1
|
Zhang YC, Wang JW, Wu Y, Tao Q, Wang FF, Wang N, Ji XR, Li YG, Yu S, Zhang JZ. Multimodal Magnetic Resonance and Fluorescence Imaging of the Induced Pluripotent Stem Cell Transplantation in the Brain. Mol Biol 2022. [DOI: 10.1134/s0026893322030153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abstract
The understanding of the engrafted cell behaviors such as the survival, growth and distribution is the prerequisite to optimize cell therapy, and a multimodal imaging at both anatomical and molecular levels is designed to achieve this goal. We constructed a lentiviral vector carrying genes of ferritin heavy chain 1 (FTH1), near-infrared fluorescent protein (iRFP) and enhanced green fluorescent protein (egfp), and established the induced pluripotent stem cells (iPSCs) culture stably expressing these three reporter genes. These iPSCs showed green and near-infrared fluorescence as well as the iron uptake capacity in vitro. After transplanted the labeled iPSCs into the rat brain, the engrafted cells could be in vivo imaged using magnetic resonance imaging (MRI) and near-infrared fluorescent imaging (NIF) up to 60 days at the anatomical level. Moreover, these cells could be detected using EGFP immunostaining and Prussian blue stain at the cellular level. The developed approach provides a novel tool to study behaviors of the transplanted cells in a multimodal way, which will be valuable for the effectiveness and safety evaluation of cell therapy.
Collapse
|
2
|
Dallet L, Stanicki D, Voisin P, Miraux S, Ribot EJ. Micron-sized iron oxide particles for both MRI cell tracking and magnetic fluid hyperthermia treatment. Sci Rep 2021; 11:3286. [PMID: 33558583 PMCID: PMC7870900 DOI: 10.1038/s41598-021-82095-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 01/14/2021] [Indexed: 12/17/2022] Open
Abstract
Iron oxide particles (IOP) are commonly used for Cellular Magnetic Resonance Imaging (MRI) and in combination with several treatments, like Magnetic Fluid Hyperthermia (MFH), due to the rise in temperature they provoke under an Alternating Magnetic Field (AMF). Micrometric IOP have a high sensitivity of detection. Nevertheless, little is known about their internalization processes or their potential heat power. Two micrometric commercial IOP (from Bangs Laboratories and Chemicell) were characterized by Transmission Electron Microscopy (TEM) and their endocytic pathways into glioma cells were analyzed. Their Specific Absorption Rate (SAR) and cytotoxicity were evaluated using a commercial AMF inductor. T2-weighted imaging was used to monitor tumor growth in vivo after MFH treatment in mice. The two micron-sized IOP had similar structures and r2 relaxivities (100 mM-1 s-1) but involved different endocytic pathways. Only ScreenMAG particles generated a significant rise in temperature following AMF (SAR = 113 W g-1 Fe). After 1 h of AMF exposure, 60% of ScreenMAG-labeled cells died. Translated to a glioma model, 89% of mice responded to the treatment with smaller tumor volume 42 days post-implantation. Micrometric particles were investigated from their characterization to their intracellular internalization pathways and applied in one in vivo cancer treatment, i.e. MFH.
Collapse
Affiliation(s)
- Laurence Dallet
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536, CNRS/Univ. Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - Dimitri Stanicki
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, 19 avenue Maistriau, 7000, Mons, Belgium
| | - Pierre Voisin
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536, CNRS/Univ. Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - Sylvain Miraux
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536, CNRS/Univ. Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - Emeline J Ribot
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536, CNRS/Univ. Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France.
