1
|
Alizadeh R, Asghari A, Taghizadeh-Hesary F, Moradi S, Farhadi M, Mehdizadeh M, Simorgh S, Nourazarian A, Shademan B, Susanabadi A, Kamrava K. Intranasal delivery of stem cells labeled by nanoparticles in neurodegenerative disorders: Challenges and opportunities. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1915. [PMID: 37414546 DOI: 10.1002/wnan.1915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 05/05/2023] [Accepted: 06/11/2023] [Indexed: 07/08/2023]
Abstract
Neurodegenerative disorders occur through progressive loss of function or structure of neurons, with loss of sensation and cognition values. The lack of successful therapeutic approaches to solve neurologic disorders causes physical disability and paralysis and has a significant socioeconomic impact on patients. In recent years, nanocarriers and stem cells have attracted tremendous attention as a reliable approach to treating neurodegenerative disorders. In this regard, nanoparticle-based labeling combined with imaging technologies has enabled researchers to survey transplanted stem cells and fully understand their fate by monitoring their survival, migration, and differentiation. For the practical implementation of stem cell therapies in the clinical setting, it is necessary to accurately label and follow stem cells after administration. Several approaches to labeling and tracking stem cells using nanotechnology have been proposed as potential treatment strategies for neurological diseases. Considering the limitations of intravenous or direct stem cell administration, intranasal delivery of nanoparticle-labeled stem cells in neurological disorders is a new method of delivering stem cells to the central nervous system (CNS). This review describes the challenges and limitations of stem cell-based nanotechnology methods for labeling/tracking, intranasal delivery of cells, and cell fate regulation as theragnostic labeling. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Rafieh Alizadeh
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alimohamad Asghari
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Salah Moradi
- Department of Life Science Engineering, Faculty of New Science and Technology, University of Tehran, Tehran, Iran
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Susanabadi
- Department of Anesthesia and Pain Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Kamran Kamrava
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Gamberoni F, Borgese M, Pagiatakis C, Armenia I, Grazù V, Gornati R, Serio S, Papait R, Bernardini G. Iron Oxide Nanoparticles with and without Cobalt Functionalization Provoke Changes in the Transcription Profile via Epigenetic Modulation of Enhancer Activity. NANO LETTERS 2023; 23:9151-9159. [PMID: 37494138 PMCID: PMC10571150 DOI: 10.1021/acs.nanolett.3c01967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/22/2023] [Indexed: 07/28/2023]
Abstract
Despite the progress in the field of nanotoxicology, much about the cellular mechanisms that mediate the adverse effects of nanoparticles (NPs) and, in particular, the possible role of epigenetics in nanotoxicity, remains to be clarified. Therefore, we studied the changes occurring in the genome-wide distribution of H3K27ac, H3K4me1, H3K9me2, and H3K27me3 histone modifications and compared them with the transcriptome after exposing NIH3T3 cells to iron-based magnetic NPs (i.e., Fe2O3 and Fe2O3@Co NPs). We found that the transcription response is mainly due to changes in the genomic distribution of H3K27ac that can modulate the activity of enhancers. We propose that alteration of the epigenetic landscape is a key mechanism in defining the gene expression program changes resulting in nanotoxicity. With this approach, it is possible to construct a data set of genomic regions that could be useful for defining toxicity in a manner that is more comprehensive than what is possible with the present toxicology assays.
Collapse
Affiliation(s)
- Federica Gamberoni
- Department
of Biotechnology and Life Sciences, University
of Insubria, via J.H. Dunant 3, 21100 Varese, Italy
| | - Marina Borgese
- Department
of Medicine and Surgery, University of Insubria, via Guicciardini 9, 21100 Varese, Italy
| | - Christina Pagiatakis
- Department
of Biotechnology and Life Sciences, University
of Insubria, via J.H. Dunant 3, 21100 Varese, Italy
- IRCCS
Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Ilaria Armenia
- BioNanoSurf
Group, Instituto de Nanociencia y Materiales
de Aragón (INMA, CSIC-UNIZAR), Edificio I + D, 50018 Zaragoza, Spain
| | - Valeria Grazù
- BioNanoSurf
Group, Instituto de Nanociencia y Materiales
de Aragón (INMA, CSIC-UNIZAR), Edificio I + D, 50018 Zaragoza, Spain
| | - Rosalba Gornati
- Department
of Biotechnology and Life Sciences, University
of Insubria, via J.H. Dunant 3, 21100 Varese, Italy
| | - Simone Serio
- Department
of Biotechnology and Life Sciences, University
of Insubria, via J.H. Dunant 3, 21100 Varese, Italy
- IRCCS
Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department
of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, 20072 Pieve Emanuele, MI, Italy
| | - Roberto Papait
- Department
of Biotechnology and Life Sciences, University
of Insubria, via J.H. Dunant 3, 21100 Varese, Italy
- IRCCS
Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giovanni Bernardini
- Department
of Biotechnology and Life Sciences, University
of Insubria, via J.H. Dunant 3, 21100 Varese, Italy
| |
Collapse
|
3
|
Singh S, Jaiswal V, Singh JK, Semwal R, Raina D. Nanoparticle formulations: A smart era of advanced treatment with nanotoxicological imprints on the human body. Chem Biol Interact 2023; 373:110355. [PMID: 36682480 DOI: 10.1016/j.cbi.2023.110355] [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: 08/03/2022] [Revised: 12/21/2022] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
In the modern era, nanoparticles are the preferred dosage form, and maximum research is going on in the field of nanoparticle formulations. But as they are so small, nanoparticles are able to slip through the body's defenses and cause damage to the organs and tissues deep inside. In recent years, most researchers have focused solely on the therapeutic value of drugs or, at times, the performance of dosage forms, but few have given toxicity studies equal weight in their research. This review demonstrates that nanoparticle formulations are not suitable from a safety standpoint. So, researchers should be focused on alternative formulations like nanoemulsion, nanogel, and other liquids as well as semisolid formulations.
