1
|
Khan S, Zhong X, Das N, Yu JH, Natarajan A, Anders D, Pratx G. Efficient radiolabeling of mesoporous silica nanoparticles for single-cell PET imaging. Eur J Nucl Med Mol Imaging 2025; 52:1778-1790. [PMID: 39729092 PMCID: PMC11928280 DOI: 10.1007/s00259-024-07027-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE Nanoparticles are highly efficient vectors for ferrying contrast agents across cell membranes, enabling ultra-sensitive in vivo tracking of single cells with positron emission tomography (PET). However, this approach must be fully characterized and understood before it can be reliably implemented for routine applications. METHODS We developed a Langmuir adsorption model that accurately describes the process of labeling mesoporous silica nanoparticles (MSNP) with 68Ga. We compared the binding efficiency of three different nanoparticle systems by fitting the model to experimental data. We then chose the MSNP with the highest affinity for 68Ga to study uptake and efflux kinetics in cancer cells. After intracardiac injection of 50-100 cells in mice, PET imaging was performed to test the effectiveness of cellular radiolabeling. RESULTS We found that highly porous mesoporous nanoparticles (d = 100 nm) with MCM-41 pore structures can achieve radiolabeling efficiency > 30 GBq/mg using 68Ga, without the need for any chelator. These 68Ga conjugated particles showed strong serum stability in vitro. In mice, the 68Ga-MSNPs predominantly accumulated in the liver with a high signal-to-background ratio and no bladder signal, indicating excellent stability of the labeled nanoparticles in vivo. Additionally, these MSNPs were efficiently taken up by B16F10 and MDA-MB-231 cancer cells, as confirmed by confocal imaging, flow cytometry analysis, and gamma counting. Finally, cardiac injection of < 100 68Ga-MSNP-labeled cells allowed PET/CT tracking of these cells in various organs in mice. CONCLUSION We characterized the critical parameters of MSNP-mediated direct cellular radiolabeling to improve the use of these nanoparticles as cellular labels for highly sensitive preclinical PET imaging.
Collapse
Affiliation(s)
- Syamantak Khan
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Xiaoxu Zhong
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Neeladrisingha Das
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Jung Ho Yu
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - David Anders
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Guillem Pratx
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Gawne P, Man F, Blower PJ, T. M. de Rosales R. Direct Cell Radiolabeling for in Vivo Cell Tracking with PET and SPECT Imaging. Chem Rev 2022; 122:10266-10318. [PMID: 35549242 PMCID: PMC9185691 DOI: 10.1021/acs.chemrev.1c00767] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Indexed: 02/07/2023]
Abstract
The arrival of cell-based therapies is a revolution in medicine. However, its safe clinical application in a rational manner depends on reliable, clinically applicable methods for determining the fate and trafficking of therapeutic cells in vivo using medical imaging techniques─known as in vivo cell tracking. Radionuclide imaging using single photon emission computed tomography (SPECT) or positron emission tomography (PET) has several advantages over other imaging modalities for cell tracking because of its high sensitivity (requiring low amounts of probe per cell for imaging) and whole-body quantitative imaging capability using clinically available scanners. For cell tracking with radionuclides, ex vivo direct cell radiolabeling, that is, radiolabeling cells before their administration, is the simplest and most robust method, allowing labeling of any cell type without the need for genetic modification. This Review covers the development and application of direct cell radiolabeling probes utilizing a variety of chemical approaches: organic and inorganic/coordination (radio)chemistry, nanomaterials, and biochemistry. We describe the key early developments and the most recent advances in the field, identifying advantages and disadvantages of the different approaches and informing future development and choice of methods for clinical and preclinical application.