| |
Collapse
|
3
|
Rbia N, Bulstra LF, Thaler R, Hovius SER, van Wijnen AJ, Shin AY. In Vivo Survival of Mesenchymal Stromal Cell-Enhanced Decellularized Nerve Grafts for Segmental Peripheral Nerve Reconstruction. J Hand Surg Am 2019; 44:514.e1-514.e11. [PMID: 30301645 DOI: 10.1016/j.jhsa.2018.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/09/2018] [Accepted: 07/18/2018] [Indexed: 02/02/2023]
Abstract
PURPOSE Adipose-derived mesenchymal stromal cells (MSCs) have emerged as promising tools for peripheral nerve reconstruction. There is a paucity of information regarding the ultimate survivorship of implanted MSCs or whether these cells remain where they are placed. The aim of the present study was to track the in vivo distribution and survival of MSCs seeded on a decellularized nerve allograft reconstruction of a peripheral nerve defect using luciferase-based bioluminescence imaging (BLI). METHODS To determine the in vivo survivability of MSCs, autologous Lewis rat MSCs were stably labeled with luciferase by lentiviral particles. Labeled cells were dynamically seeded onto a Sprague Dawley decellularized rat nerve allograft and used to bridge a 10-mm sciatic nerve defect. The MSC survival was determined by performing in vivo BLI to detect living cells. Twelve animals were examined at 24 hours after implantation, 3, 7, 9, 11, and 14 days, and at daily intervals thereafter if signals were still present. RESULTS Labeled MSCs could be detected for up to 29 days. Gradually diminishing BLI signals were observed within the first week following implantation. Implanted MSCs were not detected anywhere other than the site of surgery. CONCLUSIONS The MSCs seeded on decellularized nerve allografts can survive in vivo but have finite survival after implantation. There was no evidence of migration of MSCs to surrounding tissues. CLINICAL RELEVANCE The findings support a therapeutic approach that combines MSCs with a biological scaffold for peripheral nerve surgery. It provides understanding of the viability and distribution of implanted MSCs, which is a prerequisite before clinical translation can be considered.
Collapse
Affiliation(s)
- Nadia Rbia
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Liselotte F Bulstra
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | - Steven E R Hovius
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
4
|
Li M, Wang Y, Liu M, Lan X. Multimodality reporter gene imaging: Construction strategies and application. Theranostics 2018; 8:2954-2973. [PMID: 29896296 PMCID: PMC5996353 DOI: 10.7150/thno.24108] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/06/2018] [Indexed: 12/11/2022] Open
Abstract
Molecular imaging has played an important role in the noninvasive exploration of multiple biological processes. Reporter gene imaging is a key part of molecular imaging. By combining with a reporter probe, a reporter protein can induce the accumulation of specific signals that are detectable by an imaging device to provide indirect information of reporter gene expression in living subjects. There are many types of reporter genes and each corresponding imaging technique has its own advantages and drawbacks. Fused reporter genes or single reporter genes with products detectable by multiple imaging modalities can compensate for the disadvantages and potentiate the advantages of each modality. Reporter gene multimodality imaging could be applied to trace implanted cells, monitor gene therapy, assess endogenous molecular events, screen drugs, etc. Although several types of multimodality imaging apparatus and multimodality reporter genes are available, more sophisticated detectors and multimodality reporter gene systems are needed.
Collapse
Affiliation(s)
- Mengting Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| | - Yichun Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| | - Mei Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| |
Collapse
|
5
|
Continual conscious bioluminescent imaging in freely moving somatotransgenic mice. Sci Rep 2017; 7:6374. [PMID: 28743959 PMCID: PMC5526882 DOI: 10.1038/s41598-017-06696-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/20/2017] [Indexed: 12/31/2022] Open
Abstract
Luciferase bioimaging in living animals is increasingly being applied in many fields of biomedical research. Rodent imaging usually involves anaesthetising the animal during data capture, however, the biological consequences of anaesthesia have been largely overlooked. We have evaluated luciferase bioimaging in conscious, unrestrained mice after neonatal intracranial or intravascular administration of lentiviral, luciferase reporter cassettes (biosensors); we present real-time analyses from the first day of life to adulthood. Anaesthetics have been shown to exert both neurotoxic and neuroprotective effects during development and in models of brain injury. Mice subjected to bioimaging after neonatal intracranial or intravascular administration of biosensors, targeting the brain and liver retrospectively showed no significant difference in luciferase expression when conscious or unconscious throughout development. We applied conscious bioimaging to the assessment of NFκB and STAT3 transcription factor activated reporters during the earliest stages of development in living, unrestrained pups. Our data showed unique longitudinal activities for NFκB and STAT3 in the brain of conscious mice. Conscious bioimaging was applied to a neonatal mouse model of cerebral palsy (Hypoxic-Ischaemic Encephalopathy). Imaging of NFκB reporter before and after surgery showed a significant increase in luciferase expression, coinciding with secondary energy failure, in lesioned mice compared to controls.
