1
|
Sun A, Hayat H, Kenyon E, Quadri T, Amos D, Perkins K, Nigam S, Tarleton D, Mallett CL, Deng CX, Qiu Z, Li W, Sempere L, Fan J, Aguirre A, Wang P. Brown Adipose Tissue as a Unique Niche for Islet Organoid Transplantation: Insights From In Vivo Imaging. Transplant Direct 2024; 10:e1658. [PMID: 38881741 PMCID: PMC11177823 DOI: 10.1097/txd.0000000000001658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/15/2024] [Accepted: 04/03/2024] [Indexed: 06/18/2024] Open
Abstract
Background Transplantation of human-induced pluripotent stem cell (hiPSC)-derived islet organoids is a promising cell replacement therapy for type 1 diabetes (T1D). It is important to improve the efficacy of islet organoids transplantation by identifying new transplantation sites with high vascularization and sufficient accommodation to support graft survival with a high capacity for oxygen delivery. Methods A human-induced pluripotent stem cell line (hiPSCs-L1) was generated constitutively expressing luciferase. Luciferase-expressing hiPSCs were differentiated into islet organoids. The islet organoids were transplanted into the scapular brown adipose tissue (BAT) of nonobese diabetic/severe combined immunodeficiency disease (NOD/SCID) mice as the BAT group and under the left kidney capsule (KC) of NOD/SCID mice as a control group, respectively. Bioluminescence imaging (BLI) of the organoid grafts was performed on days 1, 7, 14, 28, 35, 42, 49, 56, and 63 posttransplantation. Results BLI signals were detected in all recipients, including both the BAT and control groups. The BLI signal gradually decreased in both BAT and KC groups. However, the graft BLI signal intensity under the left KC decreased substantially faster than that of the BAT. Furthermore, our data show that islet organoids transplanted into streptozotocin-induced diabetic mice restored normoglycemia. Positron emission tomography/MRI verified that the islet organoids were transplanted at the intended location in these diabetic mice. Immunofluorescence staining revealed the presence of functional organoid grafts, as confirmed by insulin and glucagon staining. Conclusions Our results demonstrate that BAT is a potentially desirable site for islet organoid transplantation for T1D therapy.
Collapse
Affiliation(s)
- Aixia Sun
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Hanaan Hayat
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Elizabeth Kenyon
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Tahnia Quadri
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Darius Amos
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Keenan Perkins
- Florida Agricultural and Mechanical University, Tallahassee, FL
| | - Saumya Nigam
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Deanna Tarleton
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Christiane L Mallett
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
| | - Cheri X Deng
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Zhen Qiu
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI
| | - Wen Li
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI
| | - Lorenzo Sempere
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Jinda Fan
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI
| | - Ping Wang
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| |
Collapse
|
2
|
Yun WS, Cho H, Jeon SI, Lim DK, Kim K. Fluorescence-Based Mono- and Multimodal Imaging for In Vivo Tracking of Mesenchymal Stem Cells. Biomolecules 2023; 13:1787. [PMID: 38136656 PMCID: PMC10742164 DOI: 10.3390/biom13121787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
The advancement of stem cell therapy has offered transformative therapeutic outcomes for a wide array of diseases over the past decades. Consequently, stem cell tracking has become significant in revealing the mechanisms of action and ensuring safe and effective treatments. Fluorescence stands out as a promising choice for stem cell tracking due to its myriad advantages, including high resolution, real-time monitoring, and multi-fluorescence detection. Furthermore, combining fluorescence with other tracking modalities-such as bioluminescence imaging (BLI), positron emission tomography (PET), photoacoustic (PA), computed tomography (CT), and magnetic resonance (MR)-can address the limitations of single fluorescence detection. This review initially introduces stem cell tracking using fluorescence imaging, detailing various labeling strategies such as green fluorescence protein (GFP) tagging, fluorescence dye labeling, and nanoparticle uptake. Subsequently, we present several combinations of strategies for efficient and precise detection.