Collapse
Affiliation(s)
- Peter
J. Gawne
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Francis Man
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
- Institute
of Pharmaceutical Science, School of Cancer
and Pharmaceutical Sciences, King’s College London, London, SE1 9NH, U.K.
| | - Philip J. Blower
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Rafael T. M. de Rosales
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| |
Collapse
|
3
|
Syed MBJ, Fletcher AJ, Dweck MR. Imaging cellular activity and proliferation in the aortic wall. J Nucl Cardiol 2021; 28:1972-1975. [PMID: 31832885 DOI: 10.1007/s12350-019-01987-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 11/30/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Maaz B J Syed
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| | - Alexander J Fletcher
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Butler E, Schwettmann B, Geboers S, Hao G, Kim J, Nham K, Sun X, Laetsch TW, Xu L, Williams NS, Skapek SX. Functional imaging of RAS pathway targeting in malignant peripheral nerve sheath tumor cells and xenografts. Pediatr Blood Cancer 2020; 67:e28639. [PMID: 32975370 DOI: 10.1002/pbc.28639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Malignant peripheral nerve sheath tumor (MPNST) is an aggressive form of soft-tissue sarcoma (STS) in children. Despite intensive therapy, relatively few children with metastatic and unresectable disease survive beyond three years. RAS pathway activation is common in MPNST, suggesting MEK pathway inhibition as a targeted therapy, but the impact on clinical outcome has been small to date. PROCEDURE We conducted preclinical pharmacokinetic (PK) and pharmacodynamic studies of two MEK inhibitors, trametinib and selumetinib, in two MPNST models and analyzed tumors for intratumor drug levels. We then investigated 3'-deoxy-3'-[18 F]fluorothymidine (18 F-FLT) PET imaging followed by 18 F-FDG PET/CT imaging of MPNST xenografts coupled to short-term or longer-term treatment with selumetinib focusing on PET-based imaging as a biomarker of MEK inhibition. RESULTS Trametinib decreased pERK expression in MPNST xenografts but did not prolong survival or decrease Ki67 expression. In contrast, selumetinib prolonged survival of animals bearing MPNST xenografts, and this correlated with decreased pERK and Ki67 staining. PK studies revealed a significantly higher fraction of unbound selumetinib within a responsive MPNST xenograft model. Thymidine uptake, assessed by 18 F-FLT PET/CT, positively correlated with Ki67 expression in different xenograft models and in response to selumetinib. CONCLUSION The ability of MEK inhibitors to control MPNST growth cannot simply be predicted by serum drug levels or drug-induced changes in pERK expression. Tumor cell proliferation assessed by 18 F-FLT PET imaging might be useful as an early response marker to targeted therapies, including MEK inhibition, where a primary effect is cell-cycle arrest.
Collapse
Affiliation(s)
- Erin Butler
- Department of Pediatrics Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Blake Schwettmann
- Department of Pediatrics Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sophie Geboers
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Guiyang Hao
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jiwoong Kim
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kien Nham
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas.,The Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Theodore W Laetsch
- Department of Pediatrics Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.,The Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lin Xu
- Department of Pediatrics Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.,Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,The Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stephen X Skapek
- Department of Pediatrics Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
5
|
