1
|
Zlotnick HM, Stevens MM, Mauck RL. Physical-property-based patterning: simply engineering complex tissues. Trends Biotechnol 2024; 42:1230-1240. [PMID: 38664141 PMCID: PMC11449661 DOI: 10.1016/j.tibtech.2024.03.004] [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: 11/27/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 08/09/2024]
Abstract
The field of biofabrication is rapidly expanding with the advent of new technologies and material systems to engineer complex tissues. In this opinion article, we introduce an emerging tissue patterning method, physical-property-based patterning, that has strong translational potential given its simplicity and limited dependence on external hardware. Physical-property-based patterning relies solely on the intrinsic density, magnetic susceptibility, or compressibility of an object, its surrounding solution, and the noncontact application of a remote field. We discuss how physical properties can be exploited to pattern objects and design a variety of biologic tissues. Finally, we pose several open questions that, if addressed, could transform the status quo of biofabrication, pushing us one step closer to patterning tissues in situ.
Collapse
Affiliation(s)
- Hannah M Zlotnick
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College London, London, UK; Department of Physiology, Anatomy and Genetics, Department of Engineering Science, and Kavli Institute for Nanoscience Discovery, University of Oxford, UK
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Translational Musculoskeletal Research Center, CMC VA Medical Center, Philadelphia, PA, USA.
| |
Collapse
|
2
|
He JL, You YX, Pei X, Jiang W, Zeng QM, Chen B, Wang YH, Chen EQ, Tang H, Gao XF, Wu DB. Tracking of Stem Cells in Chronic Liver Diseases: Current Trends and Developments. Stem Cell Rev Rep 2024; 20:447-454. [PMID: 37993759 DOI: 10.1007/s12015-023-10659-2] [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] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
Stem cell therapy holds great promise for future clinical practice for treatment of advanced liver diseases. However, the fate of stem cells after transplantation, including the distribution, viability, and the cell clearance, has not been fully elucidated. Herein, recent advances regarding the imaging tools for stem cells tracking mainly in chronic liver diseases with the advantages and disadvantages of each approach have been described. Magnetic resonance imaging is a promising clinical imaging modality due to non-radioactivity, excellent penetrability, and high spatial resolution. Fluorescence imaging and radionuclide imaging demonstrate relatively increased sensitivity, with the latter excelling in real-time monitoring. Reporter genes specialize in long-term tracing. Nevertheless, the disadvantages of low sensitivity, radiation, exogenous gene risk are inevitably present in each of these means, respectively. In this review, we aim to comprehensively evaluate the current state of methods for tracking of stem cell, highlighting their strengths and weaknesses, and providing insights into their future potential. Multimodality imaging strategies may overcome the inherent limitations of single-modality imaging by combining the strengths of different imaging techniques to provide more comprehensive information in the clinical setting.
Collapse
Affiliation(s)
- Jin-Long He
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610065, China
| | - Yi-Xian You
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiong Pei
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing-Min Zeng
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bin Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yong-Hong Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - En-Qiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiu-Feng Gao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610065, China.
| | - Dong-Bo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Xue J, Gurav N, Elsharkawy S, Deb S. Hydrogel Composite Magnetic Scaffolds: Toward Cell-Free In Situ Bone Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:168-181. [PMID: 38109842 PMCID: PMC10792668 DOI: 10.1021/acsabm.3c00732] [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: 08/31/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/20/2023]
Abstract
Reconstruction of critical sized bone defects in the oral and maxillofacial region continues to be clinically challenging despite the significant development of osteo-regenerative materials. Among 3D biomaterials, hydrogels and hydrogel composites have been explored for bone regeneration, however, their inferior clinical performance in comparison to autografts is mainly attributed to variable rates of degradation and lack of vascularization. In this study, we report hydrogel composite magnetic scaffolds formed from calcium carbonate, poly(vinyl alcohol) (PVA), and magnetic nanoparticles (MNPs), using PVA as matrix and calcium carbonate particles in vaterite phase as filler, to enhance the cross-linking of matrix and porosity with MNPs that can target and regulate cell signaling pathways to control cell behavior and improve the osteogenic and angiogenic potential. The physical and mechanical properties were evaluated, and cytocompatibility was investigated by culturing human osteoblast-like cells onto the scaffolds. The vaterite phase due to its higher solubility in comparison to calcium phosphates, combined with the freezing-thawing process of PVA, yielded porous scaffolds that exhibited adequate thermal stability, favorable water-absorbing capacity, excellent mineralization ability, and cytocompatibility. An increasing concentration from 1, 3, and 6 wt % MNPs in the scaffolds showed a statistically significant increase in compressive strength and modulus of the dry specimens that exhibited brittle fracture. However, the hydrated specimens were compressible and showed a slight decrease in compressive strength with 6% MNPs, although this value was higher compared to that of the scaffolds with no MNPs.
Collapse
Affiliation(s)
- Jingyi Xue
- Faculty of Dentistry, Oral
and Craniofacial Sciences, King’s
College London, London SE1 9RT, United
Kingdom
| | - Neelam Gurav
- Faculty of Dentistry, Oral
and Craniofacial Sciences, King’s
College London, London SE1 9RT, United
Kingdom
| | - Sherif Elsharkawy
- Faculty of Dentistry, Oral
and Craniofacial Sciences, King’s
College London, London SE1 9RT, United
Kingdom
| | - Sanjukta Deb
- Faculty of Dentistry, Oral
and Craniofacial Sciences, King’s
College London, London SE1 9RT, United
Kingdom
| |
Collapse
|
4
|
Merino JJ, Cabaña-Muñoz ME. Nanoparticles and Mesenchymal Stem Cell (MSC) Therapy for Cancer Treatment: Focus on Nanocarriers and a si-RNA CXCR4 Chemokine Blocker as Strategies for Tumor Eradication In Vitro and In Vivo. MICROMACHINES 2023; 14:2068. [PMID: 38004925 PMCID: PMC10673568 DOI: 10.3390/mi14112068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/13/2023] [Indexed: 11/26/2023]
Abstract
Mesenchymal stem cells (MSCs) have a high tropism for the hypoxic microenvironment of tumors. The combination of nanoparticles in MSCs decreases tumor growth in vitro as well as in rodent models of cancers in vivo. Covalent conjugation of nanoparticles with the surface of MSCs can significantly increase the drug load delivery in tumor sites. Nanoparticle-based anti-angiogenic systems (gold, silica and silicates, diamond, silver, and copper) prevented tumor growth in vitro. For example, glycolic acid polyconjugates enhance nanoparticle drug delivery and have been reported in human MSCs. Labeling with fluorescent particles (coumarin-6 dye) identified tumor cells using fluorescence emission in tissues; the conjugation of different types of nanoparticles in MSCs ensured success and feasibility by tracking the migration and its intratumor detection using non-invasive imaging techniques. However, the biosafety and efficacy; long-term stability of nanoparticles, and the capacity for drug release must be improved for clinical implementation. In fact, MSCs are vehicles for drug delivery with nanoparticles and also show low toxicity but inefficient accumulation in tumor sites by clearance of reticuloendothelial organs. To solve these problems, the internalization or conjugation of drug-loaded nanoparticles should be improved in MSCs. Finally, CXCR4 may prove to be a promising target for immunotherapy and cancer treatment since the delivery of siRNA to knock down this alpha chemokine receptor or CXCR4 antagonism has been shown to disrupt tumor-stromal interactions.
Collapse
Affiliation(s)
- José Joaquín Merino
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (U.C.M.), 28040 Madrid, Spain
| | | |
Collapse
|
5
|
Shams F, Pourjabbar B, Hashemi N, Farahmandian N, Golchin A, Nuoroozi G, Rahimpour A. Current progress in engineered and nano-engineered mesenchymal stem cells for cancer: From mechanisms to therapy. Biomed Pharmacother 2023; 167:115505. [PMID: 37716113 DOI: 10.1016/j.biopha.2023.115505] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
Mesenchymal stem cells (MSCs), as self-renewing multipotent stromal cells, have been considered promising agents for cancer treatment. A large number of studies have demonstrated the valuable properties of MSC-based treatment, such as low immunogenicity and intrinsic tumor-trophic migratory properties. To enhance the potency of MSCs for therapeutic purposes, equipping MSCs with targeted delivery functions using genetic engineering is highly beneficial. Genetically engineered MSCs can express tumor suppressor agents such as pro-apoptotic, anti-proliferative, anti-angiogenic factors and act as ideal delivery vehicles. MSCs can also be loaded with nanoparticle drugs for increased efficacy and externally moderated targeting. Moreover, exosomes secreted by MSCs have important physiological properties, so they can contribute to intercellular communication and transfer cargo into targeted tumor cells. The precise role of genetically modified MSCs in tumor environments is still up for debate, but the beginning of clinical trials has been confirmed by promising results from preclinical investigations of MSC-based gene therapy for a wide range of malignancies. This review highlights the advanced techniques of engineering/nano-engineering and MSC-derived exosomes in tumor-targeted therapy.
Collapse
Affiliation(s)
- Forough Shams
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, 1968917313 Tehran, Iran
| | - Bahareh Pourjabbar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, 1968917313 Tehran, Iran
| | - Navid Farahmandian
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Golchin
- Cellular & Molecular Research Center, Cellular & Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57157993313, Iran; Department of Clinical Biochemistry & Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia 57157993313, Islamic Republic of Iran
| | - Ghader Nuoroozi
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Rahimpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Chen Y, Hou S. Recent progress in the effect of magnetic iron oxide nanoparticles on cells and extracellular vesicles. Cell Death Discov 2023; 9:195. [PMID: 37380637 DOI: 10.1038/s41420-023-01490-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/30/2023] Open
Abstract
At present, iron oxide nanoparticles (IONPs) are widely used in the biomedical field. They have unique advantages in targeted drug delivery, imaging and disease treatment. However, there are many things to pay attention to. In this paper, we reviewed the fate of IONPs in different cells and the influence on the production, separation, delivery and treatment of extracellular vesicles. It aims to provide cutting-edge knowledge related to iron oxide nanoparticles. Only by ensuring the safety and effectiveness of IONPs can their application in biomedical research and clinic be further improved.
Collapse
Affiliation(s)
- Yuling Chen
- Institute of Disaster and Emergency Medicine, Tianjin University, 300072, Tianjin, China.
- Key Laboratory for Disaster Medicine Technology, 300072, Tianjin, China.
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, 300072, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, 300072, Tianjin, China
| |
Collapse
|
7
|
Cumpata AJ, Peptanariu D, Lungoci AL, Labusca L, Pinteala M, Radulescu L. Towards Regenerative Audiology: Immune Modulation of Adipose-Derived Mesenchymal Cells Preconditioned with Citric Acid-Coated Antioxidant-Functionalized Magnetic Nanoparticles. Medicina (B Aires) 2023; 59:587. [DOI: https:/doi.org/10.3390/medicina59030587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023] Open
Abstract
Introduction and Background: Based on stem cells, bioactive molecules and supportive structures, regenerative medicine (RM) is promising for its potential impact on field of hearing loss by offering innovative solutions for hair cell rescue. Nanotechnology has recently been regarded as a powerful tool for accelerating the efficiency of RM therapeutic solutions. Adipose-derived mesenchymal cells (ADSCs) have already been tested in clinical trials for their regenerative and immunomodulatory potential in various medical fields; however, the advancement to bedside treatment has proven to be tedious. Innovative solutions are expected to circumvent regulatory and manufacturing issues related to living cell-based therapies. The objectives of the study were to test if human primary ADSCs preconditioned with magnetic nanoparticles coated with citric acid and functionalized with antioxidant protocatechuic acid (MNP-CA-PCA) retain their phenotypic features and if conditioned media elicit immune responses in vitro. MNP-CA-PCA was synthesized and characterized regarding size, colloidal stability as well as antioxidant release profile. Human primary ADSCs preconditioned with MNP-CA-PCA were tested for viability, surface marker expression and mesenchymal lineage differentiation potential. Conditioned media (CM) from ADSCs treated with MNP-CA-PCA were tested for Il-6 and IL-8 cytokine release using ELISA and inhibition of lectin-stimulated peripheral blood monocyte proliferation. Results: MNP-CA-PCA-preconditioned ADSCs display good viability and retain their specific mesenchymal stem cell phenotype. CM from ADSCs conditioned with MNP-CA-PCA do not display increased inflammatory cytokine release and do not induce proliferation of allergen-stimulated allogeneic peripheral blood monocytes in vitro. Conclusions: While further in vitro and in vivo tests are needed to validate these findings, the present results indicated that CM from ADSCs preconditioned with MNP-CA-PCA could be developed as possible cell-free therapies for rescuing auditory hair cells.