Collapse
|
6
|
Jasmin, de Souza GT, Louzada RA, Rosado-de-Castro PH, Mendez-Otero R, Campos de Carvalho AC. Tracking stem cells with superparamagnetic iron oxide nanoparticles: perspectives and considerations. Int J Nanomedicine 2017; 12:779-793. [PMID: 28182122 PMCID: PMC5279820 DOI: 10.2147/ijn.s126530] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been used for diagnoses in biomedical applications, due to their unique properties and their apparent safety for humans. In general, SPIONs do not seem to produce cell damage, although their long-term in vivo effects continue to be investigated. The possibility of efficiently labeling cells with these magnetic nanoparticles has stimulated their use to noninvasively track cells by magnetic resonance imaging after transplantation. SPIONs are attracting increasing attention and are one of the preferred methods for cell labeling and tracking in preclinical and clinical studies. For clinical protocol approval of magnetic-labeled cell tracking, it is essential to expand our knowledge of the time course of SPIONs after cell incorporation and transplantation. This review focuses on the recent advances in tracking SPION-labeled stem cells, analyzing the possibilities and limitations of their use, not only focusing on myocardial infarction but also discussing other models.
Collapse
Affiliation(s)
- Jasmin
- NUMPEX-Bio, Federal University of Rio de Janeiro, Duque de Caxias, RJ
| | - Gustavo Torres de Souza
- Laboratory of Animal Reproduction, Embrapa Dairy Cattle, Juiz de Fora, MG
- Laboratory of Genetics, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Ruy Andrade Louzada
- Institute Gustave-Roussy of Oncology, Paris-Sud University, Villejuif, France
| | | | - Rosalia Mendez-Otero
- Institute Carlos Chagas Filho of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
7
|
Selt M, Tennstaedt A, Beyrau A, Nelles M, Schneider G, Löwik C, Hoehn M. In Vivo Non-Invasive Tracking of Macrophage Recruitment to Experimental Stroke. PLoS One 2016; 11:e0156626. [PMID: 27341631 PMCID: PMC4920382 DOI: 10.1371/journal.pone.0156626] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/17/2016] [Indexed: 11/25/2022] Open
Abstract
Brain-infiltrating monocyte-derived macrophages are one of the key players in the local immune response after stroke. It is now widely accepted that the inflammatory response is not an exclusively destructive process. However, the underlying molecular mechanisms needed for proper regulation still remain to be elucidated. Here, we propose an in vitro labelling strategy for multimodal in vivo observation of macrophage dynamics distinguished from brain-residing microglia response. Prior to intracerebral transplantation into the striatum of recipient mice or systemic administration, monocytes and macrophages, isolated from luciferase-expressing mice, were labelled with superparamagnetic iron oxide particles. Temporo-spatial localization was monitored by magnetic resonance imaging, whereas survival of grafted cells was investigated using bioluminescence imaging. The labelling procedure of the isolated cells did not significantly influence cell characteristics and resulted in detection of as few as 500 labelled cells in vivo. Two weeks after stereotactic transplantation, the luciferase signal was sustained traceable, with approximately 18% of the original luciferase signal detectable for monocytes and about 30% for macrophages. Hypointensity in MRI of the graft appeared unaltered in spatial location. In a therapeutically relevant approach, systemic cell administration after stroke resulted in accumulation mostly in thoracic regions, as could be visualized with BLI. For detection of homing to ischemic brain tissue more cells need to be administered. Nevertheless, during parallel MRI sessions recruitment of i.v. injected cells to the lesion site could be detected by day 2 post stroke as scattered hypointense signal voids. With further increase in sensitivity, our multi-facetted labelling strategy will provide the basis for in vivo tracking and fate specification of tissue-infiltrating macrophages and their distinct role in stroke-related neuro-inflammation.
Collapse
Affiliation(s)
- Marion Selt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Annette Tennstaedt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Andreas Beyrau
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Melanie Nelles
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Gabriele Schneider
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Clemens Löwik
- Dept. of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
- Dept. of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Percuros B.V., Enschede, The Netherlands
- * E-mail:
| |
Collapse
|
8
|
Praet J, Santermans E, Daans J, Le Blon D, Hoornaert C, Goossens H, Hens N, Van Der Linden A, Berneman Z, Ponsaerts P. Early Inflammatory Responses following Cell Grafting in the CNS Trigger Activation of the Subventricular Zone: A Proposed Model of Sequential Cellular Events. Cell Transplant 2015; 24:1481-92. [DOI: 10.3727/096368914x682800] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
While multiple rodent preclinical studies, and to a lesser extent human clinical trials, claim the feasibility, safety, and potential clinical benefit of cell grafting in the central nervous system (CNS), currently only little convincing knowledge exists regarding the actual fate of the grafted cells and their effect on the surrounding environment (or vice versa). Our preceding studies already indicated that only a minor fraction of the initially grafted cell population survives the grafting process, while the surviving cell population becomes invaded by highly activated microglia/macrophages and surrounded by reactive astrogliosis. In the current study, we further elaborate on early cellular and inflammatory events following syngeneic grafting of eGFP mouse embryonic fibroblasts (mEFs) in the CNS of immunocompetent mice. Based on obtained quantitative histological data, we here propose a detailed mathematically derived working model that sequentially comprises hypoxia-induced apoptosis of grafted mEFs, neutrophil invasion, neoangiogenesis, microglia/macrophage recruitment, astrogliosis, and eventually survival of a limited number of grafted mEFs. Simultaneously, we observed that the cellular events following mEF grafting activates the subventricular zone neural stem and progenitor cell compartment. This proposed model therefore further contributes to our understanding of cell graft-induced cellular responses and will eventually allow for successful manipulation of this intervention.