Belderbos S, González-Gómez MA, Cleeren F, Wouters J, Piñeiro Y, Deroose CM, Coosemans A, Gsell W, Bormans G, Rivas J, Himmelreich U. Simultaneous in vivo PET/MRI using fluorine-18 labeled Fe 3O 4@Al(OH) 3 nanoparticles: comparison of nanoparticle and nanoparticle-labeled stem cell distribution. EJNMMI Res 2020; 10:73. [PMID: 32607918 PMCID: PMC7326875 DOI: 10.1186/s13550-020-00655-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have shown potential for treatment of different diseases. However, their working mechanism is still unknown. To elucidate this, the non-invasive and longitudinal tracking of MSCs would be beneficial. Both iron oxide-based nanoparticles (Fe3O4 NPs) for magnetic resonance imaging (MRI) and radiotracers for positron emission tomography (PET) have shown potential as in vivo cell imaging agents. However, they are limited by their negative contrast and lack of spatial information as well as short half-life, respectively. In this proof-of-principle study, we evaluated the potential of Fe3O4@Al(OH)3 NPs as dual PET/MRI contrast agents, as they allow stable binding of [18F]F- ions to the NPs and thus, NP visualization and quantification with both imaging modalities. RESULTS 18F-labeled Fe3O4@Al(OH)3 NPs (radiolabeled NPs) or mouse MSCs (mMSCs) labeled with these radiolabeled NPs were intravenously injected in healthy C57Bl/6 mice, and their biodistribution was studied using simultaneous PET/MRI acquisition. While liver uptake of radiolabeled NPs was seen with both PET and MRI, mMSCs uptake in the lungs could only be observed with PET. Even some initial loss of fluoride label did not impair NPs/mMSCs visualization. Furthermore, no negative effects on blood cell populations were seen after injection of either the NPs or mMSCs, indicating good biocompatibility. CONCLUSION We present the application of novel 18F-labeled Fe3O4@Al(OH)3 NPs as safe cell tracking agents for simultaneous PET/MRI. Combining both modalities allows fast and easy NP and mMSC localization and quantification using PET at early time points, while MRI provides high-resolution, anatomic background information and long-term NP follow-up, hereby overcoming limitations of the individual imaging modalities.
Collapse
Affiliation(s)
- Sarah Belderbos
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000, Leuven, Belgium
| | - Manuel Antonio González-Gómez
- NANOMAG Group, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Jens Wouters
- Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, 3000, Leuven, Belgium
| | - Yolanda Piñeiro
- NANOMAG Group, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Christophe M Deroose
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven/UZ Leuven, 3000, Leuven, Belgium
| | - An Coosemans
- Laboratory for Tumor Immunology and Immunotherapy, ImmunOvar Research Group, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000, Leuven, Belgium.,Department of Gynaecology and Obstetrics, UZ Leuven, 3000, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000, Leuven, Belgium
| | - Guy Bormans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Jose Rivas
- NANOMAG Group, Department of Applied Physics, Technological Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
6
|
Zhao Y, Tietz O, Kuan WL, Haji-Dheere AK, Thompson S, Vallin B, Ronchi E, Tóth G, Klenerman D, Aigbirhio FI. A fluorescent molecular imaging probe with selectivity for soluble tau aggregated protein. Chem Sci 2020; 11:4773-4778. [PMID: 34122933 PMCID: PMC8159431 DOI: 10.1039/c9sc05620c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/19/2020] [Indexed: 01/25/2023] Open
Abstract
Soluble forms of aggregated tau misfolded protein, generally termed oligomers, are considered to be the most toxic species of the different assembly states that are the pathological components of neurodegenerative disorders. Therefore, a critical biomedical need exists for imaging probes that can identify and quantify them. We have designed and synthesized a novel fluorescent probe, pTP-TFE for which binding and selectivity profiles towards aggregated tau and Aβ proteins were assessed. Our results have shown pTP-TFE to be selective for early forms of soluble tau aggregates, with high affinity of dissociation constants (K d) = 66 nM, and tenfold selectivity over mature tau fibrils. Furthermore, we found that pTP-TFE is selective for tau over Aβ aggregates and had good cell permeability. This selectivity of pTP-TFE towards early forms of aggregated tau protein ex vivo was also supported with studies on human brain tissue containing tau and Aβ pathology. To the best of our knowledge, this is the first fluorescent molecule to be reported to have this form of selectivity profile, which suggests that pTP-TFE is a unique probe candidate for imaging-based detection of early stages of Alzheimer's disease and other tauopathies.