Collapse
Affiliation(s)
- Wan Su Yun
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; (W.S.Y.); (D.-K.L.)
| | - Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman’s University, Seoul 03760, Republic of Korea; (H.C.); (S.I.J.)
| | - Seong Ik Jeon
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman’s University, Seoul 03760, Republic of Korea; (H.C.); (S.I.J.)
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; (W.S.Y.); (D.-K.L.)
| | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman’s University, Seoul 03760, Republic of Korea; (H.C.); (S.I.J.)
| |
Collapse
|
3
|
Petrella F, Cassina EM, Libretti L, Pirondini E, Raveglia F, Tuoro A. Mesenchymal Stromal Cell Therapy for Thoracic Surgeons: An Update. J Pers Med 2023; 13:1632. [PMID: 38138859 PMCID: PMC10744666 DOI: 10.3390/jpm13121632] [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: 10/20/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Stem cells are undifferentiated cells presenting extensive self-renewal features and the ability to differentiate "in vitro" and "in vivo" into a range of lineage cells, like chondrogenic, osteogenic and adipogenic lineages when cultured in specific inducing media. Two major domains of clinical applications of stem cells in thoracic surgery have been investigated: regenerative medicine, which is a section of translational research in tissue engineering focusing on the replacement, renewal or regeneration of cells, tissues and organs to re-establish damaged physiologic functions; drug loading and delivery, representing a new branch proposing stem cells as carriers to provide selected districts with anti-cancer agents for targeted treatments.
Collapse
Affiliation(s)
- Francesco Petrella
- Department of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (E.M.C.); (L.L.); (E.P.); (F.R.); (A.T.)
| | | | | | | | | | | |
Collapse
|
4
|
Differentiation Capacity of Bone Marrow-Derived Rat Mesenchymal Stem Cells from DsRed and Cre Transgenic Cre/ loxP Models. Cells 2022; 11:cells11172769. [PMID: 36078177 PMCID: PMC9455627 DOI: 10.3390/cells11172769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/26/2022] Open
Abstract
Cre/loxP recombination is a well-established technique increasingly used for modifying DNA both in vitro and in vivo. Nucleotide alterations can be edited in the genomes of mammalian cells, and genetic switches can be designed to target the expression or excision of a gene in any tissue at any time in animal models. In this study, we propose a system which worked via the Cre/loxP switch gene and DsRed/emGFP dual-color fluorescence imaging. Mesenchymal stem cells (MSCs) can be used to regenerate damaged tissue because of their differentiation capacity. Although previous studies have presented evidence of fusion of transplanted MSCs with recipient cells, the possibility of fusion in such cases remains debated. Moreover, the effects and biological implications of the fusion of MSCs at the tissue and organ level have not yet been elucidated. Thus, the method for determining this issue is significant and the models we proposed can illustrate the question. However, the transgenic rats exhibited growth slower than that of wild-type rats over several weeks. The effects on the stemness, proliferation, cell cycle, and differentiation ability of bone marrow–derived rat MSCs (BM-rMSCs) from the models were examined to ensure our design was appropriate for the in vivo application. We demonstrated that MSC surface markers were maintained in DsRed and Cre transgenic rMSCs (DsRed-rMSCs and Cre-rMSCs, respectively). A WST-8 assay revealed decreased proliferative activity in these DsRed-rMSCs and Cre-rMSCs; this result was validated through cell counting. Furthermore, cell cycle analysis indicated a decrease in the proportion of G1-phase cells and a concomitant increase in the proportion of S-phase cells. The levels of cell cycle–related proteins also decreased in the DsRed-rMSCs and Cre-rMSCs, implying decelerated phase transition. However, the BM-rMSCs collected from the transgenic rats did not exhibit altered adipogenesis, osteogenesis, or chondrogenesis. The specific markers of these types of differentiation were upregulated after induction. Therefore, BM-rMSCs from DsRed and Cre transgenic models can be used to investigate the behavior of MSCs and related mechanisms. Such application may further the development of stem cell therapy for tissue damage and other diseases.