Collapse
Affiliation(s)
- Adeline Josephine Cumpata
- Doctoral School, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | - Dragos Peptanariu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers ‘‘Petru Poni’’, Institute of Macromolecular Chemistry Aleea Grigore Ghica, Voda 41A, 700487 Iasi, Romania
| | - Ana-Lacramioara Lungoci
- Centre of Advanced Research in Bionanoconjugates and Biopolymers ‘‘Petru Poni’’, Institute of Macromolecular Chemistry Aleea Grigore Ghica, Voda 41A, 700487 Iasi, Romania
| | - Luminita Labusca
- Orthopedics and Traumatology Clinic, Emergency Hospital Saint Spiridon, 1 St Independentei Boulevard, 700111 Iasi, Romania
- National Institute of Research and Development in Technical Physics Iasi Romania, 700111 Iasi, Romania
| | - Mariana Pinteala
- Centre of Advanced Research in Bionanoconjugates and Biopolymers ‘‘Petru Poni’’, Institute of Macromolecular Chemistry Aleea Grigore Ghica, Voda 41A, 700487 Iasi, Romania
| | - Luminita Radulescu
- Doctoral School, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
- ENT Clinic Department, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| |
Collapse
|
8
|
Cumpata AJ, Peptanariu D, Lungoci AL, Labusca L, Pinteala M, Radulescu L. Towards Regenerative Audiology: Immune Modulation of Adipose-Derived Mesenchymal Cells Preconditioned with Citric Acid-Coated Antioxidant-Functionalized Magnetic Nanoparticles. Medicina (B Aires) 2023; 59:medicina59030587. [PMID: 36984588 PMCID: PMC10058393 DOI: 10.3390/medicina59030587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Introduction and Background: Based on stem cells, bioactive molecules and supportive structures, regenerative medicine (RM) is promising for its potential impact on field of hearing loss by offering innovative solutions for hair cell rescue. Nanotechnology has recently been regarded as a powerful tool for accelerating the efficiency of RM therapeutic solutions. Adipose-derived mesenchymal cells (ADSCs) have already been tested in clinical trials for their regenerative and immunomodulatory potential in various medical fields; however, the advancement to bedside treatment has proven to be tedious. Innovative solutions are expected to circumvent regulatory and manufacturing issues related to living cell-based therapies. The objectives of the study were to test if human primary ADSCs preconditioned with magnetic nanoparticles coated with citric acid and functionalized with antioxidant protocatechuic acid (MNP-CA-PCA) retain their phenotypic features and if conditioned media elicit immune responses in vitro. MNP-CA-PCA was synthesized and characterized regarding size, colloidal stability as well as antioxidant release profile. Human primary ADSCs preconditioned with MNP-CA-PCA were tested for viability, surface marker expression and mesenchymal lineage differentiation potential. Conditioned media (CM) from ADSCs treated with MNP-CA-PCA were tested for Il-6 and IL-8 cytokine release using ELISA and inhibition of lectin-stimulated peripheral blood monocyte proliferation. Results: MNP-CA-PCA-preconditioned ADSCs display good viability and retain their specific mesenchymal stem cell phenotype. CM from ADSCs conditioned with MNP-CA-PCA do not display increased inflammatory cytokine release and do not induce proliferation of allergen-stimulated allogeneic peripheral blood monocytes in vitro. Conclusions: While further in vitro and in vivo tests are needed to validate these findings, the present results indicated that CM from ADSCs preconditioned with MNP-CA-PCA could be developed as possible cell-free therapies for rescuing auditory hair cells.
Collapse
Affiliation(s)
- Adeline Josephine Cumpata
- Doctoral School, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania; (A.J.C.); (L.R.)
| | - Dragos Peptanariu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers ‘‘Petru Poni’’, Institute of Macromolecular Chemistry Aleea Grigore Ghica, Voda 41A, 700487 Iasi, Romania; (D.P.)
| | - Ana-Lacramioara Lungoci
- Centre of Advanced Research in Bionanoconjugates and Biopolymers ‘‘Petru Poni’’, Institute of Macromolecular Chemistry Aleea Grigore Ghica, Voda 41A, 700487 Iasi, Romania; (D.P.)
| | - Luminita Labusca
- Orthopedics and Traumatology Clinic, Emergency Hospital Saint Spiridon, 1 St Independentei Boulevard, 700111 Iasi, Romania
- National Institute of Research and Development in Technical Physics Iasi Romania, 700111 Iasi, Romania
- Correspondence:
| | - Mariana Pinteala
- Centre of Advanced Research in Bionanoconjugates and Biopolymers ‘‘Petru Poni’’, Institute of Macromolecular Chemistry Aleea Grigore Ghica, Voda 41A, 700487 Iasi, Romania; (D.P.)
| | - Luminita Radulescu
- Doctoral School, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania; (A.J.C.); (L.R.)
- ENT Clinic Department, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| |
Collapse
|
9
|
Bulte JWM, Wang C, Shakeri-Zadeh A. In Vivo Cellular Magnetic Imaging: Labeled vs. Unlabeled Cells. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2207626. [PMID: 36589903 PMCID: PMC9798832 DOI: 10.1002/adfm.202207626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Indexed: 06/17/2023]
Abstract
Superparamagnetic iron oxide (SPIO)-labeling of cells has been applied for magnetic resonance imaging (MRI) cell tracking for over 30 years, having resulted in a dozen or so clinical trials. SPIO nanoparticles are biodegradable and can be broken down into elemental iron, and hence the tolerance of cells to magnetic labeling has been overall high. Over the years, however, single reports have accumulated demonstrating that the proliferation, migration, adhesion and differentiation of magnetically labeled cells may differ from unlabeled cells, with inhibition of chondrocytic differentiation of labeled human mesenchymal stem cells (hMSCs) as a notable example. This historical perspective provides an overview of some of the drawbacks that can be encountered with magnetic labeling. Now that magnetic particle imaging (MPI) cell tracking is emerging as a new in vivo cellular imaging modality, there has been a renaissance in the formulation of SPIO nanoparticles this time optimized for MPI. Lessons learned from the occasional past pitfalls encountered with SPIO-labeling of cells for MRI may expedite possible future clinical translation of (combined) MRI/MPI cell tracking.
Collapse
Affiliation(s)
- Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chao Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ali Shakeri-Zadeh
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
10
|
Kim JH, Bak SH, Yang HJ, Doo SW, Kim DK, Yang WJ, Kim SU, Lee HJ, Song YS. Improvement of erectile dysfunction using endothelial progenitor cells from fetal cerebral vasculature in the cavernous nerve injury of rats. Basic Clin Androl 2022; 32:21. [PMID: 36451096 PMCID: PMC9714194 DOI: 10.1186/s12610-022-00171-x] [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: 07/09/2022] [Accepted: 09/05/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Because of limited differentiation to endothelium from mesenchymal stem cells, it has been strongly recommended to use endothelial progenitor cells for the regeneration of the damaged endothelium of corpora cavernosa. This study was performed to investigate the immortalized human cerebral endothelial cells and their capability for repairing erectile dysfunction in a rat model of cavernous nerve injury. Circulating endothelial progenitor cells were isolated from human fetal brain vasculature at the periventricular region of telencephalic tissues. Over 95% of CD 31-positive cells were sorted and cultured for 10 days. Human cerebral endothelial progenitor cells were injected into the cavernosa of rats with cavernous nerve injury. Erectile response was then assessed. In in vivo assays, rats were divided into three groups: group 1, sham operation: group 2, bilateral cavernous nerve injury: and group 3, treatment with human cerebral endothelial cells after cavernous nerve injury. RESULTS Established immortalized circulating endothelial progenitor cells showed expression of human telomerase reverse transcriptase transcript by RT-PCR. They also showed the expression of vascular endothelial growth factor, von Willebrand factor, vascular endothelial growth factor receptor, and CD31, cell type-specific markers for endothelial cells by RT-PCR. In in vitro angiogenesis assays, they demonstrated tube formation that suggested morphological properties of endothelial progenitor cells. In in vivo assays, impaired erectile function of rat with cavernous nerve injury recovered at 2, 4, and 12 weeks after transplantation of human cerebral endothelial cells into the cavernosa. CONCLUSIONS Telomerase reverse transcriptase-circulating endothelial progenitor cells from fetal brain vasculature could repair erectile dysfunction of rats with cavernous nerve injury.
Collapse
Affiliation(s)
- Jae Heon Kim
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| | - Sang Hong Bak
- Research Institute, e-Biogen Inc., Seoul, Republic of Korea
| | - Hee Jo Yang
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, Cheonan, Republic of Korea
| | - Seung Whan Doo
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| | - Do Kyung Kim
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| | - Won Jae Yang
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| | - Seung U. Kim
- grid.416957.80000 0004 0633 8774Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
| | - Hong J. Lee
- Research Institute, e-Biogen Inc., Seoul, Republic of Korea ,grid.254229.a0000 0000 9611 0917Medical Research Institute, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| | - Yun Seob Song
- grid.412674.20000 0004 1773 6524Department of Urology, Soonchunhyang University School of Medicine, 04401 Seoul, Republic of Korea
| |
Collapse
|
11
|
von Pueckler K, John K, Kramer M, Bokemeyer J, Arnhold S. MRI Tracking of Iron Oxide Labelled Canine Mesenchymal Stem Cells in Artificial Stifle Defects. Vet Comp Orthop Traumatol 2022; 35:362-369. [PMID: 35790198 DOI: 10.1055/s-0042-1750432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES The aim of this study was to describe ultrasmall superparamagnetic iron oxides labelling of canine adipose-derived mesenchymal stem cells (AdMSCs) and the detection and semiquantitative evaluation of the labelled cells after implantation in artificial canine stifle defects using magnetic resonance imaging. METHODS Magnetic resonance imaging examinations of 10 paired (n = 20) cadaveric stifle joints were evaluated after creation of chondral defects and embedding of ultrasmall superparamagnetic iron oxides labelled canine mesenchymal stem cells. To prove the feasibility of the labelling for in vivo usage, Prussian blue staining, cell vitality tests and intralesional administration of labelled cells were conducted. Magnetic resonance imaging of ex vivo defects filled with different cell concentrations was obtained to depict the cell content semiquantitatively via signal intensity measurements (region of interest). RESULTS Prussian blue staining showed that the labelling was effective. According to the vitality tests, it had no significant short-term influence on cell viability and proliferation rate. For the evaluation of the defect T2* sequences were feasible and stifle defects were visible allowing measurements of the signal intensity in all cases. Increasing the cell concentration within the chondral defects resulted in an inversely proportional, significant reduction of signal intensity according to the region of interest. CLINICAL SIGNIFICANCE Ultrasmall superparamagnetic iron oxides labelling was effective. The detection of the AdMSCs in a complex anatomical structure like the surface of the femoral condyle was possible and the T2* signal intensity of the implant region was significantly correlated with the concentration of the AdMSCs.
Collapse
Affiliation(s)
- Kerstin von Pueckler
- Department of Veterinary Clinical Science, Small Animal Clinic, Justus Liebig University, Gießen, Germany
| | - Karen John
- Department of Veterinary Clinical Science, Small Animal Clinic, Justus Liebig University, Gießen, Germany
| | - Martin Kramer
- Department of Veterinary Clinical Science, Small Animal Clinic, Justus Liebig University, Gießen, Germany
| | - Jan Bokemeyer
- Tierklinik Kalbach - Fachklinik für Kleintiere Frankfurt, Frankfurt am Main, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy and Embryology, Justus Liebig University, Gießen, Germany
| |
Collapse
|
12
|
Iravani K, Mehrabani D, Doostkam A, Azarpira N, Iranpour P, Bahador M, Mehravar S. Use of MRI to Assess the Regenerative Effects of Adipose Tissue Derived Mesenchymal Stem Cells in a Rabbit Cartilaginous Laryngeal Defect Model. Curr Ther Res Clin Exp 2022; 97:100682. [PMID: 35959231 PMCID: PMC9361331 DOI: 10.1016/j.curtheres.2022.100682] [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: 11/24/2021] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
Background Stenosis and scar formation after repair of laryngeal tissue defects are serious problems that can significantly influence a patient's quality of life. Objective In this study, we evaluated the use of magnetic resonance imaging to assess the efficacy of adipose tissue-derived mesenchymal stem cells (ASCs) on cartilaginous regeneration in an experimental rabbit model. Methods Ten male white Dutch rabbits each had a 5 mm cartilaginous defect created surgically in the right and left thyroid lamina. On the right side, ASCs labeled with iron oxide particles were infused. As a control, the left side was left untreated. Repair of the defects were then evaluated by direct observation, histological evaluation, and magnetic resonance imaging monitoring done on days 1, 7, 14, and 28. Results Histological examination revealed that compared with control, transplanted ASCs significantly increased cartilage regeneration (P ˂ 0.001), reduced inflammation (P ˂ 0.001), and fibrosis (P = 0.050). Magnetic resonance imaging tracking showed accurate placement and viability of the infused ASCs, as evidenced by low signal intensity onT2 weighted images at the level of the right thyroid cartilage. Conclusions Infusion of ASCs improved laryngeal regeneration of surgically induced cartilaginous defects while decreasing fibrous tissue formation in this in vivo rabbit model. Furthermore, magnetic resonance imaging was shown to be a useful, noninvasive method to track correct ASCs placement and viability in cartilage regeneration in this animal model.
Collapse
Affiliation(s)
- Kamyar Iravani
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
- Address correspondence to: Kamyar Iravani, MD, Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Khalili Hospital, Khalili St, Shiraz, Iran, Postal code: 71936-16641.
| | - Davood Mehrabani
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Doostkam
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz Institute of Stem Cell and Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooya Iranpour
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Bahador
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Mehravar
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Andersen C, Uvebrant K, Mori Y, Aarsvold S, Jacobsen S, Berg LC, Lundgren-Åkerlund E, Lindegaard C. Human integrin α10β1-selected mesenchymal stem cells home to cartilage defects in the rabbit knee and assume a chondrocyte-like phenotype. Stem Cell Res Ther 2022; 13:206. [PMID: 35578319 PMCID: PMC9109317 DOI: 10.1186/s13287-022-02884-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have shown promising results in stimulating cartilage repair and in the treatment of osteoarthritis (OA). However, the fate of the MSCs after intra-articular injection and their role in cartilage regeneration is not clear. To address these questions, this study investigated (1) homing of labeled human adipose tissue derived integrin α10β1-selected MSCs (integrin α10-MSCs) to a cartilage defect in a rabbit model and (2) the ability of the integrin α10-MSCs to differentiate to chondrocytes and to produce cartilage matrix molecules in vivo. DESIGN Integrin α10-MSCs were labeled with superparamagnetic iron oxide nanoparticles (SPIONs) co-conjugated with Rhodamine B to allow visualization by both MRI and fluorescence microscopy. A cartilage defect was created in the articular cartilage of the intertrochlear groove of the femur of rabbits. Seven days post-surgery, labeled integrin α10-MSCs or vehicle were injected into the joint. Migration and distribution of the SPION-labeled integrin α10-MSCs was evaluated by high-field 9.4 T MRI up to 10 days after injection. Tissue sections from the repair tissue in the defects were examined by fluorescence microscopy. RESULTS In vitro characterization of the labeled integrin α10-MSCs demonstrated maintained viability, proliferation rate and trilineage differentiation capacity compared to unlabeled MSCs. In vivo MRI analysis detected the labeled integrin α10-MSCs in the cartilage defects at all time points from 12 h after injection until day 10 with a peak concentration between day 1 and 4 after injection. The labeled MSCs were also detected lining the synovial membrane at the early time points. Fluorescence analysis confirmed the presence of the labeled integrin α10-MSCs in all layers of the cartilage repair tissue and showed co-localization between the labeled cells and the specific cartilage molecules aggrecan and collagen type II indicating in vivo differentiation of the MSCs to chondrocyte-like cells. No adverse effects of the α10-MSC treatment were detected during the study period. CONCLUSION Our results demonstrated migration and homing of human integrin α10β1-selected MSCs to cartilage defects in the rabbit knee after intra-articular administration as well as chondrogenic differentiation of the MSCs in the regenerated cartilage tissue.