Collapse
Affiliation(s)
- Yanyan Zhao
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge Cambridge UK
| | - Ole Tietz
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge Cambridge UK
| | - Wei-Li Kuan
- John van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge Cambridge UK
| | - Abdul K Haji-Dheere
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge Cambridge UK
| | - Stephen Thompson
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge Cambridge UK
| | - Benjamin Vallin
- John van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge Cambridge UK
| | - Elisabetta Ronchi
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge Cambridge UK
| | - Gergely Tóth
- TTK-NAP B - Drug Discovery Research Group - Neurodegenerative Diseases, Institute of Organic Chemistry, Research Center for Natural Sciences Budapest Hungary
| | - David Klenerman
- Department of Chemistry, University of Cambridge Lensfield Road Cambridge UK
- UK Dementia Research Institute, University of Cambridge Cambridge UK
| | - Franklin I Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge Cambridge UK
| |
Collapse
|
7
|
Perrin J, Capitao M, Mougin-Degraef M, Guérard F, Faivre-Chauvet A, Rbah-Vidal L, Gaschet J, Guilloux Y, Kraeber-Bodéré F, Chérel M, Barbet J. Cell Tracking in Cancer Immunotherapy. Front Med (Lausanne) 2020; 7:34. [PMID: 32118018 PMCID: PMC7033605 DOI: 10.3389/fmed.2020.00034] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
The impressive development of cancer immunotherapy in the last few years originates from a more precise understanding of control mechanisms in the immune system leading to the discovery of new targets and new therapeutic tools. Since different stages of disease progression elicit different local and systemic inflammatory responses, the ability to longitudinally interrogate the migration and expansion of immune cells throughout the whole body will greatly facilitate disease characterization and guide selection of appropriate treatment regiments. While using radiolabeled white blood cells to detect inflammatory lesions has been a classical nuclear medicine technique for years, new non-invasive methods for monitoring the distribution and migration of biologically active cells in living organisms have emerged. They are designed to improve detection sensitivity and allow for a better preservation of cell activity and integrity. These methods include the monitoring of therapeutic cells but also of all cells related to a specific disease or therapeutic approach. Labeling of therapeutic cells for imaging may be performed in vitro, with some limitations on sensitivity and duration of observation. Alternatively, in vivo cell tracking may be performed by genetically engineering cells or mice so that may be revealed through imaging. In addition, SPECT or PET imaging based on monoclonal antibodies has been used to detect tumors in the human body for years. They may be used to detect and quantify the presence of specific cells within cancer lesions. These methods have been the object of several recent reviews that have concentrated on technical aspects, stressing the differences between direct and indirect labeling. They are briefly described here by distinguishing ex vivo (labeling cells with paramagnetic, radioactive, or fluorescent tracers) and in vivo (in vivo capture of injected radioactive, fluorescent or luminescent tracers, or by using labeled antibodies, ligands, or pre-targeted clickable substrates) imaging methods. This review focuses on cell tracking in specific therapeutic applications, namely cell therapy, and particularly CAR (Chimeric Antigen Receptor) T-cell therapy, which is a fast-growing research field with various therapeutic indications. The potential impact of imaging on the progress of these new therapeutic modalities is discussed.