Collapse
|
5
|
Jung KO, Theruvath AJ, Nejadnik H, Liu A, Xing L, Sulchek T, Daldrup-Link HE, Pratx G. Mechanoporation enables rapid and efficient radiolabeling of stem cells for PET imaging. Sci Rep 2022; 12:2955. [PMID: 35194089 PMCID: PMC8863797 DOI: 10.1038/s41598-022-06938-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/27/2022] [Indexed: 11/09/2022] Open
Abstract
Regenerative medicine uses the patient own stem cells to regenerate damaged tissues. Molecular imaging techniques are commonly used to image the transplanted cells, either right after surgery or at a later time. However, few techniques are fast or straightforward enough to label cells intraoperatively. Adipose tissue-derived stem cells (ADSCs) were harvested from knee joints of minipigs. The cells were labeled with PET contrast agent by flowing mechanoporation using a microfluidic device. While flowing through a series of microchannels, cells are compressed repeatedly by micro-ridges, which open transient pores in their membranes and induce convective transport, intended to facilitate the transport of 68Ga-labeled and lipid-coated mesoporous nanoparticles (MSNs) into the cells. This process enables cells to be labeled in a matter of seconds. Cells labeled with this approach were then implanted into cartilage defects, and the implant was imaged using positron emission tomography (PET) post-surgery. The microfluidic device can efficiently label millions of cells with 68Ga-labeled MSNs in as little as 15 min. The method achieved labeling efficiency greater than 5 Bq/cell on average, comparable to 30 min-long passive co-incubation with 68Ga-MSNs, but with improved biocompatibility due to the reduced exposure to ionizing radiation. Labeling time could also be accelerated by increasing throughput through more parallel channels. Finally, as a proof of concept, ADSCs were labeled with 68Ga-MSNs and quantitatively assessed using clinical PET/MR in a mock transplant operation in pig knee joints. MSN-assisted mechanoporation is a rapid, effective and straightforward approach to label cells with 68Ga. Given its high efficiency, this labeling method can be used to track small cells populations without significant effects on viability. The system is applicable to a variety of cell tracking studies for cancer therapy, regenerative therapy, and immunotherapy.
Collapse
Affiliation(s)
- Kyung Oh Jung
- Division of Medical Physics, Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, 94305, USA.
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Korea.
| | - Ashok Joseph Theruvath
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Hossein Nejadnik
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, Stanford, CA, 94305, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anna Liu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Lei Xing
- Division of Medical Physics, Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, 94305, USA
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, 94305, USA
| | - Todd Sulchek
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Heike E Daldrup-Link
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Guillem Pratx
- Division of Medical Physics, Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
6
|
Sun A, Hayat H, Liu S, Tull E, Bishop JO, Dwan BF, Gudi M, Talebloo N, Dizon JR, Li W, Gaudet J, Alessio A, Aguirre A, Wang P. 3D in vivo Magnetic Particle Imaging of Human Stem Cell-Derived Islet Organoid Transplantation Using a Machine Learning Algorithm. Front Cell Dev Biol 2021; 9:704483. [PMID: 34458264 PMCID: PMC8397508 DOI: 10.3389/fcell.2021.704483] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022] Open
Abstract
Stem cell-derived islet organoids constitute a promising treatment of type 1 diabetes. A major hurdle in the field is the lack of appropriate in vivo method to determine graft outcome. Here, we investigate the feasibility of in vivo tracking of transplanted stem cell-derived islet organoids using magnetic particle imaging (MPI) in a mouse model. Human induced pluripotent stem cells-L1 were differentiated to islet organoids and labeled with superparamagnetic iron oxide nanoparticles. The phantoms comprising of different numbers of labeled islet organoids were imaged using an MPI system. Labeled islet organoids were transplanted into NOD/scid mice under the left kidney capsule and were then scanned using 3D MPI at 1, 7, and 28 days post transplantation. Quantitative assessment of the islet organoids was performed using the K-means++ algorithm analysis of 3D MPI. The left kidney was collected and processed for immunofluorescence staining of C-peptide and dextran. Islet organoids expressed islet cell markers including insulin and glucagon. Image analysis of labeled islet organoids phantoms revealed a direct linear correlation between the iron content and the number of islet organoids. The K-means++ algorithm showed that during the course of the study the signal from labeled islet organoids under the left kidney capsule decreased. Immunofluorescence staining of the kidney sections showed the presence of islet organoid grafts as confirmed by double staining for dextran and C-peptide. This study demonstrates that MPI with machine learning algorithm analysis can monitor islet organoids grafts labeled with super-paramagnetic iron oxide nanoparticles and provide quantitative information of their presence in vivo.