Collapse
Affiliation(s)
- Camilla Andersen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark.
| | | | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | | | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| | - Lise Charlotte Berg
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| | | | - Casper Lindegaard
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| |
Collapse
|
14
|
In vivo tracking of unlabelled mesenchymal stromal cells by mannose-weighted chemical exchange saturation transfer MRI. Nat Biomed Eng 2022; 6:658-666. [PMID: 35132228 PMCID: PMC9425291 DOI: 10.1038/s41551-021-00822-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 08/05/2021] [Indexed: 12/13/2022]
Abstract
The tracking of the in vivo biodistribution of transplanted human mesenchymal stromal cells (hMSCs) relies on reporter genes or on the addition of exogenous imaging agents. However, reporter genes and exogenous labels may require bespoke manufacturing and regulatory processes if used in cell therapies, and the labels may alter the cells' properties and are diluted on cellular division. Here we show that high-mannose N-linked glycans, which are abundantly expressed on the surface of hMSCs, can serve as a biomarker for the label-free tracking of transplanted hMSCs by mannose-weighted chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI). For live mice with luciferase-transfected hMSCs transplanted into their brains, post-mortem fluorescence staining with a mannose-specific lectin showed that increases in the CEST MRI signal, which correlated well with the bioluminescence intensity of viable hMSCs for 14 days, corresponded to the presence of mannose. In vitro, osteogenically differentiated hMSCs led to lower CEST MRI signal intensities owing to the concomitantly reduced expression of mannose. The label-free imaging of hMSCs may facilitate the development and testing of cell therapies.
Collapse
|
15
|
Abstract
Mesenchymal stem cells (MSCs) exhibit regenerative and reparative properties. However, most MSC-related studies remain to be translated for regular clinical usage, partly due to challenges in pre-transplantation cell labelling and post-transplantation cell tracking. Amidst this, there are growing concerns over the toxicity of commonly used gadolinium-based contrast agents that mediate in-vivo cell detection via MRI. This urges to search for equally effective but less toxic alternatives that would facilitate and enhance MSC detection post-administration and provide therapeutic benefits in-vivo. MSCs labelled with iron oxide nanoparticles (IONPs) have shown promising results in-vitro and in-vivo. Thus, it would be useful to revisit these studies before inventing new labelling approaches. Aiming to inform regenerative medicine and augment clinical applications of IONP-labelled MSCs, this review collates and critically evaluates the utility of IONPs in enhancing MSC detection and therapeutics. It explains the rationale, principle, and advantages of labelling MSCs with IONPs, and describes IONP-induced intracellular alterations and consequent cellular manifestations. By exemplifying clinical pathologies, it examines contextual in-vitro, animal, and clinical studies that used IONP-labelled bone marrow-, umbilical cord-, adipose tissue- and dental pulp-derived MSCs. It compiles and discusses studies involving MSC-labelling of IONPs in combinations with carbohydrates (Venofer, ferumoxytol, dextran, glucosamine), non-carbohydrate polymers [poly(L-lysine), poly(lactide-co-glycolide), poly(L-lactide), polydopamine], elements (ruthenium, selenium, gold, zinc), compounds/stains (silica, polyethylene glycol, fluorophore, rhodamine B, DAPI, Prussian blue), DNA, Fibroblast growth Factor-2 and the drug doxorubicin. Furthermore, IONP-labelling of MSC exosomes is reviewed. Also, limitations of IONP-labelling are addressed and methods of tackling those challenges are suggested.
Collapse
|
16
|
Islam J, So KH, Kc E, Moon HC, Kim A, Hyun SH, Kim S, Park YS. Transplantation of human embryonic stem cells alleviates motor dysfunction in AAV2-Htt171-82Q transfected rat model of Huntington's disease. Stem Cell Res Ther 2021; 12:585. [PMID: 34809707 PMCID: PMC8607638 DOI: 10.1186/s13287-021-02653-7] [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: 04/05/2021] [Accepted: 11/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human embryonic stem cells (hESCs) transplantation had shown to provide a potential source of cells in neurodegenerative disease studies and lead to behavioral recovery in lentivirus transfected or, toxin-induced Huntington's disease (HD) rodent model. Here, we aimed to observe if transplantation of superparamagnetic iron oxide nanoparticle (SPION)-labeled hESCs could migrate in the neural degenerated area and improve motor dysfunction in an AAV2-Htt171-82Q transfected Huntington rat model. METHODS All animals were randomly allocated into three groups at first: HD group, sham group, and control group. After six weeks, the animals of the HD group and sham group were again divided into two subgroups depending on animals receiving either ipsilateral or contralateral hESCs transplantation. We performed cylinder test and stepping test every two weeks after AAV2-Htt171-82Q injection and hESCs transplantation. Stem cell tracking was performed once per two weeks using T2 and T2*-weighted images at 4.7 Tesla MRI. We also performed immunohistochemistry and immunofluorescence staining to detect the presence of hESCs markers, huntingtin protein aggregations, and iron in the striatum. RESULTS After hESCs transplantation, the Htt virus-injected rats exhibited significant behavioral improvement in behavioral tests. SPION labeled hESCs showed migration with hypointense signal in MRI. The cells were positive with βIII-tubulin, GABA, and DARPP32. CONCLUSION Collectively, our results suggested that hESCs transplantation can be a potential treatment for motor dysfunction of Huntington's disease.
Collapse
Affiliation(s)
- Jaisan Islam
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyoung Ha So
- Institute for Stem Cell & Regenerative Medicine (ISCRM), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Elina Kc
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyeong Cheol Moon
- Department of Neurosurgery, Gammaknife Icon Center, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Aryun Kim
- Department of Neurology, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Sang Hwan Hyun
- Institute for Stem Cell & Regenerative Medicine (ISCRM), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Soochong Kim
- Institute for Stem Cell & Regenerative Medicine (ISCRM), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Young Seok Park
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea.
- Institute for Stem Cell & Regenerative Medicine (ISCRM), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.
- Department of Neurosurgery, Gammaknife Icon Center, Chungbuk National University Hospital, Cheongju, Republic of Korea.
- Department of Neurosurgery, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, 776, 1 Sunhwanro, Seowon-gu, Cheongju-si, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
17
|
Mundy DC, Goldberg JL. Nanoparticles as Cell Tracking Agents in Human Ocular Cell Transplantation Therapy. CURRENT OPHTHALMOLOGY REPORTS 2021. [DOI: 10.1007/s40135-021-00275-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
18
|
Moonshi SS, Wu Y, Ta HT. Visualizing stem cells in vivo using magnetic resonance imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1760. [PMID: 34651465 DOI: 10.1002/wnan.1760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/18/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022]
Abstract
Stem cell (SC) therapies displayed encouraging efficacy and clinical outcome in various disorders. Despite this huge hype, clinical translation of SC therapy has been disheartening due to contradictory results from clinical trials. The ability to monitor migration and engraftment of cells in vivo represents an ideal strategy in cell therapy. Therefore, suitable imaging approach to track MSCs would allow understanding of migratory and homing efficiency, optimal route of delivery and engraftment of cells at targeted location. Hence, longitudinal tracking of SCs is crucial for the optimization of treatment parameters, leading to improved clinical outcome and translation. Magnetic resonance imaging (MRI) represents a suitable imaging modality to observe cells non-invasively and repeatedly. Tracking is achieved when cells are incubated prior to implantation with appropriate contrast agents (CA) or tracers which can then be detected in an MRI scan. This review explores and emphasizes the importance of monitoring the distribution and fate of SCs post-implantation using current contrast agents, such as positive CAs including paramagnetic metals (gadolinium), negative contrast agents such as superparamagnetic iron oxides and 19 F containing tracers, specifically for the in vivo tracking of MSCs using MRI. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Shehzahdi Shebbrin Moonshi
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia
| | - Yuao Wu
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia
| | - Hang Thu Ta
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia.,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland, Australia.,School of Environment and Science, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
19
|
Mousavinejad M, Skidmore S, Barone FG, Tyers P, Pisupati V, Poptani H, Plagge A, Barker RA, Murray P, Taylor A, Hill CJ. Assessing Human Embryonic Stem Cell-Derived Dopaminergic Neuron Progenitor Transplants Using Non-invasive Imaging Techniques. Mol Imaging Biol 2021; 22:1244-1254. [PMID: 32378000 PMCID: PMC7497430 DOI: 10.1007/s11307-020-01499-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Human pluripotent stem cell (hPSC)-derived dopaminergic neuron progenitor cells (DAPCs) are a potential therapy for Parkinson's disease (PD). However, their intracranial administration raises safety concerns including uncontrolled proliferation, migration and inflammation. Here, we apply a bimodal imaging approach to investigate the fate of DAPC transplants in the rat striatum. PROCEDURES DAPCs co-expressing luciferase and ZsGreen or labelled with micron-sized particles of iron oxide (MPIOs) were transplanted in the striatum of RNU rats (n = 6 per group). DAPCs were tracked in vivo using bioluminescence and magnetic resonance (MR) imaging modalities. RESULTS Transgene silencing in differentiating DAPCs accompanied with signal attenuation due to animal growth rendered the bioluminescence undetectable by week 2 post intrastriatal transplantation. However, MR imaging of MPIO-labelled DAPCs showed that transplanted cells remained at the site of injection for over 120 days. Post-mortem histological analysis of DAPC transplants demonstrated that labelling with either luciferase/ZsGreen or MPIOs did not affect the ability of cells to differentiate into mature dopaminergic neurons. Importantly, labelled cells did not elicit increased glial reactivity compared to non-labelled cells. CONCLUSIONS In summary, our findings support the transplantation of hPSC-derived DAPCs as a safe treatment for PD.
Collapse
Affiliation(s)
- M Mousavinejad
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - S Skidmore
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.,WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - F G Barone
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - P Tyers
- John van Geest Centre for Brain Repair & Department of Neurology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - V Pisupati
- WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - H Poptani
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - A Plagge
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - R A Barker
- WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.,John van Geest Centre for Brain Repair & Department of Neurology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - P Murray
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - A Taylor
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| | - C J Hill
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK. .,Centre for Women's Health Research, Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, L8 7SS, UK.
| |
Collapse
|
20
|
Glover JC, Aswendt M, Boulland JL, Lojk J, Stamenković S, Andjus P, Fiori F, Hoehn M, Mitrecic D, Pavlin M, Cavalli S, Frati C, Quaini F. In vivo Cell Tracking Using Non-invasive Imaging of Iron Oxide-Based Particles with Particular Relevance for Stem Cell-Based Treatments of Neurological and Cardiac Disease. Mol Imaging Biol 2021; 22:1469-1488. [PMID: 31802361 DOI: 10.1007/s11307-019-01440-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell-based therapeutics is a rapidly developing field associated with a number of clinical challenges. One such challenge lies in the implementation of methods to track stem cells and stem cell-derived cells in experimental animal models and in the living patient. Here, we provide an overview of cell tracking in the context of cardiac and neurological disease, focusing on the use of iron oxide-based particles (IOPs) visualized in vivo using magnetic resonance imaging (MRI). We discuss the types of IOPs available for such tracking, their advantages and limitations, approaches for labeling cells with IOPs, biological interactions and effects of IOPs at the molecular and cellular levels, and MRI-based and associated approaches for in vivo and histological visualization. We conclude with reviews of the literature on IOP-based cell tracking in cardiac and neurological disease, covering both preclinical and clinical studies.