Collapse
Affiliation(s)
- Justine Perrin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marisa Capitao
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marie Mougin-Degraef
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - François Guérard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - Latifa Rbah-Vidal
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Joëlle Gaschet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yannick Guilloux
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | | |
Collapse
|
8
|
Chetty SS, Praneetha S, Vadivel Murugan A, Govarthanan K, Verma RS. Human Umbilical Cord Wharton's Jelly-Derived Mesenchymal Stem Cells Labeled with Mn 2+ and Gd 3+ Co-Doped CuInS 2-ZnS Nanocrystals for Multimodality Imaging in a Tumor Mice Model. ACS APPLIED MATERIALS & INTERFACES 2020; 12:3415-3429. [PMID: 31875453 DOI: 10.1021/acsami.9b19054] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mesenchymal stem cell (MSCs) therapy has recently received profound interest as a targeting platform in cancer theranostics because of inherent tumor-homing abilities. However, the terminal tracking of MSCs engraftment by fluorescent in situ hybridization, immuno-histochemistry, and flow-cytometry techniques to translate into clinics is still challenging because of a dearth of inherent MSCs-specific markers and FDA approval for genetic modifications of MSCs. To address this challenge, a cost-effective noninvasive imaging technology based on multifunctional nanocrystals (NCs) with enhanced detection sensitivity, spatial-temporal resolution, and deep-tissue diagnosis is needed to be developed to track the transplanted stem cells. A hassle-free labeling of human umbilical cord Wharton's Jelly (WJ)-derived MSCs with Mn2+ and Gd3+ co-doped CuInS2-ZnS (CIS-ZMGS) NCs has been demonstrated in 2 h without requiring an electroporation process or transfection agents. It has been found that WJ-MSCs labeling did not affect their multilineage differentiation (adipocyte, osteocyte, chondrocyte), immuno-phenotypes (CD44+, CD105+, CD90+), protein (β-actin, vimentin, CD73, α-SMCA), and gene expressions. Interestingly, CIS-ZMGS-NCs-labeled WJ-MSCs exhibit near-infrared (NIR) fluorescence with a quantum yield of 84%, radiant intensity of ∼3.999 × 1011 (p/s/cm2/sr)/(μW/cm2), magnetic relaxivity (longitudinal r1 = 2.26 mM-1 s-1, transverse r2 = 16.47 mM-1 s-1), and X-ray attenuation (78 HU) potential for early noninvasive multimodality imaging of a subcutaneous melanoma in B16F10-tumor-bearing C57BL/6 mice in 6 h. The ex vivo imaging and inductively coupled plasma mass-spectroscopy analyses of excised organs along with confocal microscopy and immunofluorescence of tumor results also significantly confirmed the positive tropism of CIS-ZMGS-NCs-labeled WJ-MSCs in the tumor environment. Hence, we propose the magnetofluorescent CIS-ZMGS-NCs-labeled WJ-MSCs as a next-generation nanobioprobe of three commonly used imaging modalities for stem cell-assisted anticancer therapy and tracking tissue/organ regenerations.
Collapse
Affiliation(s)
- Shashank Shankar Chetty
- Advanced Functional Nanostructured Materials Laboratory, Centre for Nanoscience and Technology, Madanjeet School of Green Energy Technologies , Pondicherry University (A Central University) , Puducherry 605014 , India
| | - Selvarasu Praneetha
- Advanced Functional Nanostructured Materials Laboratory, Centre for Nanoscience and Technology, Madanjeet School of Green Energy Technologies , Pondicherry University (A Central University) , Puducherry 605014 , India
| | - Arumugam Vadivel Murugan
- Advanced Functional Nanostructured Materials Laboratory, Centre for Nanoscience and Technology, Madanjeet School of Green Energy Technologies , Pondicherry University (A Central University) , Puducherry 605014 , India
| | - Kavitha Govarthanan
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology , Indian Institute of Technology-Madras (IIT-M) , Chennai 600036 , India
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology , Indian Institute of Technology-Madras (IIT-M) , Chennai 600036 , India
| |
Collapse
|
9
|