Collapse
Affiliation(s)
- Aixia Sun
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Hasaan Hayat
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Lyman Briggs College, Michigan State University, East Lansing, MI, United States
| | - Sihai Liu
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
- Department of Orthopedics, Beijing Charity Hospital, Capital Medical University, Beijing, China
| | - Eliah Tull
- Medgar Evers College, City University of New York, Brooklyn, NY, United States
| | - Jack Owen Bishop
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Neuroscience, College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Bennett Francis Dwan
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Mithil Gudi
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Lyman Briggs College, Michigan State University, East Lansing, MI, United States
| | - Nazanin Talebloo
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - James Raynard Dizon
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Wen Li
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Jeffery Gaudet
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
- Magnetic Insight Inc., Alameda, CA, United States
| | - Adam Alessio
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
- Department of Computational Mathematics, Science and Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Ping Wang
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
7
|
Rizzo S, Padelli F, Rinaldi E, Gioeni D, Aquino D, Brizzola S, Acocella F, Spaggiari L, Baggi F, Bellomi M, Bruzzone MG, Petrella F. 7-T MRI tracking of mesenchymal stromal cells after lung injection in a rat model. Eur Radiol Exp 2020; 4:54. [PMID: 33029694 PMCID: PMC7541802 DOI: 10.1186/s41747-020-00183-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/04/2020] [Indexed: 01/01/2023] Open
Abstract
Background Mesenchymal stromal cells (MSCs) are able to migrate and engraft at sites of inflammation, injuries, and tumours, but little is known about their fate after local injection. The purpose of this study is to perform MSC tracking, combining in vivo 7-T magnetic resonance imaging (MRI) and histological assessment, following lung injection in a rat model. Methods Five lungs were injected with ferumoxide-labelled MSCs and five with perfluorocarbon-labelled MSCs and underwent 7-T MRI. MRI acquisitions were recorded immediately (T0), at 24 h (T24) and/or 48 h (T48) after injection. For each rat, labelled cells were assessed in the main organs by MRI. Target organs were harvested under sterile conditions from rats sacrificed 0, 24, or 48 h after injection and fixed for histological analysis via confocal and structured illumination microscopy. Results Ferumoxide-labelled MSCs were not detectable in the lungs, whereas they were not visible in the distant sites. Perfluorocarbon-labelled MSCs were seen in 5/5 injected lungs at T0, in 1/2 at T24, and in 1/3 at T48. The fluorine signal in the liver was seen in 3/5 at T0, in 1/2 at T24, and in 2/3 at T48. Post-mortem histology confirmed the presence of MSCs in the injected lung. Conclusions Ferumoxide-labelled cells were not seen at distant sites; a linear decay of injected perfluorocarbon-labelled MSCs was observed at T0, T24, and T48 in the lung. In more than half of the experiments, perfluorocarbon-labelled MSCs scattering to the liver was observed, with a similar decay over time as observed in the lung.
Collapse
Affiliation(s)
- Stefania Rizzo
- Imaging Institute of the Southern Switzerland (IIMSI), Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900, Lugano, Switzerland. .,Facoltà di Scienze biomediche, Università della Svizzera italiana (USI), Via G. Buffi 13, 6904, Lugano, Switzerland. .,Clinica di Radiologia EOC, Istituto di Imaging della Svizzera Italiana (IIMSI), via Tesserete 46, 6900, Lugano, Switzerland.