Collapse
Affiliation(s)
- Jelle Praet
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- BioImaging Laboratory, University of Antwerp, Wilrijk, Belgium
| | - Eva Santermans
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium
| | - Jasmijn Daans
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Debbie Le Blon
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Chloé Hoornaert
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium
- Centre for Health Economic Research and Modeling Infectious Diseases (Chermid), University of Antwerp, Wilrijk, Belgium
| | | | - Zwi Berneman
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Peter Ponsaerts
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
9
|
Distinct spatial distribution of microglia and macrophages following mesenchymal stem cell implantation in mouse brain. Immunol Cell Biol 2014; 92:650-8. [DOI: 10.1038/icb.2014.49] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/09/2014] [Accepted: 05/23/2014] [Indexed: 12/18/2022]
|
10
|
Roose D, Leroux F, De Vocht N, Guglielmetti C, Pintelon I, Adriaensen D, Ponsaerts P, Van der Linden A, Bals S. Multimodal imaging of micron-sized iron oxide particles following in vitro and in vivo uptake by stem cells: down to the nanometer scale. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 9:400-8. [PMID: 24753446 DOI: 10.1002/cmmi.1594] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 10/10/2013] [Accepted: 01/08/2014] [Indexed: 11/08/2022]
Abstract
In this study, the interaction between cells and micron-sized paramagnetic iron oxide (MPIO) particles was investigated by characterizing MPIO in their original state, and after cellular uptake in vitro as well as in vivo. Moreover, MPIO in the olfactory bulb were studied 9 months after injection. Using various imaging techniques, cell-MPIO interactions were investigated with increasing spatial resolution. Live cell confocal microscopy demonstrated that MPIO co-localize with lysosomes after in vitro cellular uptake. In more detail, a membrane surrounding the MPIO was observed by high-angle annular dark-field scanning transmission electron microscopy (HAADF-STEM). Following MPIO uptake in vivo, the same cell-MPIO interaction was observed by HAADF-STEM in the subventricular zone at 1 week and in the olfactory bulb at 9 months after MPIO injection. These findings provide proof for the current hypothesis that MPIO are internalized by the cell through endocytosis. The results also show MPIO are not biodegradable, even after 9 months in the brain. Moreover, they show the possibility of HAADF-STEM generating information on the labeled cell as well as on the MPIO. In summary, the methodology presented here provides a systematic route to investigate the interaction between cells and nanoparticles from the micrometer level down to the nanometer level and beyond.
Collapse
Affiliation(s)
- Dimitri Roose
- EMAT, University of Antwerp, Antwerp, Belgium; Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Boehm-Sturm P, Aswendt M, Minassian A, Michalk S, Mengler L, Adamczak J, Mezzanotte L, Löwik C, Hoehn M. A multi-modality platform to image stem cell graft survival in the naïve and stroke-damaged mouse brain. Biomaterials 2013; 35:2218-26. [PMID: 24355489 DOI: 10.1016/j.biomaterials.2013.11.085] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 11/27/2013] [Indexed: 02/08/2023]
Abstract
Neural stem cell implantations have been extensively investigated for treatment of brain diseases such as stroke. In order to follow the localization and functional status of cells after implantation noninvasive imaging is essential. Therefore, we developed a comprehensive multi-modality platform for in vivo imaging of graft localization, density, and survival using 19F magnetic resonance imaging in combination with bioluminescence imaging. We quantitatively analyzed cell graft survival over the first 4 weeks after transplantation in both healthy and stroke-damaged mouse brain and correlated our findings of graft vitality with the host innate immune response. The multi-modality imaging platform will help to improve cell therapy also in context other than stroke and to gain indispensable information for clinical translation.