Collapse
Affiliation(s)
- Joel C Glover
- Laboratory for Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, Norway. .,Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway.
| | - Markus Aswendt
- Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich, Leo-Brandt-Str. 5, 52425, Jülich, Germany
| | - Jean-Luc Boulland
- Laboratory for Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway
| | - Jasna Lojk
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, Ljubljana, Slovenia
| | - Stefan Stamenković
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, PB 52, 10001 Belgrade, Serbia
| | - Pavle Andjus
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, PB 52, 10001 Belgrade, Serbia
| | - Fabrizio Fiori
- Department of Applied Physics, Università Politecnica delle Marche - Di.S.C.O., Via Brecce Bianche, 60131, Ancona, Italy
| | - Mathias Hoehn
- Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich, Leo-Brandt-Str. 5, 52425, Jülich, Germany
| | - Dinko Mitrecic
- Laboratory for Stem Cells, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mojca Pavlin
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, Ljubljana, Slovenia.,Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Stefano Cavalli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | |
Collapse
|
21
|
Theruvath AJ, Mahmoud EE, Wu W, Nejadnik H, Kiru L, Liang T, Felt S, Daldrup-Link HE. Ascorbic Acid and Iron Supplement Treatment Improves Stem Cell-Mediated Cartilage Regeneration in a Minipig Model. Am J Sports Med 2021; 49:1861-1870. [PMID: 33872071 PMCID: PMC8177720 DOI: 10.1177/03635465211005754] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The transplantation of mesenchymal stem cells (MSCs) into cartilage defects has led to variable cartilage repair outcomes. Previous in vitro studies have shown that ascorbic acid and reduced iron independently can improve the chondrogenic differentiation of MSCs. However, the combined effect of ascorbic acid and iron supplementation on MSC differentiation has not been investigated. PURPOSE To investigate the combined in vivo effects of ascorbic acid and a US Food and Drug Administration (FDA)-approved iron supplement on MSC-mediated cartilage repair in mature Göttingen minipigs. STUDY DESIGN Controlled laboratory study. METHODS We pretreated bone marrow-derived MSCs with ascorbic acid and the FDA-approved iron supplement ferumoxytol and then transplanted the MSCs into full-thickness cartilage defects in the distal femurs of Göttingen minipigs. Untreated cartilage defects served as negative controls. We evaluated the cartilage repair site with magnetic resonance imaging at 4 and 12 weeks after MSC implantation, followed by histological examination and immunofluorescence staining at 12 weeks. RESULTS Ascorbic acid plus iron-pretreated MSCs demonstrated a significantly better MOCART (magnetic resonance observation of cartilage repair tissue) score (73.8 ± 15.5), better macroscopic cartilage regeneration score according to the International Cartilage Repair Society (8.6 ± 2.0), better Pineda score (2.9 ± 0.8), and larger amount of collagen type II (28,469 ± 21,313) compared with untreated controls (41.3 ± 2.5, 1.8 ± 2.9, 12.8 ± 1.9, and 905 ± 1326, respectively). The obtained scores were also better than scores previously reported in the same animal model for MSC implants without ascorbic acid. CONCLUSION Pretreatment of MSCs with ascorbic acid and an FDA-approved iron supplement improved the chondrogenesis of MSCs and led to hyaline-like cartilage regeneration in the knee joints of minipigs. CLINICAL RELEVANCE Ascorbic acid and iron supplements are immediately clinically applicable. Thus, these results, in principle, could be translated into clinical applications.
Collapse
Affiliation(s)
- Ashok Joseph Theruvath
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, California, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Elhussein Elbadry Mahmoud
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, California, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA.,Department of Surgery, School of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, California, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Hossein Nejadnik
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, California, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Louise Kiru
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, California, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Tie Liang
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, California, USA
| | - Stephen Felt
- Department of Comparative Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Heike Elisabeth Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, California, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA.,Department of Pediatrics, School of Medicine, Stanford University, Stanford, California, USA.,Address correspondence to Heike E. Daldrup-Link, MD, PhD, Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, CA, 94305, USA ()
| |
Collapse
|
22
|
Chouhan RS, Horvat M, Ahmed J, Alhokbany N, Alshehri SM, Gandhi S. Magnetic Nanoparticles-A Multifunctional Potential Agent for Diagnosis and Therapy. Cancers (Basel) 2021; 13:2213. [PMID: 34062991 PMCID: PMC8124749 DOI: 10.3390/cancers13092213] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023] Open
Abstract
Magnetic nanoparticles gained considerable attention in last few years due to their remarkable properties. Superparamaganetism, non-toxicity, biocompatibility, chemical inertness, and environmental friendliness are some of the properties that make iron oxide nanoparticles (IONPs) an ideal choice for biomedical applications. Along with being easily tuneable and a tailored surface for conjugation of IONPs, their physio-chemical and biological properties can also be varied by modifying the basic parameters for synthesis that enhances the additional possibilities for designing novel magnetic nanomaterial for theranostic applications. This review highlights the synthesis, surface modification, and different applications of IONPs for diagnosis, imaging, and therapy. Furthermore, it also represents the recent report on the application of IONPs as enzyme mimetic compounds and a contrasting agent, and its significance in the field as an anticancer and antimicrobial agent.
Collapse
Affiliation(s)
- Raghuraj Singh Chouhan
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia;
| | - Milena Horvat
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia;
| | - Jahangeer Ahmed
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (J.A.); (N.A.)
| | - Norah Alhokbany
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (J.A.); (N.A.)
| | - Saad M. Alshehri
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (J.A.); (N.A.)
| | - Sonu Gandhi
- Amity Institute of Biotechnology, Amity University, Noida 201301, India
- DBT-National Institute of Animal Biotechnology (DBT-NIAB), Hyderabad 500032, India
| |
Collapse
|
23
|
Berninger MT, Rodriguez-Gonzalez P, Schilling F, Haller B, Lichtenstein T, Imhoff AB, Rummeny EJ, Anton M, Vogt S, Henning TD. Bifunctional Labeling of Rabbit Mesenchymal Stem Cells for MR Imaging and Fluorescence Microscopy. Mol Imaging Biol 2021; 22:303-312. [PMID: 31209781 DOI: 10.1007/s11307-019-01385-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE Longitudinal imaging studies are important in the translational process of stem cell-based therapies. Small animal imaging models are widely available and practical but insufficiently depict important morphologic detail. In contrary, large animal models are logistically challenging and costly but offer greater imaging quality. In order to combine the advantages of both, we developed an intermediate-sized rabbit animal model for cartilage imaging studies. PROCEDURES Rabbit mesenchymal stem cells (rMSC) were isolated as primary cultures from the bone marrow of New Zealand white rabbits. rMSC were subsequentially transduced lentivirally with eGFP and magnetically labeled with the iron oxide ferucarbotran. eGFP expression was evaluated by flow cytometry and iron uptake was analyzed by isotope dilution mass spectrometry and Prussian blue staining. Fluorescence microscopy of eGFP-transduced rMSC was performed. Viability and induction of apoptosis were assessed by XTT and caspase-3/-7 measurements. The chondrogenic potential of labeled cells was quantified by glycosaminoglycan contents in TGF-β3 induced pellet cultures. Labeled and unlabeled cells underwent magnetic resonance imaging (MRI) at 1.5 T before and after differentiation using T1-, T2-, and T2*-weighted pulse sequences. Relaxation rates were calculated. rMSCs were implanted in fibrin clots in osteochondral defects of cadaveric rabbit knees and imaged by 7 T MRI. T2* maps were calculated. Statistical analyses were performed using multiple regression models. RESULTS Efficiency of lentiviral transduction was greater than 90 %. Fluorescence signal was dose dependent. Cellular iron uptake was significant for all concentrations (p < 0.05) and dose dependent (3.3-56.5 pg Fe/cell). Labeled rMSC showed a strong, dose-dependent contrast on all MR pulse sequences and a significant decrease in T2 and T2* relaxation rates. Compared with non-transduced or unlabeled controls, there were no adverse effects on cell viability, rate of apoptosis, or chondrogenic differentiation. MRI of labeled rMSCs in osteochondral defects showed a significant signal of the transplant with additional high-resolution anatomical information. CONCLUSIONS This intermediate-sized rabbit model and its bifunctional labeling technique allow for improved depiction of anatomic detail for noninvasive in vivo rMSC tracking with MRI and for immunohistological correlation by fluorescence microscopy.
Collapse
Affiliation(s)
- Markus T Berninger
- Department of Orthopaedic Sports Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
- Department of Trauma Surgery, BG Trauma Center Murnau, Prof.-Küntscher-Strasse 8, 82418, Murnau, Germany.
| | | | - Franz Schilling
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Bernhard Haller
- Institute for Medical Statistics and Epidemiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Andreas B Imhoff
- Department of Orthopaedic Sports Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Ernst J Rummeny
- Department of Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Martina Anton
- Institute for Experimental Oncology and Therapy Research, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stephan Vogt
- Department of Orthopaedic Sports Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Tobias D Henning
- Section of Neuroradiology, Uniklinik Köln, Cologne, Germany
- Section of Neuroradiology, Krankenhaus der Barmherzigen Brüder, Trier, Germany
| |
Collapse
|
24
|
Radeloff K, Ramos Tirado M, Haddad D, Breuer K, Müller J, Hochmuth S, Hackenberg S, Scherzad A, Kleinsasser N, Radeloff A. Superparamagnetic Iron Oxide Particles (VSOPs) Show Genotoxic Effects but No Functional Impact on Human Adipose Tissue-Derived Stromal Cells (ASCs). MATERIALS 2021; 14:ma14020263. [PMID: 33430323 PMCID: PMC7825809 DOI: 10.3390/ma14020263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
Abstract
Adipose tissue-derived stromal cells (ASCs) represent a capable source for cell-based therapeutic approaches. For monitoring a cell-based application in vivo, magnetic resonance imaging (MRI) of cells labeled with iron oxide particles is a common method. It is the aim of the present study to analyze potential DNA damage, cytotoxicity and impairment of functional properties of human (h)ASCs after labeling with citrate-coated very small superparamagnetic iron oxide particles (VSOPs). Cytotoxic as well as genotoxic effects of the labeling procedure were measured in labeled and unlabeled hASCs using the MTT assay, comet assay and chromosomal aberration test. Trilineage differentiation was performed to evaluate an impairment of the differentiation potential due to the particles. Proliferation as well as migration capability were analyzed after the labeling procedure. Furthermore, the labeling of the hASCs was confirmed by Prussian blue staining, transmission electron microscopy (TEM) and high-resolution MRI. Below the concentration of 0.6 mM, which was used for the procedure, no evidence of genotoxic effects was found. At 0.6 mM, 1 mM as well as 1.5 mM, an increase in the number of chromosomal aberrations was determined. Cytotoxic effects were not observed at any concentration. Proliferation, migration capability and differentiation potential were also not affected by the procedure. Labeling with VSOPs is a useful labeling method for hASCs that does not affect their proliferation, migration and differentiation potential. Despite the absence of cytotoxicity, however, indications of genotoxic effects have been demonstrated.
Collapse
Affiliation(s)
- Katrin Radeloff
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Oldenburg, 26122 Oldenburg, Germany; (J.M.); (S.H.); (A.R.)
- Correspondence:
| | - Mario Ramos Tirado
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (M.R.T.); (S.H.); (A.S.); (N.K.)
| | - Daniel Haddad
- Fraunhofer Development Center X-ray Technology EZRT, Department Magnetic Resonance and X-ray Imaging, A Division of Fraunhofer Institute for Integrated Circuits IIS, 97074 Wuerzburg, Germany;
| | - Kathrin Breuer
- Department of Radiation Oncology, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Jana Müller
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Oldenburg, 26122 Oldenburg, Germany; (J.M.); (S.H.); (A.R.)
| | - Sabine Hochmuth
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Oldenburg, 26122 Oldenburg, Germany; (J.M.); (S.H.); (A.R.)
| | - Stephan Hackenberg
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (M.R.T.); (S.H.); (A.S.); (N.K.)
| | - Agmal Scherzad
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (M.R.T.); (S.H.); (A.S.); (N.K.)
| | - Norbert Kleinsasser
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (M.R.T.); (S.H.); (A.S.); (N.K.)
| | - Andreas Radeloff
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Oldenburg, 26122 Oldenburg, Germany; (J.M.); (S.H.); (A.R.)
| |
Collapse
|
25
|
Zlotnick HM, Clark AT, Gullbrand SE, Carey JL, Cheng XM, Mauck RL. Magneto-Driven Gradients of Diamagnetic Objects for Engineering Complex Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2005030. [PMID: 33073437 PMCID: PMC7723011 DOI: 10.1002/adma.202005030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/10/2020] [Indexed: 05/23/2023]
Abstract
Engineering complex tissues represents an extraordinary challenge and, to date, there have been few strategies developed that can easily recapitulate native-like cell and biofactor gradients in 3D materials. This is true despite the fact that mimicry of these gradients may be essential for the functionality of engineered graft tissues. Here, a non-traditional magnetics-based approach is developed to predictably position naturally diamagnetic objects in 3D hydrogels. Rather than magnetizing the objects within the hydrogel, the magnetic susceptibility of the surrounding hydrogel precursor solution is enhanced. In this way, a range of diamagnetic objects (e.g., polystyrene beads, drug delivery microcapsules, and living cells) are patterned in response to a brief exposure to a magnetic field. Upon photo-crosslinking the hydrogel precursor, object positioning is maintained, and the magnetic contrast agent diffuses out of the hydrogel, supporting long-term construct viability. This approach is applied to engineer cartilage constructs with a depth-dependent cellularity mirroring that of native tissue. These are thought to be the first results showing that magnetically unaltered cells can be magneto-patterned in hydrogels and cultured to generate heterogeneous tissues. This work provides a foundation for the formation of opposing magnetic-susceptibility-based gradients within a single continuous material.