| | - Francesco Padelli
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Rinaldi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Gioeni
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milan, Italy
| | - Domenico Aquino
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefano Brizzola
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy
| | - Fabio Acocella
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Spaggiari
- Department of Thoracic Surgery, IRCCS European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Fulvio Baggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Massimo Bellomi
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,Department of Radiology, IRCCS European Institute of Oncology, Milan, Italy
| | - Maria Grazia Bruzzone
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesco Petrella
- Department of Thoracic Surgery, IRCCS European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,CRC StaMeTec Università degli studi di Milano, Milan, Italy
| |
Collapse
|
8
|
Nucci MP, Filgueiras IS, Ferreira JM, de Oliveira FA, Nucci LP, Mamani JB, Rego GNA, Gamarra LF. Stem cell homing, tracking and therapeutic efficiency evaluation for stroke treatment using nanoparticles: A systematic review. World J Stem Cells 2020; 12:381-405. [PMID: 32547686 PMCID: PMC7280869 DOI: 10.4252/wjsc.v12.i5.381] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/02/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Stroke is the second leading cause of death worldwide. There is a real need to develop treatment strategies for reducing neurological deficits in stroke survivors, and stem cell (SC) therapeutics appear to be a promising alternative for stroke therapy that can be used in combination with approved thrombolytic or thrombectomy approaches. However, the efficacy of SC therapy depends on the SC homing ability and engraftment into the injury site over a long period of time. Nonetheless, tracking SCs from their niche to the target tissues is a complex process.
AIM To evaluate SC migration homing, tracking and therapeutic efficacy in the treatment of stroke using nanoparticles
METHODS A systematic literature search was performed to identify articles published prior to November 2019 that were indexed in PubMed and Scopus. The following inclusion criteria were used: (1) Studies that used in vivo models of stroke or ischemic brain lesions; (2) Studies of SCs labeled with some type of contrast agent for cell migration detection; and (3) Studies that involved in vivo cellular homing and tracking analysis.
RESULTS A total of 82 articles were identified by indexing in Scopus and PubMed. After the inclusion criteria were applied, 35 studies were selected, and the articles were assessed for eligibility; ultimately, only 25 studies were included. Most of the selected studies used SCs from human and mouse bone marrow labeled with magnetic nanoparticles alone or combined with fluorophore dyes. These cells were administered in the stroke model (to treat middle cerebral artery occlusion in 74% of studies and for photothrombotic induction in 26% of studies). Fifty-three percent of studies used xenogeneic grafts for cell therapy, and the migration homing and tracking evaluation was performed by magnetic resonance imaging as well as other techniques, such as near-infrared fluorescence imaging (12%) or bioluminescence assays (12%).
CONCLUSION Our systematic review provided an up-to-date evaluation of SC migration homing and the efficacy of cellular therapy for stroke treatment in terms of functional and structural improvements in the late stage.
Collapse
Affiliation(s)
- Mariana Penteado Nucci
- LIM44, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05529-060, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Oliveira FA, Nucci MP, Filgueiras IS, Ferreira JM, Nucci LP, Mamani JB, Alvieri F, Souza LEB, Rego GNA, Kondo AT, Hamerschlak N, Gamarra LF. Noninvasive Tracking of Hematopoietic Stem Cells in a Bone Marrow Transplant Model. Cells 2020; 9:cells9040939. [PMID: 32290257 PMCID: PMC7226958 DOI: 10.3390/cells9040939] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/31/2020] [Accepted: 04/03/2020] [Indexed: 12/11/2022] Open
Abstract
The hematopoietic stem cell engraftment depends on adequate cell numbers, their homing, and the subsequent short and long-term engraftment of these cells in the niche. We performed a systematic review of the methods employed to track hematopoietic reconstitution using molecular imaging. We searched articles indexed, published prior to January 2020, in PubMed, Cochrane, and Scopus with the following keyword sequences: (Hematopoietic Stem Cell OR Hematopoietic Progenitor Cell) AND (Tracking OR Homing) AND (Transplantation). Of 2191 articles identified, only 21 articles were included in this review, after screening and eligibility assessment. The cell source was in the majority of bone marrow from mice (43%), followed by the umbilical cord from humans (33%). The labeling agent had the follow distribution between the selected studies: 14% nanoparticle, 29% radioisotope, 19% fluorophore, 19% luciferase, and 19% animal transgenic. The type of graft used in the studies was 57% allogeneic, 38% xenogeneic, and 5% autologous, being the HSC receptor: 57% mice, 9% rat, 19% fish, 5% for dog, porcine and salamander. The imaging technique used in the HSC tracking had the following distribution between studies: Positron emission tomography/single-photon emission computed tomography 29%, bioluminescence 33%, fluorescence 19%, magnetic resonance imaging 14%, and near-infrared fluorescence imaging 5%. The efficiency of the graft was evaluated in 61% of the selected studies, and before one month of implantation, the cell renewal was very low (less than 20%), but after three months, the efficiency was more than 50%, mainly in the allogeneic graft. In conclusion, our review showed an increase in using noninvasive imaging techniques in HSC tracking using the bone marrow transplant model. However, successful transplantation depends on the formation of engraftment, and the functionality of cells after the graft, aspects that are poorly explored and that have high relevance for clinical analysis.
