monitor these cells have been developed and are discussed. In some cases, stem cell monitoring even reached the clinical setting. We anticipate that by further exploring these imaging possibilities and unraveling their in vivo behavior further improvement in stem cell transplantations will be achieved.
Collapse
|
21
|
Wu CG, Zhang JC, Xie CQ, Parolini O, Silini A, Huang YZ, Lian B, Zhang M, Huang YC, Deng L. In vivo tracking of human placenta derived mesenchymal stem cells in nude mice via ¹⁴C-TdR labeling. BMC Biotechnol 2015; 15:55. [PMID: 26070459 PMCID: PMC4465458 DOI: 10.1186/s12896-015-0174-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 05/29/2015] [Indexed: 02/05/2023] Open
Abstract
Background In order to shed light on the regenerative mechanism of mesenchymal stem cells (MSCs) in vivo, the bio-distribution profile of implanted cells using a stable and long-term tracking method is needed. We herein investigated the bio-distribution of human placental deciduas basalis derived MSCs (termed as PDB-MSCs) in nude mice after intravenous injection by carbon radioisotope labeling thymidine (14C-TdR), which is able to incorporate into new DNA strands during cell replication. Results The proliferation rate and radioactive emission of human PDB-MSCs after labeled with different concentrations of 14C-TdR were measured. PDB-MSCs labeled with 1 μCi possessed high radioactivity, and the biological characteristics (i.e. morphology, colony forming ability, differentiation capabilities, karyotype and cell cycle) showed no significant changes after labeling. Thus, 1 μCi was the optimal concentration in this experimental design. In nude mice, 1 × 10614C-TdR-labeled PDB-MSCs were injected intravenously and the organs were collected at days 1, 2, 3, 5, 30 and 180 after injection, respectively. Radiolabeled PDB-MSCs were found mainly in the lung, liver, spleen, stomach and left femur of the recipient nude mice at the whole observation period. Conclusions This work provided solid evidence that 14C-TdR labeling did not alter the biological characteristics of human placental MSCs, and that this labeling method has potential to decrease the signal from non-infused or dead cells for cell tracking. Therefore, this labeling technique can be utilized to quantify the infused cells after long-term follow-up in pre-clinical studies.
Collapse
Affiliation(s)
- Cheng-Guang Wu
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Ji-Chun Zhang
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Cheng-Quan Xie
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Ornella Parolini
- Centro di Ricerca E.Menni, Fondazione Poliambulanza, Brescia, Italy.
| | - Antonietta Silini
- Centro di Ricerca E.Menni, Fondazione Poliambulanza, Brescia, Italy.
| | - Yi-Zhou Huang
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Bing Lian
- West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China.
| | - Min Zhang
- Center Laboratory For Isotopy, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Yong-Can Huang
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| | - Li Deng
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|