Collapse
Affiliation(s)
- Philipp Boehm-Sturm
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Markus Aswendt
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Anuka Minassian
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Stefanie Michalk
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Luam Mengler
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Joanna Adamczak
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Laura Mezzanotte
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clemens Löwik
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mathias Hoehn
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany; Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
12
|
De Vocht N, Praet J, Reekmans K, Le Blon D, Hoornaert C, Daans J, Berneman Z, Van der Linden A, Ponsaerts P. Tackling the physiological barriers for successful mesenchymal stem cell transplantation into the central nervous system. Stem Cell Res Ther 2013; 4:101. [PMID: 23998480 PMCID: PMC3854758 DOI: 10.1186/scrt312] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over the past decade a lot of research has been performed towards the therapeutic use of mesenchymal stem cells (MSCs) in neurodegenerative and neuroinflammatory diseases. MSCs have shown to be beneficial in different preclinical studies of central nervous system (CNS) disorders due to their immunomodulatory properties and their capacity to secrete various growth factors. Nevertheless, most of the transplanted cells die within the first hours after transplantation and induce a neuroinflammatory response. In order to increase the efficacy of MSC transplantation, it is thus imperative to completely characterise the mechanisms mediating neuroinflammation and cell death following MSC transplantation into the CNS. Consequently, different components of these cell death- and neuroinflammation-inducing pathways can be targeted in an attempt to improve the therapeutic potential of MSCs for CNS disorders.
Collapse
|
13
|
Boeykens N, Ponsaerts P, Van der Linden A, Berneman Z, Ysebaert D, De Greef K. Injury-dependent retention of intraportally administered mesenchymal stromal cells following partial hepatectomy of steatotic liver does not lead to improved liver recovery. PLoS One 2013; 8:e69092. [PMID: 23874878 PMCID: PMC3715456 DOI: 10.1371/journal.pone.0069092] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/04/2013] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to evaluate the effect of bone marrow-derived mesenchymal stromal cell (BM-MSC) administration on liver function following partial hepatectomy (PHx) of methionine/choline-deficient (MCD) diet induced steatotic livers in rodents. Here we identified and validated serum cholinesterase (CHE) and triglyceride (TG) levels as non-invasive markers to longitudinally monitor rat liver function. Using in vivo bioluminescence imaging, retention of BM-MSC in the liver was observed following intraportal administration, but not after intravenous administration. Therefore, BM-MSC were intraportally delivered to investigate the effect on liver recovery and/or regeneration after PHx. However, despite recovery to normal body weight, liver weight and NAS score, both serum CHE and TG levels of non-treated and cell-treated rats with PHx after MCD diet remained significantly lower as compared to those of control rats. Importantly, serum CHE levels, but not TG levels, of cell-treated rats remained significantly lower as compared to those of non-treated rats, thereby warranting that certain caution should be considered for future clinical application of IP BM-MSC administration in order to promote liver regeneration and/or function.
Collapse
Affiliation(s)
- Nele Boeykens
- Laboratory of Experimental Surgery, Antwerp Surgical Training and Research Centre, University of Antwerp/University Hospital of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | | | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Dirk Ysebaert
- Laboratory of Experimental Surgery, Antwerp Surgical Training and Research Centre, University of Antwerp/University Hospital of Antwerp, Antwerp, Belgium
- * E-mail:
| | - Kathleen De Greef
- Laboratory of Experimental Surgery, Antwerp Surgical Training and Research Centre, University of Antwerp/University Hospital of Antwerp, Antwerp, Belgium
| |
Collapse
|
14
|
Reekmans K, De Vocht N, Praet J, Le Blon D, Hoornaert C, Daans J, Van der Linden A, Berneman Z, Ponsaerts P. Quantitative evaluation of stem cell grafting in the central nervous system of mice by in vivo bioluminescence imaging and postmortem multicolor histological analysis. Methods Mol Biol 2013; 1052:125-141. [PMID: 23733539 DOI: 10.1007/7651_2013_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Stem cell transplantation in the central nervous system (CNS) is currently under intensive investigation as a novel therapeutic approach for a variety of brain disorders and/or injuries. However, one of the main hurdles at the moment is the lack of standardized procedures to evaluate cell graft survival and behavior following transplantation into CNS tissue, thereby leading to the publication of confusing and/or conflicting research results. In this chapter, we therefore provide validated in vivo bioluminescence and postmortem histological procedures to quantitatively determine: (a) the survival of grafted stem cells, and (b) the microglial and astroglial cell responses following cell grafting.