Collapse
Affiliation(s)
- Hannah M Zlotnick
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
- McKay Orthopedic Research Laboratory, Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Andy T Clark
- Department of Physics and Center for Engineering MechanoBiology, Bryn Mawr College, Bryn Mawr, PA, 19010, USA
| | - Sarah E Gullbrand
- McKay Orthopedic Research Laboratory, Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - James L Carey
- McKay Orthopedic Research Laboratory, Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xuemei M Cheng
- Department of Physics and Center for Engineering MechanoBiology, Bryn Mawr College, Bryn Mawr, PA, 19010, USA
| | - Robert L Mauck
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
- McKay Orthopedic Research Laboratory, Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| |
Collapse
|
26
|
Umbilical Cord Mesenchymal Stem Cell-Derived Nanovesicles Potentiate the Bone-Formation Efficacy of Bone Morphogenetic Protein 2. Int J Mol Sci 2020; 21:ijms21176425. [PMID: 32899307 PMCID: PMC7504262 DOI: 10.3390/ijms21176425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/22/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
Recombinant human bone morphogenetic protein 2 (rhBMP-2) is one of the most potent osteogenic factors used to treat bone loss. However, at higher doses, rhBMP-2 does not necessarily increase bone formation but rather increases the incidence of adverse side effects. Here, we investigated whether umbilical cord mesenchymal stem cell (UCMSC)-derived nanovesicles (NVs) further increase the in vivo bone formation at high doses of rhBMP-2. In the presence of UCMSC-derived NVs, proliferation, migration, and tube formation of human umbilical vein endothelial cells were stimulated in vitro. Furthermore, migration and osteogenesis of human bone marrow-derived mesenchymal stem cells were stimulated. To examine the efficacy of UCMSC-derived NVs on in vivo bone formation, collagen sponges soaked with rhBMP-2 and UCMSC-derived NVs were used in athymic nude mice with calvarial defects. At a high rhBMP-2 dosage (500 ng/mL), UCMSC-derived NVs significantly promoted bone formation in calvarial defects; however, the UCMSC-derived NVs alone did not induce in vivo bone formation. Our results indicate that UCMSC-derived NVs can potentiate the bone formation efficacy of rhBMP-2 at a high dosage.
Collapse
|
27
|
McGinley LM, Willsey MS, Kashlan ON, Chen KS, Hayes JM, Bergin IL, Mason SN, Stebbins AW, Kwentus JF, Pacut C, Kollmer J, Sakowski SA, Bell CB, Chestek CA, Murphy GG, Patil PG, Feldman EL. Magnetic resonance imaging of human neural stem cells in rodent and primate brain. Stem Cells Transl Med 2020; 10:83-97. [PMID: 32841522 PMCID: PMC7780819 DOI: 10.1002/sctm.20-0126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Stem cell transplantation therapies are currently under investigation for central nervous system disorders. Although preclinical models show benefit, clinical translation is somewhat limited by the absence of reliable noninvasive methods to confirm targeting and monitor transplanted cells in vivo. Here, we assess a novel magnetic resonance imaging (MRI) contrast agent derived from magnetotactic bacteria, magneto‐endosymbionts (MEs), as a translatable methodology for in vivo tracking of stem cells after intracranial transplantation. We show that ME labeling provides robust MRI contrast without impairment of cell viability or other important therapeutic features. Labeled cells were visualized immediately post‐transplantation and over time by serial MRI in nonhuman primate and mouse brain. Postmortem tissue analysis confirmed on‐target grft location, and linear correlations were observed between MRI signal, cell engraftment, and tissue ME levels, suggesting that MEs may be useful for determining graft survival or rejection. Overall, these findings indicate that MEs are an effective tool for in vivo tracking and monitoring of cell transplantation therapies with potential relevance to many cellular therapy applications.
Collapse
Affiliation(s)
- Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew S Willsey
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Osama N Kashlan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin S Chen
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - John M Hayes
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ingrid L Bergin
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shayna N Mason
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Aaron W Stebbins
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Crystal Pacut
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer Kollmer
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Caleb B Bell
- Bell Biosystems, San Francisco, California, USA.,G4S Capital & Ikigai Accelerator, Santa Clara, California, USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Electrical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Neuroscience and Robotics Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Geoffrey G Murphy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Parag G Patil
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
28
|
Suciu M, Ionescu CM, Ciorita A, Tripon SC, Nica D, Al-Salami H, Barbu-Tudoran L. Applications of superparamagnetic iron oxide nanoparticles in drug and therapeutic delivery, and biotechnological advancements. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2020; 11:1092-1109. [PMID: 32802712 PMCID: PMC7404288 DOI: 10.3762/bjnano.11.94] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 07/07/2020] [Indexed: 05/13/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have unique properties with regard to biological and medical applications. SPIONs have been used in clinical settings although their safety of use remains unclear due to the great differences in their structure and in intra- and inter-patient absorption and response. This review addresses potential applications of SPIONs in vitro (formulations), ex vivo (in biological cells and tissues) and in vivo (preclinical animal models), as well as potential biomedical applications in the context of drug targeting, disease treatment and therapeutic efficacy, and safety studies.
Collapse
Affiliation(s)
- Maria Suciu
- Department of Molecular Biology and Biotechnology, Electron Microscopy Laboratory, Biology and Geology Faculty, Babes-Bolyai University, 5–7 Clinicilor Str., Cluj-Napoca, Cluj County, 400006, Romania
- Electron Microscopy Integrated Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donath Str., Cluj-Napoca, Cluj County, 400293, Romania
| | - Corina M Ionescu
- Department of Molecular Biology and Biotechnology, Electron Microscopy Laboratory, Biology and Geology Faculty, Babes-Bolyai University, 5–7 Clinicilor Str., Cluj-Napoca, Cluj County, 400006, Romania
| | - Alexandra Ciorita
- Department of Molecular Biology and Biotechnology, Electron Microscopy Laboratory, Biology and Geology Faculty, Babes-Bolyai University, 5–7 Clinicilor Str., Cluj-Napoca, Cluj County, 400006, Romania
- Electron Microscopy Integrated Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donath Str., Cluj-Napoca, Cluj County, 400293, Romania
| | - Septimiu C Tripon
- Department of Molecular Biology and Biotechnology, Electron Microscopy Laboratory, Biology and Geology Faculty, Babes-Bolyai University, 5–7 Clinicilor Str., Cluj-Napoca, Cluj County, 400006, Romania
- Electron Microscopy Integrated Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donath Str., Cluj-Napoca, Cluj County, 400293, Romania
| | - Dragos Nica
- Functional Sciences Department, Medical Faculty, University of Medicine and Pharmacy “Victor Babes”, 2 Eftimie Murgu, Timisoara, Timis County, 300041, Romania
| | - Hani Al-Salami
- Biotechnology and Drug Development Research Laboratory, the School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth Western Australia 6845, Australia
| | - Lucian Barbu-Tudoran
- Department of Molecular Biology and Biotechnology, Electron Microscopy Laboratory, Biology and Geology Faculty, Babes-Bolyai University, 5–7 Clinicilor Str., Cluj-Napoca, Cluj County, 400006, Romania
- Electron Microscopy Integrated Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donath Str., Cluj-Napoca, Cluj County, 400293, Romania
| |
Collapse
|
29
|
Nejadnik H, Jung KO, Theruvath AJ, Kiru L, Liu A, Wu W, Sulchek T, Pratx G, Daldrup-Link HE. Instant labeling of therapeutic cells for multimodality imaging. Theranostics 2020; 10:6024-6034. [PMID: 32483435 PMCID: PMC7255004 DOI: 10.7150/thno.39554] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 03/03/2020] [Indexed: 12/21/2022] Open
Abstract
Autologous therapeutic cells are typically harvested and transplanted in one single surgery. This makes it impossible to label them with imaging biomarkers through classical transfection techniques in a laboratory. To solve this problem, we developed a novel microfluidic device, which provides highly efficient labeling of therapeutic cells with imaging biomarkers through mechanoporation. Methods: Studies were performed with a new, custom-designed microfluidic device, which contains ridges, which compress adipose tissue-derived stem cells (ADSCs) during their device passage. Cell relaxation after compression leads to cell volume exchange for convective transfer of nanoparticles and nanoparticle uptake into the cell. ADSCs were passed through the microfluidic device doped with iron oxide nanoparticles and 18F-fluorodeoxyglucose (FDG). The cellular nanoparticle and radiotracer uptake was evaluated with DAB-Prussian blue, fluorescent microscopy, and inductively coupled plasma spectrometry (ICP). Labeled and unlabeled ADSCs were imaged in vitro as well as ex vivo in pig knee specimen with magnetic resonance imaging (MRI) and positron emission tomography (PET). T2 relaxation times and radiotracer signal were compared between labeled and unlabeled cell transplants using Student T-test with p<0.05. Results: We report significant labeling of ADSCs with iron oxide nanoparticles and 18F-FDG within 12+/-3 minutes. Mechanoporation of ADSCs with our microfluidic device led to significant nanoparticle (> 1 pg iron per cell) and 18F-FDG uptake (61 mBq/cell), with a labeling efficiency of 95%. The labeled ADSCs could be detected with MRI and PET imaging technologies: Nanoparticle labeled ADSC demonstrated significantly shorter T2 relaxation times (24.2±2.1 ms) compared to unlabeled cells (79.6±0.8 ms) on MRI (p<0.05) and 18F-FDG labeled ADSC showed significantly higher radiotracer uptake (614.3 ± 9.5 Bq / 1×104 cells) compared to controls (0.0 ± 0.0 Bq/ 1×104 cells) on gamma counting (p<0.05). After implantation of dual-labeled ADSCs into pig knee specimen, the labeled ADSCs revealed significantly shorter T2 relaxation times (41±0.6 ms) compared to unlabeled controls (90±1.8 ms) (p<0.05). Conclusion: The labeling of therapeutic cells with our new microfluidic device does not require any chemical intervention, therefore it is broadly and immediately clinically applicable. Cellular labeling using mechanoporation can improve our understanding of in vivo biodistributions of therapeutic cells and ultimately improve long-term outcomes of therapeutic cell transplants.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
| | - Kyung Oh Jung
- Department of Radiation Oncology, Stanford University, CA, 94305, USA
| | - Ashok J. Theruvath
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Louise Kiru
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
| | - Anna Liu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
| | - Todd Sulchek
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Guillem Pratx
- Department of Radiation Oncology, Stanford University, CA, 94305, USA
| | - Heike E. Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
- Department of Pediatrics, Stanford University, CA, 94305, USA
| |
Collapse
|
30
|
Assessment of proliferation, migration and differentiation potentials of bone marrow mesenchymal stem cells labeling with silica-coated and amine-modified superparamagnetic iron oxide nanoparticles. Cytotechnology 2020; 72:513-525. [PMID: 32394163 DOI: 10.1007/s10616-020-00397-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/04/2020] [Indexed: 10/24/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles have been widely used for cell labeling in preclinical and clinical studies, to improve labeling efficiency, particle conjugation and surface modifications are developed, but some modified SPIONs exert side-effect on physiological activity of cells, which cannot be served as ideal cell tracker. In this study, amine-modified silica-coated SPIO (SPIO@SiO2-NH2, SPIO@S-N) nanoparticles were used to label bone marrow derived mesenchymal stem cells (BM-MSCs), then the stem cell potentials were evaluated. It was found BM-MSCs could be efficiently labeled by SPIO@S-N nanoparticles. After labeling, the BM-MSCs viability kept well and the migration ability increased, but the osteogenesis and adipogenesis potentials were not impaired. In steroid associated osteonecrosis (SAON) bone defect model, stem cell implantation was performed by injection of SPIO@S-N labeled BM-MSCs into marrow cavity locally, it was found the SPIO positive cells homed to the periphery of defect region in control group, but were recruited to the defect region in poly lactic-coglycolic acid/tricalcium phosphate (PLGA/TCP) scaffold implantation group. In conclusion, SPIO@S-N nanoparticles promoted migration while retained proliferation and differentiation ability of BM-MSCs, implying this kind of nanoparticles could be served not only an ideal tracking marker but also an accelerator for stem cell homing during tissue repair.
Collapse
|
31
|
Fahmy HM, Abd El-Daim TM, Mohamed HAAENE, Mahmoud EAAEQ, Abdallah EAS, Mahmoud Hassan FEZ, Maihop DI, Amin AEAE, Mustafa ABE, Hassan FMA, Mohamed DME, Shams-Eldin EMM. Multifunctional nanoparticles in stem cell therapy for cellular treating of kidney and liver diseases. Tissue Cell 2020; 65:101371. [PMID: 32746989 DOI: 10.1016/j.tice.2020.101371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
The review gives an overview of the mechanisms of internalization and distribution of nanoparticles in stem cells this is achieved via providing analysis of the methods used in exploring the migration routes of stem cells, and their reciprocity. In addition, exploring microenvironment target in the body, and tracking the fate of exogenously transplanted stem cells by using innovative and non-invasive techniques will also be discussed. Such techniques like magnetic resonance imaging (MRI), multimodality tracking, optical imaging, and nuclear medicine imaging, which were designed to follow up stem cell migration. This review will explain the various distinctive strategies to enhance homing of labeled stem cells with nanoparticles into damaged hepatic and renal tissues, this purpose was obtained by inducing a specific gene into stem cells, various chemokines, and applying an external magnetic field. Also, this work illustrates how to improve nanoparticles uptake by using transfection agents or covalently binding an exogenous protein (i.e., Human immunodeficiency virus-Tat protein) or conjugating a receptor-specific monoclonal antibody or make modifications to iron coat. It contains stem cell labeling methods such as extracellular labeling and internalization approaches. Ultimately, our review indicates trails of researchers in nanoparticles utilization in stem cell therapy in both kidney and liver diseases.
Collapse
|
32
|
Lee JR, Park BW, Kim J, Choo YW, Kim HY, Yoon JK, Kim H, Hwang JW, Kang M, Kwon SP, Song SY, Ko IO, Park JA, Ban K, Hyeon T, Park HJ, Kim BS. Nanovesicles derived from iron oxide nanoparticles-incorporated mesenchymal stem cells for cardiac repair. SCIENCE ADVANCES 2020; 6:eaaz0952. [PMID: 32494669 PMCID: PMC7195131 DOI: 10.1126/sciadv.aaz0952] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 02/12/2020] [Indexed: 05/18/2023]
Abstract
Because of poor engraftment and safety concerns regarding mesenchymal stem cell (MSC) therapy, MSC-derived exosomes have emerged as an alternative cell-free therapy for myocardial infarction (MI). However, the diffusion of exosomes out of the infarcted heart following injection and the low productivity limit the potential of clinical applications. Here, we developed exosome-mimetic extracellular nanovesicles (NVs) derived from iron oxide nanoparticles (IONPs)-incorporated MSCs (IONP-MSCs). The retention of injected IONP-MSC-derived NVs (IONP-NVs) within the infarcted heart was markedly augmented by magnetic guidance. Furthermore, IONPs significantly increased the levels of therapeutic molecules in IONP-MSCs and IONP-NVs, which can reduce the concern of low exosome productivity. The injection of IONP-NVs into the infarcted heart and magnetic guidance induced an early shift from the inflammation phase to the reparative phase, reduced apoptosis and fibrosis, and enhanced angiogenesis and cardiac function recovery. This approach can enhance the therapeutic potency of an MSC-derived NV therapy.