Chetty SS, Praneetha S, Govarthanan K, Verma RS, Vadivel Murugan A. Noninvasive Tracking and Regenerative Capabilities of Transplanted Human Umbilical Cord-Derived Mesenchymal Stem Cells Labeled with I-III-IV Semiconducting Nanocrystals in Liver-Injured Living Mice. ACS APPLIED MATERIALS & INTERFACES 2019; 11:8763-8778. [PMID: 30741534 DOI: 10.1021/acsami.8b19953] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Acute liver injury is a critical syndrome ascribed to prevalent death of hepatocytes and imperatively requires liver transplantation. Such a methodology is certainly hampered due to the deficit of healthy donors. In this regard, stem cell-based regenerative therapies are attractive in repairing injured tissues and organs for medical applications. However, it is crucial to understand the migration, engraftment, and regeneration capabilities of transplanted stem cells in the living animal models. For the first time, we demonstrate rapid labeling of umbilical cord-derived mesenchymal stem cells (MSCs) with near-infrared (NIR)-fluorescent CuInS2-ZnS nanocrystals (CIZS-NCs) to develop innovative nanobioconjugates (MSCs-CIZS-NBCs) that exhibit 98% labeling efficiency. Before nanobioconjugate synthesis, the pristine CIZS-NCs were prepared via a two-step, hot-injection, rapid and low-cost domestic-microwave-refluxing (MW-R) method within 6 min. The as-synthesized CIZS-NCs display high photoluminescence quantum yield (∼88%) and long-lived lifetime (23.4 μs). In contrast to unlabeled MSCs, the MSCs-CIZS nanobioconjugates show excellent biocompatibility without affecting the stemness, as confirmed by cell viability, immunophenotyping (CD44+, CD105+, CD90+), multi-lineage-specific gene expressions, and differentiation into adipocytes, osteocytes, and chondrocytes. The in vivo fluorescence tracking analyses revealed that the MSCs-CIZS-NBCs after tail-vein injection were initially trapped in the lungs and gradually engrafted in the injured liver within 2 h. The regeneration potential of MSCs-CIZS-NBCs was confirmed via renewal of the portal tract composed of portal veins, bile ducts, and hepatic arteries around the hepatocytes. Consequently, no apparent inflammations, necrosis, or apoptosis was observed in the acetaminophen (APAP)-induced liver-injured BALB/c mice model over 3 days after transplantation, as corroborated using laser-scanning confocal microscopy and histopathological and hematological analyses. Hence, our innovative NIR-fluorescent MSCs-CIZS-NBCs offer an off-the-self technology for noninvasive tracking of transplanted MSCs in an acute-liver-injured animal model for future image-guided cell-therapies.
Collapse
Affiliation(s)
- Shashank Shankar Chetty
- Advanced Functional Nanostructured Materials Laboratory, Centre for Nanoscience and Technology, Madanjeet School of Green Energy Technologies , Pondicherry University (A Central University) , Puducherry 605 014 , India
| | - Selvarasu Praneetha
- Advanced Functional Nanostructured Materials Laboratory, Centre for Nanoscience and Technology, Madanjeet School of Green Energy Technologies , Pondicherry University (A Central University) , Puducherry 605 014 , India
| | - Kavitha Govarthanan
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology , Indian Institute of Technology-Madras (IIT-M) , Chennai 600 036 , India
| | - Rama Shanker Verma
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology , Indian Institute of Technology-Madras (IIT-M) , Chennai 600 036 , India
| | - Arumugam Vadivel Murugan
- Advanced Functional Nanostructured Materials Laboratory, Centre for Nanoscience and Technology, Madanjeet School of Green Energy Technologies , Pondicherry University (A Central University) , Puducherry 605 014 , India
| |
Collapse
|
10
|
Rothe M, Jahn A, Weiss K, Hwang JH, Szendroedi J, Kelm M, Schrader J, Roden M, Flögel U, Bönner F. In vivo 19F MR inflammation imaging after myocardial infarction in a large animal model at 3 T. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2018; 32:5-13. [DOI: 10.1007/s10334-018-0714-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/04/2018] [Accepted: 10/22/2018] [Indexed: 12/19/2022]
|
11
|
MacAskill MG, Saif J, Condie A, Jansen MA, MacGillivray TJ, Tavares AAS, Fleisinger L, Spencer HL, Besnier M, Martin E, Biglino G, Newby DE, Hadoke PWF, Mountford JC, Emanueli C, Baker AH. Robust Revascularization in Models of Limb Ischemia Using a Clinically Translatable Human Stem Cell-Derived Endothelial Cell Product. Mol Ther 2018; 26:1669-1684. [PMID: 29703701 PMCID: PMC6035339 DOI: 10.1016/j.ymthe.2018.03.