Collapse
Affiliation(s)
- Kristien Reekmans
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Campus Drie Eiken (CDE-S6.51), Universiteitsplein 1, Antwerp (Wilrijk), 2610, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Reekmans K, De Vocht N, Praet J, Fransen E, Le Blon D, Hoornaert C, Daans J, Goossens H, Van der Linden A, Berneman Z, Ponsaerts P. Spatiotemporal evolution of early innate immune responses triggered by neural stem cell grafting. Stem Cell Res Ther 2012; 3:56. [PMID: 23241452 PMCID: PMC3580486 DOI: 10.1186/scrt147] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/12/2012] [Indexed: 12/12/2022] Open
Abstract
Introduction Transplantation of neural stem cells (NSCs) is increasingly suggested to become part of future therapeutic approaches to improve functional outcome of various central nervous system disorders. However, recently it has become clear that only a small fraction of grafted NSCs display long-term survival in the (injured) adult mouse brain. Given the clinical invasiveness of NSC grafting into brain tissue, profound characterisation and understanding of early post-transplantation events is imperative to claim safety and efficacy of cell-based interventions. Methods Here, we applied in vivo bioluminescence imaging (BLI) and post-mortem quantitative histological analysis to determine the localisation and survival of grafted NSCs at early time points post-transplantation. Results An initial dramatic cell loss (up to 80% of grafted cells) due to apoptosis could be observed within the first 24 hours post-implantation, coinciding with a highly hypoxic NSC graft environment. Subsequently, strong spatiotemporal microglial and astroglial cell responses were initiated, which stabilised by day 5 post-implantation and remained present during the whole observation period. Moreover, the increase in astrocyte density was associated with a high degree of astroglial scarring within and surrounding the graft site. During the two-week follow up in this study, the NSC graft site underwent extensive remodelling with NSC graft survival further declining to around 1% of the initial number of grafted cells. Conclusions The present study quantitatively describes the early post-transplantation events following NSC grafting in the adult mouse brain and warrants that such intervention is directly associated with a high degree of cell loss, subsequently followed by strong glial cell responses.
Collapse
|
16
|
Reekmans K, Praet J, De Vocht N, Daans J, Van der Linden A, Berneman Z, Ponsaerts P. Stem cell therapy for multiple sclerosis: preclinical evidence beyond all doubt? Regen Med 2012; 7:245-59. [PMID: 22397612 DOI: 10.2217/rme.12.5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stem cell transplantation holds great promise for restoration of neural function in various neurodegenerative disorders, including multiple sclerosis (MS). However, many questions remain regarding the true efficacy and precise mode of action of stem cell-based therapeutic approaches. Therefore, in this article, we will first discuss the ideal route and/or timing of stem cell-based therapies for experimental autoimmune encephalomyelitis (EAE), the most used preclinical animal model for MS. Next, we will provide an overview of the proposed mechanisms that contribute to the beneficial effects of stem cell transplantation observed during the treatment of rodent EAE. Reviews of current and past literature clearly demonstrate conceptual changes in the development of stem cell-based approaches for EAE/MS, leading to the identification of several major challenges to be tackled before (stem) cell therapy for rodent EAE can be safely and successfully translated to human therapy for MS.