Collapse
Affiliation(s)
- Ju-Ro Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Bong-Woo Park
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jonghoon Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Yeon Woong Choo
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Han Young Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Kee Yoon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeok Kim
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, Seoul 06591, Republic of Korea
| | - Ji-Won Hwang
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, Seoul 06591, Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seuk Young Song
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - In Ok Ko
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Ji-Ae Park
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Taeghwan Hyeon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul 08826, Republic of Korea
- Institute of Chemical Processes, Institute of Engineering Research, BIOMAX, Seoul National University, Seoul 08826, Republic of Korea
| | - Hun-Jun Park
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, Seoul 06591, Republic of Korea
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Corresponding author. (B.-S.K.); (H.-J.P.)
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Chemical Processes, Institute of Engineering Research, BIOMAX, Seoul National University, Seoul 08826, Republic of Korea
- Corresponding author. (B.-S.K.); (H.-J.P.)
| |
Collapse
|
33
|
Toxicity and Functional Impairment in Human Adipose Tissue-Derived Stromal Cells (hASCs) Following Long-Term Exposure to Very Small Iron Oxide Particles (VSOPs). NANOMATERIALS 2020; 10:nano10040741. [PMID: 32294970 PMCID: PMC7221569 DOI: 10.3390/nano10040741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/03/2020] [Accepted: 04/05/2020] [Indexed: 11/20/2022]
Abstract
Magnetic nanoparticles (NPs), such as very small iron oxide NPs (VSOPs) can be used for targeted drug delivery, cancer treatment or tissue engineering. Another important field of application is the labelling of mesenchymal stem cells to allow in vivo tracking and visualization of transplanted cells using magnetic resonance imaging (MRI). For these NPs, however, various toxic effects, as well as functional impairment of the exposed cells, are described. The present study evaluates the influence of VSOPs on the multilineage differentiation ability and cytokine secretion of human adipose tissue derived stromal cells (hASCs) after long-term exposure. Human ASCs were labelled with VSOPs, and the efficacy of the labelling was documented over 4 weeks in vitro cultivation of the labelled cells. Unlabelled hASCs served as negative controls. Four weeks after labelling, adipogenic and osteogenic differentiation was histologically evaluated and quantified by polymerase chain reaction (PCR). Changes in gene expression of IL-6, IL-8, VEGF and caspase 3 were determined over 4 weeks. Four weeks after the labelling procedure, labelled and unlabelled hASCs did not differ in the gene expression of IL-6, IL-8, VEGF and caspase 3. Furthermore, the labelling procedure had no influence on the multidifferentiation ability of hASC. The percentage of labelled cells decreased during in vitro expansion over 4 weeks. Labelling with VSOPs and long-term intracellular disposition probably have no influence on the physiological functions of hASCs. This could be important for the future in vivo use of iron oxide NPs.
Collapse
|
34
|
Goranov V, Shelyakova T, De Santis R, Haranava Y, Makhaniok A, Gloria A, Tampieri A, Russo A, Kon E, Marcacci M, Ambrosio L, Dediu VA. 3D Patterning of cells in Magnetic Scaffolds for Tissue Engineering. Sci Rep 2020; 10:2289. [PMID: 32041994 PMCID: PMC7010825 DOI: 10.1038/s41598-020-58738-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 12/18/2019] [Indexed: 12/03/2022] Open
Abstract
A three dimensional magnetic patterning of two cell types was realised in vitro inside an additive manufactured magnetic scaffold, as a conceptual precursor for the vascularised tissue. The realisation of separate arrangements of vascular and osteoprogenitor cells, labelled with biocompatible magnetic nanoparticles, was established on the opposite sides of the scaffold fibres under the effect of non-homogeneous magnetic gradients and loading magnetic configuration. The magnetisation of the scaffold amplified the guiding effects by an additional trapping of cells due to short range magnetic forces. The mathematical modelling confirmed the strong enhancement of the magnetic gradients and their particular geometrical distribution near the fibres, defining the preferential cell positioning on the micro-scale. The manipulation of cells inside suitably designed magnetic scaffolds represents a unique solution for the assembling of cellular constructs organised in biologically adequate arrangements.
Collapse
Affiliation(s)
- V Goranov
- Institute for Nanostructured Materials, CNR-ISMN, Via Gobetti 101, 40129, Bologna, Italy.
- BioDevice Systems, Praha 10, Vršovice, Bulharská, 996/20, Czech Republic.
| | - T Shelyakova
- IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136, Bologna, Italy.
| | - R De Santis
- Institute of Polymers, Composites and Biomaterials, CNR-IPCB, V.le J.F. Kennedy 54 - Pad. 20 Mostra d'Oltremare, 80125, Naples, Italy
| | - Y Haranava
- BioDevice Systems, Praha 10, Vršovice, Bulharská, 996/20, Czech Republic
| | - A Makhaniok
- BioDevice Systems, Praha 10, Vršovice, Bulharská, 996/20, Czech Republic
| | - A Gloria
- Institute of Polymers, Composites and Biomaterials, CNR-IPCB, V.le J.F. Kennedy 54 - Pad. 20 Mostra d'Oltremare, 80125, Naples, Italy
| | - A Tampieri
- Institute of Science and Technology for Ceramics, CNR-ISTEC, Via Granarolo 64, 48018, Faenza, Italy
| | - A Russo
- IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136, Bologna, Italy
| | - E Kon
- Humanitas University Department of Biomedical Sciences, Via Manzoni 113, 20089 Rozzano, Milano, Italy
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano - Milan, Italy
- First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - M Marcacci
- Humanitas University Department of Biomedical Sciences, Via Manzoni 113, 20089 Rozzano, Milano, Italy
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano - Milan, Italy
| | - L Ambrosio
- Institute of Polymers, Composites and Biomaterials, CNR-IPCB, V.le J.F. Kennedy 54 - Pad. 20 Mostra d'Oltremare, 80125, Naples, Italy
| | - V A Dediu
- Institute for Nanostructured Materials, CNR-ISMN, Via Gobetti 101, 40129, Bologna, Italy.
| |
Collapse
|
35
|
Ledda M, Fioretti D, Lolli MG, Papi M, Di Gioia C, Carletti R, Ciasca G, Foglia S, Palmieri V, Marchese R, Grimaldi S, Rinaldi M, Lisi A. Biocompatibility assessment of sub-5 nm silica-coated superparamagnetic iron oxide nanoparticles in human stem cells and in mice for potential application in nanomedicine. NANOSCALE 2020; 12:1759-1778. [PMID: 31895375 DOI: 10.1039/c9nr09683c] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ultrasmall superparamagnetic iron oxide nanoparticles with a size <5 nm are emerging nanomaterials for their excellent biocompatibility, chemical stability, and tunable surface modifications. The applications explored include dual-modal or multi-modal imaging, drug delivery, theranostics and, more recently, magnetic resonance angiography. Good biocompatibility and biosafety are regarded as the preliminary requirements for their biomedical applications and further exploration in this field is still required. We previously synthesized and characterized ultrafine (average core size of 3 nm) silica-coated superparamagnetic iron oxide fluorescent nanoparticles, named sub-5 SIO-Fl, uniform in size, shape, chemical properties and composition. The cellular uptake and in vitro biocompatibility of the as-synthesized nanoparticles were demonstrated in a human colon cancer cellular model. Here, we investigated the biocompatibility of sub-5 SIO-Fl nanoparticles in human Amniotic Mesenchymal Stromal/Stem Cells (hAMSCs). Kinetic analysis of cellular uptake showed a quick nanoparticle internalization in the first hour, increasing over time and after long exposure (48 h), the uptake rate gradually slowed down. We demonstrated that after internalization, sub-5 SIO-Fl nanoparticles neither affect hAMSC growth, viability, morphology, cytoskeletal organization, cell cycle progression, immunophenotype, and the expression of pro-angiogenic and immunoregulatory paracrine factors nor the osteogenic and myogenic differentiation markers. Furthermore, sub-5 SIO-Fl nanoparticles were intravenously injected into mice to investigate the in vivo biodistribution and toxicity profile for a time period of 7 weeks. Our findings showed an immediate transient accumulation of nanoparticles in the kidney, followed by the liver and lungs, where iron contents increased over a 7-week period. Histopathology, hematology, serum pro-inflammatory response, body weight and mortality studies demonstrated a short- and long-term biocompatibility and biosafety profile with no apparent acute and chronic toxicity caused by these nanoparticles in mice. Overall, these results suggest the feasibility of using sub-5 SIO-Fl nanoparticles as a promising agent for stem cell magnetic targeting as well as for diagnostic and therapeutic applications in oncology.
Collapse
Affiliation(s)
- Mario Ledda
- Institute of Translational Pharmacology (IFT), Department of Biomedical Sciences, National Research Council (CNR), via del Fosso del Cavaliere 100, 00133 Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Avval ZM, Malekpour L, Raeisi F, Babapoor A, Mousavi SM, Hashemi SA, Salari M. Introduction of magnetic and supermagnetic nanoparticles in new approach of targeting drug delivery and cancer therapy application. Drug Metab Rev 2019; 52:157-184. [PMID: 31834823 DOI: 10.1080/03602532.2019.1697282] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In this article, the recent applications of different types of magnetic nanoparticles such as α-Fe2O3 (hematite), γ-Fe2O3 (maghemite), Fe3O4 (magnetite), hexagonal (MFe12O19), garnet (M3Fe5O12) and spinel (MFe2O4), where M represents one or more bivalent transition metals (Mn, Fe, Co, Ni, Ba, Sr, Cu, and Zn), and different materials for coating the surface of magnetic nanoparticles like poly lactic acid (PLA), doxorubicin hydrophobic (DOX-HCL), paclitaxel (PTX), EPPT-FITC, oleic acid, tannin, 3-Aminopropyltriethoxysilane (APTES), multi-wall carbon nanotubes (CNTs), polyethylenimine (PEI) and polyarabic acid in drug delivery, biomedicine and treatment of cancer, specially chemotherapy, are reviewed. MNPs possess large surface area to volume ratios because of their nano-size, low surface charge at physiological pH and they aggregate easily in solution due to their essential magnetic nature. These materials are widely used in biology and medicine in many cases. One targeted delivery technique that has gained prominence in recent years is the use of magnetic nanoparticles. In these systems, therapeutic compounds are attached to biocompatible magnetic nanoparticles and magnetic fields generated outside the body are focused on specific targets in vivo. The fields capture the particle complex, resulting in enhanced delivery to the target site. Also, the application of brand new supermagnetic nanoparticles, like Ba,SrFe12O19, is considered and studied in this paper.
Collapse
Affiliation(s)
| | - Leila Malekpour
- Department of Chemistry, Payame Noor University, Ardabil, Iran
| | - Farzad Raeisi
- Department of Chemistry, Payame Noor University, Ardabil, Iran
| | - Aziz Babapoor
- Department of Chemical Engineering, University of Mohaghegh Ardabili (UMA), Ardabil, Iran
| | - Seyyed Mojtaba Mousavi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Alireza Hashemi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marjan Salari
- Department of Civil and Environmental Engineering, Sirjan University of Technology, Kerman, Iran
| |
Collapse
|
37
|
Arifin DR, Kulkarni M, Kadayakkara D, Bulte JWM. Fluorocapsules allow in vivo monitoring of the mechanical stability of encapsulated islet cell transplants. Biomaterials 2019; 221:119410. [PMID: 31421313 PMCID: PMC6717436 DOI: 10.1016/j.biomaterials.2019.119410] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/02/2019] [Indexed: 02/06/2023]
Abstract
Clinical trials that have used encapsulated islet cell therapy have been few and overall disappointing. This is due in part to the lack of suitable methods to monitor the integrity vs. rupture of transplanted microcapsules over time. Fluorocapsules were synthesized by embedding emulsions of perfluoro-15-crown-5-ether (PFC), a bioinert compound detectable by 19F MRI, into dual-alginate layer, Ba2+-gelled alginate microcapsules. Fluorocapsules were spherical with an apparent smooth surface and an average diameter of 428 ± 52 μm. After transplantation into mice, the 19F MRI signal of capsules remained stable for up to 90 days, corresponding to the total number of intact fluorocapsules. When single-alginate layer capsules were ruptured with alginate lyase, the 19F MRI signal dissipated within 4 days. For fluoroencapsulated luciferase-expressing mouse βTC6 insulinoma cells implanted into autoimmune NOD/ShiLtJ mice and subjected to alginate-lyase induced capsule rupture in vivo, the 19F MRI signal decreased sharply over time along with a decrease in bioluminescence imaging signal used as a measure of cell viability in vivo. These results indicate that maintenance of capsule integrity is essential for preserving transplanted cell survival, where a decrease in 19F MRI signal may serve as a predictive imaging surrogate biomarker for impending failure of encapsulated islet cell therapy.