Collapse
Affiliation(s)
- Fernando A. Oliveira
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Mariana P. Nucci
- LIM44—Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil;
| | - Igor S. Filgueiras
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - João M. Ferreira
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Leopoldo P. Nucci
- Centro Universitário do Planalto Central, Brasília DF 72445-020, Brazil;
| | - Javier B. Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Fernando Alvieri
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Lucas E. B. Souza
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto SP 14049-900, Brazil;
| | - Gabriel N. A. Rego
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Andrea T. Kondo
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Nelson Hamerschlak
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
| | - Lionel F. Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.A.O.); (I.S.F.); (J.M.F.); (J.B.M.); (F.A.); (G.N.A.R.); (A.T.K.); (N.H.)
- Correspondence: ; Tel.: +55-11-2151-0243
| |
Collapse
|
10
|
Xie P, Hu X, Li D, Xie S, Zhou Z, Meng X, Shan H. Bioluminescence Imaging of Transplanted Mesenchymal Stem Cells by Overexpression of Hepatocyte Nuclear Factor4α: Tracking Biodistribution and Survival. Mol Imaging Biol 2019; 21:44-53. [PMID: 29761416 DOI: 10.1007/s11307-018-1204-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE The purposes of this study were to construct immortalized human bone marrow mesenchymal stem cells (UE7T-13) with overexpression of the hepatocyte nuclear factor4α (hHNF4α) and luciferase2-mKate2 dual-fusion reporter gene, further investigate their impact on treating acute liver injury (ALI) in rats, and track their biodistribution and survival by bioluminescence imaging (BLI). PROCEDURES The hHNF4α and luciferase2-mKate2 genes were transduced by a lentiviral vector into UE7T-13 cells (named E7-hHNF4α-R cells), and expression was verified by immunofluorescence, RT-PCR, and flow cytometry. E7-hGFP-R cells expressing the luciferase2-mKate2/hGFP gene served as a negative group. A correlation between the bioluminescence signal and cell number was detected by BLI. The ALI rats were established and divided into three groups: PBS, E7-hGFP-R, and E7-hHNF4α-R. After transplantation of 2.0 × 106 cells, BLI was used to dynamically track their biodistribution and survival. The restoration of biological functions was assessed by serum biochemical and histological analyses. RESULTS Stable high-level expression of hHNF4α and mKate2 protein was established in the E7-hHNF4α-R cells in vitro. The E7-hHNF4α-R cells strongly expressed hGFP, hHNF4α, and mKate2 proteins, and the hHNF4α gene. hGFP-mKate2 dual-positive cell expression reached approximately 93 %. BLI verified that a linear relationship existed between the cell number and bioluminescence signal (R2 = 0.9991). The cells improved liver function in vivo after transplantation into the ALI rat liver, as evidenced by the fact that AST and ALT temporarily returned to normal levels in the recipient ALI rats. The presence of the transplanted E7-hGFP-R and E7-hHNF4α-R cells in recipient rat livers was confirmed by BLI and immunohistochemistry. However, the cells were cleared by the immune system a short time after transplantation into ALI rats with a normal immune system. CONCLUSION Our data revealed that the E7-hHNF4α-R cells can transiently improve damaged liver function and were rapidly cleared by the immune system. In addition, BLI is a useful tool to track transplanted cell biodistribution and survival.