Collapse
Affiliation(s)
- Kristien Reekmans
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | | | | | | | | | | | | |
Collapse
|
17
|
De Vocht N, Lin D, Praet J, Hoornaert C, Reekmans K, Le Blon D, Daans J, Pauwels P, Goossens H, Hens N, Berneman Z, Van der Linden A, Ponsaerts P. Quantitative and phenotypic analysis of mesenchymal stromal cell graft survival and recognition by microglia and astrocytes in mouse brain. Immunobiology 2012; 218:696-705. [PMID: 22944251 DOI: 10.1016/j.imbio.2012.08.266] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 08/02/2012] [Accepted: 08/05/2012] [Indexed: 01/18/2023]
Abstract
Although cell transplantation is increasingly suggested to be beneficial for the treatment of various neurodegenerative diseases, the therapeutic application of such intervention is currently hindered by the limited knowledge regarding central nervous system (CNS) transplantation immunology. In this study, we aimed to investigate the early post transplantation innate immune events following grafting of autologous mesenchymal stromal cells (MSC) in the CNS of immune competent mice. First, the survival of grafted Luciferase/eGFP-expressing MSC (MSC-Luc/eGFP) was demonstrated to be stable from on day 3 post implantation using in vivo bioluminescence imaging (BLI), which was further confirmed by quantitative histological analysis of MSC-Luc/eGFP graft survival. Additional histological analyses at week 1 and week 2 post grafting revealed the appearance of (i) graft-surrounding/-invading Iba1+ microglia and (ii) graft-surrounding GFAP+ astrocytes, as compared to day 0 post grafting. While the density of graft-surrounding astrocytes and microglia did not change between week 1 and week 2 post grafting, the density of graft-invading microglia significantly decreased between week 1 and week 2 post implantation. However, despite the observed decrease in microglial density within the graft site, additional phenotypic analysis of graft-invading microglia, based on CD11b- and MHCII-expression, revealed >50% of graft-invading microglia at week 2 post implantation to display an activated status. Although microglial expression of CD11b and MHCII is already suggestive for a pro-inflammatory M1-oriented phenotype, the latter was further confirmed by: (i) the expression of NOS2 by microglia within the graft site, and (ii) the absence of arginase 1-expression, an enzyme known to suppress NO activity in M2-oriented microglia, on graft-surrounding and -invading microglia. In summary, we here provide a detailed phenotypic analysis of post transplantation innate immune events in the CNS of mice, and warrant that such intervention is associated with an M1-oriented microglia response and severe astrogliosis.
Collapse
Affiliation(s)
- Nathalie De Vocht
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
De Vocht N, Reekmans K, Bergwerf I, Praet J, Hoornaert C, Le Blon D, Daans J, Berneman Z, Van der Linden A, Ponsaerts P. Multimodal imaging of stem cell implantation in the central nervous system of mice. J Vis Exp 2012:e3906. [PMID: 22733218 DOI: 10.3791/3906] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During the past decade, stem cell transplantation has gained increasing interest as primary or secondary therapeutic modality for a variety of diseases, both in preclinical and clinical studies. However, to date results regarding functional outcome and/or tissue regeneration following stem cell transplantation are quite diverse. Generally, a clinical benefit is observed without profound understanding of the underlying mechanism(s). Therefore, multiple efforts have led to the development of different molecular imaging modalities to monitor stem cell grafting with the ultimate aim to accurately evaluate survival, fate and physiology of grafted stem cells and/or their micro-environment. Changes observed in one or more parameters determined by molecular imaging might be related to the observed clinical effect. In this context, our studies focus on the combined use of bioluminescence imaging (BLI), magnetic resonance imaging (MRI) and histological analysis to evaluate stem cell grafting. BLI is commonly used to non-invasively perform cell tracking and monitor cell survival in time following transplantation, based on a biochemical reaction where cells expressing the Luciferase-reporter gene are able to emit light following interaction with its substrate (e.g. D-luciferin). MRI on the other hand is a non-invasive technique which is clinically applicable and can be used to precisely locate cellular grafts with very high resolution, although its sensitivity highly depends on the contrast generated after cell labeling with an MRI contrast agent. Finally, post-mortem histological analysis is the method of choice to validate research results obtained with non-invasive techniques with highest resolution and sensitivity. Moreover end-point histological analysis allows us to perform detailed phenotypic analysis of grafted cells and/or the surrounding tissue, based on the use of fluorescent reporter proteins and/or direct cell labeling with specific antibodies. In summary, we here visually demonstrate the complementarities of BLI, MRI and histology to unravel different stem cell- and/or environment-associated characteristics following stem cell grafting in the CNS of mice. As an example, bone marrow-derived stromal cells, genetically engineered to express the enhanced Green Fluorescent Protein (eGFP) and firefly Luciferase (fLuc), and labeled with blue fluorescent micron-sized iron oxide particles (MPIOs), will be grafted in the CNS of immune-competent mice and outcome will be monitored by BLI, MRI and histology (Figure 1).