Collapse
Affiliation(s)
- Dian R Arifin
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mangesh Kulkarni
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Deepak Kadayakkara
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Chemical & Biomolecular Engineering, The Johns Hopkins University Whiting School of Engineering, Baltimore, MD, 21218, USA; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
38
|
Liu XL, Chen S, Zhang H, Zhou J, Fan HM, Liang XJ. Magnetic Nanomaterials for Advanced Regenerative Medicine: The Promise and Challenges. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1804922. [PMID: 30511746 DOI: 10.1002/adma.201804922] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/24/2018] [Indexed: 06/09/2023]
Abstract
The recent emergence of numerous nanotechnologies is expected to facilitate the development of regenerative medicine, which is a tissue regeneration technique based on the replacement/repair of diseased tissue or organs to restore the function of lost, damaged, and aging cells in the human body. In particular, the unique magnetic properties and specific dimensions of magnetic nanomaterials make them promising innovative components capable of significantly advancing the field of tissue regeneration. Their potential applications in tissue regeneration are the focus here, beginning with the fundamentals of magnetic nanomaterials. How nanomaterials-both those that are intrinsically magnetic and those that respond to an externally applied magnetic field-can enhance the efficiency of tissue regeneration is also described. Applications including magnetically controlled cargo delivery and release, real-time visualization and tracking of transplanted cells, magnetic regulation of cell proliferation/differentiation, and magnetic activation of targeted ion channels and signal pathways involved in regeneration are highlighted, and comments on the perspectives and challenges in magnetic nanomaterial-based tissue regeneration are given.
Collapse
Affiliation(s)
- Xiao-Li Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shizhu Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, P. R. China
| | - Huan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710069, P. R. China
| | - Jin Zhou
- Tissue Engineering Research Center of the Academy of Military Medical Sciences, No. 27, Taiping Road, Haidian District, Beijing, 100850, P. R. China
| | - Hai-Ming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710069, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
39
|
Cai X, Zhu Q, Zeng Y, Zeng Q, Chen X, Zhan Y. Manganese Oxide Nanoparticles As MRI Contrast Agents In Tumor Multimodal Imaging And Therapy. Int J Nanomedicine 2019; 14:8321-8344. [PMID: 31695370 PMCID: PMC6814316 DOI: 10.2147/ijn.s218085] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 10/02/2019] [Indexed: 01/09/2023] Open
Abstract
Contrast agents (CAs) play a crucial role in high-quality magnetic resonance imaging (MRI) applications. At present, as a result of the Gd-based CAs which are associated with renal fibrosis as well as the inherent dark imaging characteristics of superparamagnetic iron oxide nanoparticles, Mn-based CAs which have a good biocompatibility and bright images are considered ideal for MRI. In addition, manganese oxide nanoparticles (MONs, such as MnO, MnO2, Mn3O4, and MnOx) have attracted attention as T1-weighted magnetic resonance CAs due to the short circulation time of Mn(II) ion chelate and the size-controlled circulation time of colloidal nanoparticles. In this review, recent advances in the use of MONs as MRI contrast agents for tumor detection and diagnosis are reported, as are the advances in in vivo toxicity, distribution and tumor microenvironment-responsive enhanced tumor chemotherapy and radiotherapy as well as photothermal and photodynamic therapies.
Collapse
Affiliation(s)
- Xiaoxia Cai
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, People’s Republic of China
| | - Qingxia Zhu
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, People’s Republic of China
| | - Yun Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, People’s Republic of China
| | - Qi Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, People’s Republic of China
| | - Xueli Chen
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, People’s Republic of China
| | - Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
40
|
Wang P, Ma S, Ning G, Chen W, Wang B, Ye D, Chen B, Yang Y, Jiang Q, Gu N, Sun J. Entry-Prohibited Effect of kHz Pulsed Magnetic Field Upon Interaction Between SPIO Nanoparticles and Mesenchymal Stem Cells. IEEE Trans Biomed Eng 2019; 67:1152-1158. [PMID: 31369367 DOI: 10.1109/tbme.2019.2931774] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE The interaction between superparamagnetic iron oxide (SPIO) nanoparticles and mesenchymal stem cells (MSCs) in the presence of pulsed magnetic field (PMF) has become an important area of research in recent years. METHODS A parameter-adjustable pulsed magnetic field was developed based on the principle of insulated gate bipolar translator transistor-controlled discharge of large capacitances. The internalizations of SPIO nanoparticles by MSCs were investigated under the treatment of PMF in both intermittent stimulation mode and continuous stimulation mode. RESULTS The intensities and frequencies of pulsed magnetic field can be adjustable in the range of 1.9-4.6 mT and 3-5 kHz, respectively. This PMF was safe to the MSCs. However, the uptake of SPIO nanoparticles by MSCs was significantly prohibited under the treatment of kHz-ranged PMF while the 10 Hz PMF enhanced the cellular uptake of nanoparticles. This phenomenon was relative with the magnetic effect of the PMF with different frequency. CONCLUSION The PMF can be used to effectively regulate the cellular uptake of SPIO nanoparticles and the mechanism lies in the magnetic effect. SIGNIFICANCE The interaction between SPIO nanoparticles and the MSCs is a fundamental and important issue for nanomedicine and stem cell research. Our results demonstrate that the external magnetic field can be used to regulate their interaction. We believe that this safe, facile, and flexible method will greatly promote the development and clinical translation of regenerative medicine and nanomedicine.
Collapse
|
41
|
The Effect of Uncoated SPIONs on hiPSC-Differentiated Endothelial Cells. Int J Mol Sci 2019; 20:ijms20143536. [PMID: 31331030 PMCID: PMC6678752 DOI: 10.3390/ijms20143536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 01/29/2023] Open
Abstract
Background: Endothelial progenitor cells (EPCs) were indicated in vascular repair, angiogenesis of ischemic organs, and inhibition of formation of initial hyperplasia. Differentiation of endothelial cells (ECs) from human induced pluripotent stem cells (hiPSC)-derived endothelial cells (hiPSC-ECs) provides an unlimited supply for clinical application. Furthermore, magnetic cell labelling offers an effective way of targeting and visualization of hiPSC-ECs and is the next step towards in vivo studies. Methods: ECs were differentiated from hiPSCs and labelled with uncoated superparamagnetic iron-oxide nanoparticles (uSPIONs). uSPION uptake was compared between hiPSC-ECs and mature ECs isolated from patients by software analysis of microscopy pictures after Prussian blue cell staining. The acute and long-term cytotoxic effects of uSPIONs were evaluated by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay) and Annexin assay. Results: We showed, for the first time, uptake of uncoated SPIONs (uSPIONs) by hiPSC-ECs. In comparison with mature ECs of identical genetic background hiPSC-ECs showed lower uSPION uptake. However, all the studied endothelial cells were effectively labelled and showed magnetic properties even with low labelling concentration of uSPIONs. uSPIONs prepared by microwave plasma synthesis did not show any cytotoxicity nor impair endothelial properties. Conclusion: We show that hiPSC-ECs labelling with low concentration of uSPIONs is feasible and does not show any toxic effects in vitro, which is an important step towards animal studies.
Collapse
|
42
|
Radeloff K, Radeloff A, Tirado MR, Scherzad A, Hagen R, Kleinsasser NH, Hackenberg S. Long-Term Impact of Zinc Oxide Nanoparticles on Differentiation and Cytokine Secretion of Human Adipose-Derived Stromal Cells. MATERIALS 2019; 12:ma12111823. [PMID: 31195623 PMCID: PMC6600967 DOI: 10.3390/ma12111823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/24/2019] [Accepted: 06/04/2019] [Indexed: 01/01/2023]
Abstract
Zinc oxide nanoparticles (ZnO-NPs) are widely utilized, for example in manufacturing paints and in the cosmetic industry. In addition, there is raising interest in the application of NPs in stem cell research. However, cytotoxic, genotoxic and pro-inflammatory effects were shown for NPs. The aim of this study was to evaluate the impact of ZnO-NPs on cytokine secretion and differentiation properties of human adipose tissue-derived stromal cells (ASCs). Human ASCs were exposed to the subtoxic concentration of 0.2 µg/mL ZnO-NPs for 24 h. After four weeks of cultivation, adipogenic and osteogenic differentiation procedures were performed. The multi-differentiation potential was confirmed histologically and using polymerase chain reaction (PCR). In addition, the gene expression of IL-6, IL-8, vascular endothelial growth factor (VEGF) and caspase 3 was analyzed. Over the course of four weeks after ZnO-NPs exposure, no significant differences were detected in the gene expression of IL-6, IL-8, VEGF and caspase 3 compared to non-exposed cells. The differentiation was also not affected by the ZnO-NPs. These findings underline the fact, that functionality of ASCs is likely to be unaffected by ZnO-NPs, despite a long-term disposition of NPs in the cells, supposing that the starting concentration was safely in the non-toxic range. This might provide important information for single-use nanomedical applications of ZnO-NPs.
Collapse
Affiliation(s)
- Katrin Radeloff
- Department of Otorhinolaryngology, Head and Neck Surgery, Evangelisches Krankenhaus, Carl von Ossietzky-University, 26122 Oldenburg, Germany.
| | - Andreas Radeloff
- Department of Otorhinolaryngology, Head and Neck Surgery, Evangelisches Krankenhaus, Carl von Ossietzky-University, 26122 Oldenburg, Germany.
| | - Mario Ramos Tirado
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius-Maximilian-University, 97080 Wuerzburg, Germany.
| | - Agmal Scherzad
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius-Maximilian-University, 97080 Wuerzburg, Germany.
| | - Rudolf Hagen
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius-Maximilian-University, 97080 Wuerzburg, Germany.
| | - Norbert H Kleinsasser
- Department of Otorhinolaryngology, Head and Neck Surgery, Kepler-University, 4021 Linz, Austria.
| | - Stephan Hackenberg
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius-Maximilian-University, 97080 Wuerzburg, Germany.
| |
Collapse
|
43
|
Theruvath AJ, Nejadnik H, Lenkov O, Yerneni K, Li K, Kuntz L, Wolterman C, Tuebel J, Burgkart R, Liang T, Felt S, Daldrup-Link HE. Tracking Stem Cell Implants in Cartilage Defects of Minipigs by Using Ferumoxytol-enhanced MRI. Radiology 2019; 292:129-137. [PMID: 31063081 DOI: 10.1148/radiol.2019182176] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Cartilage repair outcomes of matrix-associated stem cell implants (MASIs) in patients have been highly variable. Conventional MRI cannot help distinguish between grafts that will and grafts that will not repair the underlying cartilage defect until many months after the repair. Purpose To determine if ferumoxytol nanoparticle labeling could be used to depict successful or failed MASIs compared with conventional MRI in a large-animal model. Materials and Methods Between January 2016 and December 2017, 10 Göttingen minipigs (n = 5 male; n = 5 female; mean age, 6 months ± 5.1; age range, 4-20 months) received implants of unlabeled (n = 12) or ferumoxytol-labeled (n = 20) viable and apoptotic MASIs in cartilage defects of the distal femur. All MASIs were serially imaged with MRI on a 3.0-T imaging unit at week 1 and weeks 2, 4, 8, 12, and 24, with calculation of T2 relaxation times. Cartilage regeneration outcomes were assessed by using the MR observation of cartilage repair tissue (MOCART) score (scale, 0-100), the Pineda score, and histopathologic quantification of collagen 2 production in the cartilage defect. Findings were compared by using the unpaired Wilcoxon rank sum test, a linear regression model, the Fisher exact test, and Pearson correlation. Results Ferumoxytol-labeled MASIs showed significant T2 shortening (22.2 msec ± 3.2 vs 27.9 msec ± 1.8; P < .001) and no difference in cartilage repair outcomes compared with unlabeled control MASIs (P > .05). At week 2 after implantation, ferumoxytol-labeled apoptotic MASIs showed a loss of iron signal and higher T2 relaxation times compared with ferumoxytol-labeled viable MASIs (26.6 msec ± 4.9 vs 20.8 msec ± 5.3; P = .001). Standard MRI showed incomplete cartilage defect repair of apoptotic MASIs at 24 weeks. Iron signal loss at 2 weeks correlated with incomplete cartilage repair, diagnosed at histopathologic examination at 12-24 weeks. Conclusion Ferumoxytol nanoparticle labeling can accelerate the diagnosis of successful and failed matrix-associated stem cell implants at MRI in a large-animal model. © RSNA, 2019 Online supplemental material is available for this article. See also the editorial by Sneag and Potter in this issue.
Collapse
Affiliation(s)
- Ashok J Theruvath
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Hossein Nejadnik
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Olga Lenkov
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Ketan Yerneni
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Kai Li
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Lara Kuntz
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Cody Wolterman
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Jutta Tuebel
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Rainer Burgkart
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Tie Liang
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Stephen Felt
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| | - Heike E Daldrup-Link
- From the Department of Radiology and Molecular Imaging Program at Stanford (MIPS) (A.J.T., H.N., O.L., K.Y., K.L., L.K., C.W., T.L., H.E.D.), Department of Comparative Medicine (S.F.), and Department of Pediatrics (H.E.D.), Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); and Department of Orthopedics and Sportorthopedics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany (L.K., J.T., R.B.)
| |
Collapse
|
44
|
Negi H, Takeuchi S, Kamei N, Yanada S, Adachi N, Ochi M. In Vitro Safety and Quality of Magnetically Labeled Human Mesenchymal Stem Cells Preparation for Cartilage Repair. Tissue Eng Part C Methods 2019; 25:324-333. [PMID: 31002015 DOI: 10.1089/ten.tec.2019.0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
IMPACT STATEMENT This study is very important for a preclinical assessment of the safety and quality of magnetically labeled mesenchymal stem cells (MSCs) for use in cartilage repair. The findings of this study show that magnetic labeling with an appropriate density of magnetic particles has no harmful effects on the safety and quality of MSCs.