Collapse
Affiliation(s)
- Peiyi Xie
- Guang Dong Provincial Engineering Research Center of Molecular Imaging, Zhuhai, China.,Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojun Hu
- Guang Dong Provincial Engineering Research Center of Molecular Imaging, Zhuhai, China.,Interventional Medicine Department, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Interventional Radiology Institute, Sun Yat-sen University, Zhuhai, China
| | - Dan Li
- Guang Dong Provincial Engineering Research Center of Molecular Imaging, Zhuhai, China.,Interventional Medicine Department, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Interventional Radiology Institute, Sun Yat-sen University, Zhuhai, China
| | - Sidong Xie
- The Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiyang Zhou
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Hong Shan
- Guang Dong Provincial Engineering Research Center of Molecular Imaging, Zhuhai, China. .,Interventional Medicine Department, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China. .,Interventional Radiology Institute, Sun Yat-sen University, Zhuhai, China.
| |
Collapse
|
11
|
Rizzo S, Petrella F, Zucca I, Rinaldi E, Barbaglia A, Padelli F, Baggi F, Spaggiari L, Bellomi M, Bruzzone MG. In vitro labelling and detection of mesenchymal stromal cells: a comparison between magnetic resonance imaging of iron-labelled cells and magnetic resonance spectroscopy of fluorine-labelled cells. Eur Radiol Exp 2017; 1:6. [PMID: 29708157 PMCID: PMC5909334 DOI: 10.1186/s41747-017-0010-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/31/2017] [Indexed: 12/31/2022] Open
Abstract
Background Among the various stem cell populations used for cell therapy, adult mesenchymal stromal cells (MSCs) have emerged as a major new cell technology. These cells must be tracked after transplantation to monitor their migration within the body and quantify their accumulation at the target site. This study assessed whether rat bone marrow MSCs can be labelled with superparamagnetic iron oxide (SPIO) nanoparticles and perfluorocarbon (PFC) nanoemulsion formulations without altering cell viability and compared magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) results from iron-labelled and fluorine-labelled MSCs, respectively. Methods Of MSCs, 2 × 106 were labelled with Molday ION Rhodamine-B (MIRB) and 2 × 106 were labelled with Cell Sense. Cell viability was evaluated by trypan blue exclusion method. Labelled MSCs were divided into four samples containing increasing cell numbers (0.125 × 106, 0.25 × 106, 0.5 × 106, 1 × 106) and scanned on a 7T MRI: for MIRB-labelled cells, phantoms and cells negative control, T1, T2 and T2* maps were acquired; for Cell Sense labelled cells, phantoms and unlabelled cells, a 19F non-localised single-pulse MRS sequence was acquired. Results In total, 86.8% and 83.6% of MIRB-labelled cells and Cell Sense-labelled cells were viable, respectively. MIRB-labelled cells were visible in all samples with different cell numbers; pellets containing 0.5 × 106 and 1 × 106 of Cell Sense-labelled cells showed a detectable 19F signal. Conclusions Our data support the use of both types of contrast material (SPIO and PFC) for MSCs labelling, although further efforts should be dedicated to improve the efficiency of PFC labelling.
Collapse
Affiliation(s)
- Stefania Rizzo
- 1Department of Radiology, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Francesco Petrella
- 2Department of Thoracic Surgery, European Institute of Oncology, Milan, Italy.,5Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, via Festa del Perdono 7, 20142 Milan, Italy
| | - Ileana Zucca
- Scientific Department, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Elena Rinaldi
- Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Andrea Barbaglia
- Scientific Department, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Francesco Padelli
- Scientific Department, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Fulvio Baggi
- Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Lorenzo Spaggiari
- 2Department of Thoracic Surgery, European Institute of Oncology, Milan, Italy.,5Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, via Festa del Perdono 7, 20142 Milan, Italy
| | - Massimo Bellomi
- 1Department of Radiology, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy.,5Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, via Festa del Perdono 7, 20142 Milan, Italy
| | - Maria Grazia Bruzzone
- Department of Neuroradiology, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| |
Collapse
|