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cell-derived differentiated endothelial cells offer high potential in regenerative medicine in the cardiovascular system. With the aim of translating the use of a human stem cell-derived endothelial cell product (hESC-ECP) for treatment of critical limb ischemia (CLI) in man, we report a good manufacturing practice (GMP)-compatible protocol and detailed cell tracking and efficacy data in multiple preclinical models. The clinical-grade cell line RC11 was used to generate hESC-ECP, which was identified as mostly endothelial (60% CD31+/CD144+), with the remainder of the subset expressing various pericyte/mesenchymal stem cell markers. Cell tracking using MRI, PET, and qPCR in a murine model of limb ischemia demonstrated that hESC-ECP was detectable up to day 7 following injection. Efficacy in several murine models of limb ischemia (immunocompromised/immunocompetent mice and mice with either type I/II diabetes mellitus) demonstrated significantly increased blood perfusion and capillary density. Overall, we demonstrate a GMP-compatible hESC-ECP that improved ischemic limb perfusion and increased local angiogenesis without engraftment, paving the way for translation of this therapy.
Collapse
Affiliation(s)
- Mark G MacAskill
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Jaimy Saif
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Alison Condie
- Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Maurits A Jansen
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | | | - Adriana A S Tavares
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Lucija Fleisinger
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Helen L Spencer
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Marie Besnier
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Ernesto Martin
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Giovanni Biglino
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - David E Newby
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Patrick W F Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Joanne C Mountford
- Scottish National Blood Transfusion Service, Edinburgh, UK; Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Costanza Emanueli
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK; National Heart and Lung Institute, Imperial College London, London, UK
| | - Andrew H Baker
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
12
|
Facilitated recruitment of mesenchymal stromal cells by bone marrow concentrate and platelet rich plasma. PLoS One 2018; 13:e0194567. [PMID: 29566102 PMCID: PMC5864018 DOI: 10.1371/journal.pone.0194567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 02/14/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Biologics containing growth factors are frequently used to enhance healing after musculoskeletal injuries. One mechanism of action is thought to be though the ability of biologics to induce homing and migration of endogenous mesenchymal stromal cells (MSCs) to a target tissue. However, the ability of biologics to stimulate chemotaxis (directed migration of cells) and chemokinesis (increase rate of cell migration) of MSCs is unknown. HYPOTHESIS/PURPOSE The aim of this study was to directly compare the ability of biologics including platelet rich plasma (PRP) and bone marrow concentrate (BMC) to induce MSC migration. The hypothesis was that leukocyte-low platelet rich plasma (Llo PRP) would induce migration to a greater extent than leukocyte-high platelet rich plasma (Lhi PRP) or BMC. METHODS Bone marrow-derived MSCs were isolated from 8 horses. Migration of MSCs toward a biologic (BMC, Llo PRP, and Lhi PRP) or the positive control platelet derived growth factor (PDGF) was continuously traced and measured for 24hrs using time-lapse microscopy and a microfluidics device. Cell migration, chemotaxis and chemokinesis were determined by measurements of displacement, number of cells migrated, and cell flux. RESULTS All biologics resulted in a significantly greater percentage of MSCs migrated compared to the positive control (PDGF). MSCs migrated further toward BMC compared to Llo PRP. Cell migration, measured as cell flux, was greater toward BMC and Lhi PRP than Llo PRP. CONCLUSION The biologics BMC and Lhi PRP elicit greater chemotaxis and chemokinesis of MSCs than Llo PRP. However, all biologics recruited the same number of MSCs suggesting that differences in other regenerative effects, such as growth factor concentration, between biologics should be strongly considered when choosing a biologic for treatment of musculoskeletal injuries. The results of this study have the potential to reduce the need, risks, and costs associated with MSC culture and delivery.
Collapse
|