Collapse
|
19
|
Praet J, Reekmans K, Lin D, De Vocht N, Bergwerf I, Tambuyzer B, Daans J, Hens N, Goossens H, Pauwels P, Berneman Z, Van der Linden A, Ponsaerts P. Cell type-associated differences in migration, survival, and immunogenicity following grafting in CNS tissue. Cell Transplant 2012; 21:1867-81. [PMID: 22472278 DOI: 10.3727/096368912x636920] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell transplantation has been suggested to display several neuroprotective and/or neuroregenerative effects in animal models of central nervous system (CNS) trauma. However, while most studies report on clinical observations, currently little is known regarding the actual fate of the cell populations grafted and whether or how the brain's innate immune system, mainly directed by activated microglia and astrocytes, interacts with autologous cellular implants. In this study, we grafted well-characterized neural stem cell, mouse embryonic fibroblast, dendritic cell, bone marrow mononuclear cell, and splenocyte populations, all isolated or cultured from C57BL/6-eGFP transgenic mice, below the capsula externa (CE) of healthy C57BL/6 mice and below the inflamed/demyelinated CE of cuprizone-treated C57BL/6 mice. Two weeks postgrafting, an extensive quantitative multicolor histological analysis was performed in order (i) to quantify cell graft localization, migration, survival, and toxicity and (ii) to characterize endogenous CNS immune responses against the different cell grafts. Obtained results indicate dependence on the cell type grafted: (i) a different degree of cell graft migration, survival, and toxicity and (ii) a different organization of the endogenous immune response. Based on these observations, we warrant that further research should be undertaken to understand-and eventually control-cell graft-induced tissue damage and activation of the brain's innate immune system. The latter will be inevitable before cell grafting in the CNS can be performed safely and successfully in clinical settings.
Collapse
Affiliation(s)
- Jelle Praet
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Parolini O, Alviano F, Betz A, Bianchi D, Götherström C, Manuelpillai U, Mellor A, Ofir R, Ponsaerts P, Scherjon S, Weiss M, Wolbank S, Wood K, Borlongan C. Meeting report of the first conference of the International Placenta Stem Cell Society (IPLASS). Placenta 2011; 32 Suppl 4:S285-90. [PMID: 21575989 PMCID: PMC4437622 DOI: 10.1016/j.placenta.2011.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 04/22/2011] [Accepted: 04/26/2011] [Indexed: 12/14/2022]
Abstract
The International Placenta Stem Cell Society (IPLASS) was founded in June 2010. Its goal is to serve as a network for advancing research and clinical applications of stem/progenitor cells isolated from human term placental tissues, including the amnio-chorionic fetal membranes and Wharton's jelly. The commitment of the Society to champion placenta as a stem cell source was realized with the inaugural meeting of IPLASS held in Brescia, Italy, in October 2010. Officially designated as an EMBO-endorsed scientific activity, international experts in the field gathered for a 3-day meeting, which commenced with "Meet with the experts" sessions, IPLASS member and board meetings, and welcome remarks by Dr. Ornella Parolini, President of IPLASS. The evening's highlight was a keynote plenary lecture by Dr. Diana Bianchi. The subsequent scientific program consisted of morning and afternoon oral and poster presentations, followed by social events. Both provided many opportunities for intellectual exchange among the 120 multi-national participants. This allowed a methodical and deliberate evaluation of the status of placental cells in research in regenerative and reparative medicine. The meeting concluded with Dr. Parolini summarizing the meeting's highlights. This further prepared the fertile ground on which to build the promising potential of placental cell research. The second IPLASS meeting will take place in September 2012 in Vienna, Austria. This meeting report summarizes the thought-provoking lectures delivered at the first meeting of IPLASS.
Collapse
Affiliation(s)
- O. Parolini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Brescia, Italy
| | - F. Alviano
- Department of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - A.G. Betz
- Medical Research Council, Cambridge, UK
| | - D.W. Bianchi
- Mother Infant Research Institute, Tufts Medical Center, Boston, USA
| | - C. Götherström
- Karolinska Institutet, CLINTEC, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - U. Manuelpillai
- Monash Institute of Medical Research, Monash University, Clayton, Australia
| | - A.L. Mellor
- Immunotherapy Center, Medical College of Georgia, Augusta, GA, USA
| | - R. Ofir
- Pluristem Therapeutics Inc., Haifa, Israel
| | - P. Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - S.A. Scherjon
- Department of Immunohematology and Bloodbank, Leiden University Medical Center, Leiden, The Netherlands
| | - M.L. Weiss
- Department of Anatomy and Physiology, Kansas State University College of Veterinary Medicine, Manhattan, USA
| | - S. Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austrian Cluster for Tissue Regeneration, Austria
| | - K.J. Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - C.V. Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| |
Collapse
|