Collapse
Affiliation(s)
- Hiroshi Negi
- 1 Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Naosuke Kamei
- 1 Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan.,3 Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | | | - Nobuo Adachi
- 1 Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mitsuo Ochi
- 4 Hiroshima University, Higashihiroshima, Japan
| |
Collapse
|
45
|
Wu MR, Hsiao JK, Liu HM, Huang YY, Tseng YJ, Chou PT, Weng TI, Yang CY. In vivo imaging of insulin-secreting human pancreatic ductal cells using MRI reporter gene technique: A feasibility study. Magn Reson Med 2019; 82:763-774. [PMID: 30957300 DOI: 10.1002/mrm.27749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE The purpose of this study was to investigate the feasibility of in vivo imaging of human pancreatic ductal cells by OATP1B3 reporter gene under MRI. METHODS A human cell line (PANC-1) derived from the pancreatic ductal epithelium was used in this study. After transduction of OATP1B3, the cellular physiological functions and the ability of intracellular uptake of the MRI contrast medium (Gd-EOB-DTPA) were examined. Induced differentiation of the PANC-1 cells into hormone-secreting cells were performed to simulate pancreatic β-like cells. The hormone-secreting cells were implanted into rats and in vivo MRI was evaluated. RESULTS The mRNA and proteins of OATP1B3 were highly expressed. No significant change of cellular physiological functions was found after the expression. After induced differentiation, the hormone secretion capacities of the OATP1B3-expressing PANC-1 cells were confirmed. Intra-cellular uptake of Gd-EOB-DTPA was determined in vitro by inductively coupled plasma mass spectrometry and MRI. In vivo MRI of the OATP1B3-expressing xenograft revealed an increased signal intensity after contrast enhancement. CONCLUSION OATP1B3 can be used as a safe and feasible in vivo MRI gene reporter for human pancreatic ductal cells.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Medical Imaging, Buddhist Tzu Chi General Hospital, Taipei Branch, New Taipei city, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Buddhist Tzu Chi General Hospital, Taipei Branch, New Taipei city, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hon-Man Liu
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan.,Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Radiology and Medical Imaging, Fu-Jen Catholic University and Hospital, New Taipei City, Taiwan
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Jui Tseng
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, Taiwan
| | - Pi-Tai Chou
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, Taiwan
| | - Te-I Weng
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Yi Yang
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Medical Imaging, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan.,School of Medicine for International Students, I-Shou University, Kaohsiung City, Taiwan
| |
Collapse
|
46
|
Noninvasive Monitoring of Allogeneic Stem Cell Delivery with Dual-Modality Imaging-Visible Microcapsules in a Rabbit Model of Peripheral Arterial Disease. Stem Cells Int 2019; 2019:9732319. [PMID: 31001343 PMCID: PMC6437732 DOI: 10.1155/2019/9732319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/06/2019] [Accepted: 01/28/2019] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapies, although promising for treating peripheral arterial disease (PAD), often suffer from low engraftment rates and the inability to confirm the delivery success and track cell distribution and engraftment. Stem cell microencapsulation combined with imaging contrast agents may provide a means to simultaneously enhance cell survival and enable cell tracking with noninvasive imaging. Here, we have evaluated a novel MRI- and X-ray-visible microcapsule formulation for allogeneic mesenchymal stem cell (MSC) delivery and tracking in a large animal model. Bone marrow-derived MSCs from male New Zealand White rabbits were encapsulated using a modified cell encapsulation method to incorporate a dual-modality imaging contrast agent, perfluorooctyl bromide (PFOB). PFOB microcapsules (PFOBCaps) were then transplanted into the medial thigh of normal or PAD female rabbits. In vitro MSC viability remained high (79 ± 5% at 4 weeks of postencapsulation), and as few as two and ten PFOBCaps could be detected in phantoms using clinical C-arm CT and 19F MRI, respectively. Successful injections of PFOBCaps in the medial thigh of normal (n = 15) and PAD (n = 16) rabbits were demonstrated on C-arm CT at 1-14 days of postinjection. Using 19F MRI, transplanted PFOBCaps were clearly identified as “hot spots” and showed one-to-one correspondence to the radiopacities on C-arm CT. Concordance of 19F MRI and C-arm CT locations of PFOBCaps with postmortem locations was high (95%). Immunohistological analysis revealed high MSC survival in PFOBCaps (>56%) two weeks after transplantation while naked MSCs were no longer viable beyond three days after delivery. These findings demonstrate that PFOBCaps could maintain cell viability even in the ischemic tissue and provide a means to monitor cell delivery and track engraftment using clinical noninvasive imaging systems.
Collapse
|
47
|
Biosynthesis of magnetic nanoparticles from nano-degradation products revealed in human stem cells. Proc Natl Acad Sci U S A 2019; 116:4044-4053. [PMID: 30760598 DOI: 10.1073/pnas.1816792116] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
While magnetic nanoparticles offer exciting possibilities for stem cell imaging or tissue bioengineering, their long-term intracellular fate remains to be fully documented. Besides, it appears that magnetic nanoparticles can occur naturally in human cells, but their origin and potentially endogenous synthesis still need further understanding. In an effort to explore the life cycle of magnetic nanoparticles, we investigated their transformations upon internalization in mesenchymal stem cells and as a function of the cells' differentiation status (undifferentiated, or undergoing adipogenesis, osteogenesis, and chondrogenesis). Using magnetism as a fingerprint of the transformation process, we evidenced an important degradation of the nanoparticles during chondrogenesis. For the other pathways, stem cells were remarkably "remagnetized" after degradation of nanoparticles. This remagnetization phenomenon is the direct demonstration of a possible neosynthesis of magnetic nanoparticles in cellulo and could lay some foundation to understand the presence of magnetic crystals in human cells. The neosynthesis was shown to take place within the endosomes and to involve the H-subunit of ferritin. Moreover, it appeared to be the key process to avoid long-term cytotoxicity (impact on differentiation) related to high doses of magnetic nanoparticles within stem cells.
Collapse
|
48
|
Guo R, Li Q, Yang F, Hu X, Jiao J, Guo Y, Wang J, Zhang Y. In Vivo MR Imaging of Dual MRI Reporter Genes and Deltex-1 Gene-modified Human Mesenchymal Stem Cells in the Treatment of Closed Penile Fracture. Mol Imaging Biol 2019; 20:417-427. [PMID: 28971290 DOI: 10.1007/s11307-017-1128-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE The purpose of this study was to investigate the feasibility of dual magnetic resonance imaging (MRI) reporter genes, including ferritin heavy subunit (Fth) and transferrin receptor (TfR), which provide sufficient MRI contrast for in vivo MRI tracking, and the Deltex-1 (DTX1) gene, which promotes human mesenchymal stem cell (hMSC) differentiation to smooth muscle cells (SMCs), to treat closed penile fracture (CPF). METHODS Multi-gene co-expressing hMSCs were generated. The expression of mRNA and proteins was assessed, and the original biological properties of hMSCs were determined and compared. The intracellular uptake of iron was evaluated, and the ability to differentiate into SMCs was detected. Fifty rabbits with CPF were randomly transplanted with PBS, hMSCs, Fth-TfR-hMSCs, DTX1-hMSCs, and Fth-TfR-DTX1-hMSCs. In vivo MRI was performed to detect the distribution and migration of the grafted cells and healing progress of CPF, and the results were correlated with histology. RESULTS The mRNA and proteins of the multi-gene were highly expressed. The transgenes could not influence the original biological properties of hMSCs. The dual MRI reporter genes increased the iron accumulation capacity, and the DTX1 gene promoted hMSC differentiation into SMCs. The distribution and migration of the dual MRI reporter gene-modified hMSCs, and the healing state of CPF could be obviously detected by MRI and confirmed by histology. CONCLUSION The dual MRI reporter genes could provide sufficient MRI contrast, and the distribution and migration of MSCs could be detected in vivo. The DTX1 gene can promote MSC differentiation into SMCs for the treatment of CPF and effectively inhibit granulation tissue formation.
Collapse
Affiliation(s)
- Ruomi Guo
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qingling Li
- Department of VIP Medical Center, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fei Yang
- Department of Urology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaojun Hu
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ju Jiao
- Department of Nuclear Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yu Guo
- Department of VIP Medical Center, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jin Wang
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yong Zhang
- Department of Nuclear Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
49
|
Markides H, Newell KJ, Rudorf H, Ferreras LB, Dixon JE, Morris RH, Graves M, Kaggie J, Henson F, El Haj AJ. Ex vivo MRI cell tracking of autologous mesenchymal stromal cells in an ovine osteochondral defect model. Stem Cell Res Ther 2019; 10:25. [PMID: 30635066 PMCID: PMC6330448 DOI: 10.1186/s13287-018-1123-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/04/2018] [Accepted: 12/25/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Osteochondral injuries represent a significant clinical problem requiring novel cell-based therapies to restore function of the damaged joint with the use of mesenchymal stromal cells (MSCs) leading research efforts. Pre-clinical studies are fundamental in translating such therapies; however, technologies to minimally invasively assess in vivo cell fate are currently limited. We investigate the potential of a MRI- (magnetic resonance imaging) and superparamagnetic iron oxide nanoparticle (SPION)-based technique to monitor cellular bio-distribution in an ovine osteochondral model of acute and chronic injuries. METHODS MSCs were isolated, expanded and labelled with Nanomag, a 250-nm SPION, and using a novel cell-penetrating technique, glycosaminoglycan-binding enhanced transduction (GET). MRI visibility thresholds, cellular toxicity and differentiation potential post-labelling were assessed in vitro. A single osteochondral defect was created in the medial femoral condyle in the left knee joint of each sheep with the contralateral joint serving as the control. Cells, either GET-Nanomag labelled or unlabelled, were delivered 1 week or 4.5 weeks later. Sheep were sacrificed 7 days post implantation and immediately MR imaged using a 0.2-T MRI scanner and validated on a 3-T MRI scanner prior to histological evaluation. RESULTS MRI data demonstrated a significant increase in MRI contrast as a result of GET-Nanomag labelling whilst cell viability, proliferation and differentiation capabilities were not affected. MRI results revealed evidence of implanted cells within the synovial joint of the injured leg of the chronic model only with no signs of cell localisation to the defect site in either model. This was validated histologically determining the location of implanted cells in the synovium. Evidence of engulfment of Nanomag-labelled cells by leukocytes is observed in the injured legs of the chronic model only. Finally, serum c-reactive protein (CRP) levels were measured by ELISA with no obvious increase in CRP levels observed as a result of P21-8R:Nanomag delivery. CONCLUSION This study has the potential to be a powerful translational tool with great implications in the clinical translation of stem cell-based therapies. Further, we have demonstrated the ability to obtain information linked to key biological events occurring post implantation, essential in designing therapies and selecting pre-clinical models.
Collapse
Affiliation(s)
- Hareklea Markides
- Institute of Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent, ST4 7QB UK
- Department of Chemical Engineering, Healthcare Technologies Institute, Birmingham University, B15 2TT, Birmingham, UK
| | - Karin J. Newell
- Department of Surgery, University of Cambridge, Addenbrooke’s Hospital, Hills Road Cambridge, Cambridge, CB2 0QQ UK
| | - Heike Rudorf
- Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge, CB3 0ES UK
| | - Lia Blokpoel Ferreras
- Centre for Biomolecular Sciences, The University of Nottingham, University Park, Nottingham, NG7 2RD UK
| | - James E. Dixon
- Centre for Biomolecular Sciences, The University of Nottingham, University Park, Nottingham, NG7 2RD UK
- School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, NG11 8NF UK
| | - Robert H. Morris
- School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, NG11 8NF UK
- Department of Radiology, University of Cambridge, Hills Rd, Cambridge, CB2 0QQ UK
| | - Martin Graves
- Department of Radiology, University of Cambridge, Hills Rd, Cambridge, CB2 0QQ UK
| | - Joshua Kaggie
- Department of Radiology, University of Cambridge, Hills Rd, Cambridge, CB2 0QQ UK
| | - Frances Henson
- Department of Surgery, University of Cambridge, Addenbrooke’s Hospital, Hills Road Cambridge, Cambridge, CB2 0QQ UK
- Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge, CB3 0ES UK
| | - Alicia J. El Haj
- Institute of Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent, ST4 7QB UK
- Department of Chemical Engineering, Healthcare Technologies Institute, Birmingham University, B15 2TT, Birmingham, UK
| |
Collapse
|
50
|
Yaman S, Anil-Inevi M, Ozcivici E, Tekin HC. Magnetic Force-Based Microfluidic Techniques for Cellular and Tissue Bioengineering. Front Bioeng Biotechnol 2018; 6:192. [PMID: 30619842 PMCID: PMC6305723 DOI: 10.3389/fbioe.2018.00192] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/23/2018] [Indexed: 01/21/2023] Open
Abstract
Live cell manipulation is an important biotechnological tool for cellular and tissue level bioengineering applications due to its capacity for guiding cells for separation, isolation, concentration, and patterning. Magnetic force-based cell manipulation methods offer several advantages, such as low adverse effects on cell viability and low interference with the cellular environment. Furthermore, magnetic-based operations can be readily combined with microfluidic principles by precisely allowing control over the spatiotemporal distribution of physical and chemical factors for cell manipulation. In this review, we present recent applications of magnetic force-based cell manipulation in cellular and tissue bioengineering with an emphasis on applications with microfluidic components. Following an introduction of the theoretical background of magnetic manipulation, components of magnetic force-based cell manipulation systems are described. Thereafter, different applications, including separation of certain cell fractions, enrichment of rare cells, and guidance of cells into specific macro- or micro-arrangements to mimic natural cell organization and function, are explained. Finally, we discuss the current challenges and limitations of magnetic cell manipulation technologies in microfluidic devices with an outlook on future developments in the field.
Collapse
|