Collapse
Affiliation(s)
- Siddharth Singh
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun, 248007, India
| | - Vishakha Jaiswal
- Faculty of Pharmacy, BBDNIIT, Lucknow, Uttar Pradesh, 226028, India
| | | | - Ravindra Semwal
- Research and Development Centre, Faculty of Biomedical Sciences, Uttarakhand Ayurved University, Harrawala, Dehradun, 248001, India
| | - Deepika Raina
- School of Pharmacy, Graphic Era Hill University, Dehradun, India.
| |
Collapse
|
4
|
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
|
5
|
Gawne P, Man F, Blower PJ, T. M. de Rosales R. Direct Cell Radiolabeling for in Vivo Cell Tracking with PET and SPECT Imaging. Chem Rev 2022; 122:10266-10318. [PMID: 35549242 PMCID: PMC9185691 DOI: 10.1021/acs.chemrev.1c00767] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Indexed: 02/07/2023]
Abstract
The arrival of cell-based therapies is a revolution in medicine. However, its safe clinical application in a rational manner depends on reliable, clinically applicable methods for determining the fate and trafficking of therapeutic cells in vivo using medical imaging techniques─known as in vivo cell tracking. Radionuclide imaging using single photon emission computed tomography (SPECT) or positron emission tomography (PET) has several advantages over other imaging modalities for cell tracking because of its high sensitivity (requiring low amounts of probe per cell for imaging) and whole-body quantitative imaging capability using clinically available scanners. For cell tracking with radionuclides, ex vivo direct cell radiolabeling, that is, radiolabeling cells before their administration, is the simplest and most robust method, allowing labeling of any cell type without the need for genetic modification. This Review covers the development and application of direct cell radiolabeling probes utilizing a variety of chemical approaches: organic and inorganic/coordination (radio)chemistry, nanomaterials, and biochemistry. We describe the key early developments and the most recent advances in the field, identifying advantages and disadvantages of the different approaches and informing future development and choice of methods for clinical and preclinical application.
Collapse
Affiliation(s)
- Peter
J. Gawne
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Francis Man
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
- Institute
of Pharmaceutical Science, School of Cancer
and Pharmaceutical Sciences, King’s College London, London, SE1 9NH, U.K.
| | - Philip J. Blower
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Rafael T. M. de Rosales
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| |
Collapse
|
6
|
Al-Zoubi MS, Al-Zoubi RM. Nanomedicine Tactics in Cancer Treatment: Challenge and Hope. Crit Rev Oncol Hematol 2022; 174:103677. [PMID: 35385774 DOI: 10.1016/j.critrevonc.2022.103677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 10/18/2022] Open
|
7
|
Talib WH, Alsalahat I, Daoud S, Abutayeh RF, Mahmod AI. Plant-Derived Natural Products in Cancer Research: Extraction, Mechanism of Action, and Drug Formulation. Molecules 2020; 25:E5319. [PMID: 33202681 PMCID: PMC7696819 DOI: 10.3390/molecules25225319] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the main causes of death globally and considered as a major challenge for the public health system. The high toxicity and the lack of selectivity of conventional anticancer therapies make the search for alternative treatments a priority. In this review, we describe the main plant-derived natural products used as anticancer agents. Natural sources, extraction methods, anticancer mechanisms, clinical studies, and pharmaceutical formulation are discussed in this review. Studies covered by this review should provide a solid foundation for researchers and physicians to enhance basic and clinical research on developing alternative anticancer therapies.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan;
| | - Izzeddin Alsalahat
- Department of Pharmaceutical Chemistry and Pharmacognosy, Applied Science Private University, Amman 11931, Jordan; (I.A.); (S.D.); (R.F.A.)
| | - Safa Daoud
- Department of Pharmaceutical Chemistry and Pharmacognosy, Applied Science Private University, Amman 11931, Jordan; (I.A.); (S.D.); (R.F.A.)
| | - Reem Fawaz Abutayeh
- Department of Pharmaceutical Chemistry and Pharmacognosy, Applied Science Private University, Amman 11931, Jordan; (I.A.); (S.D.); (R.F.A.)
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan;
| |
Collapse
|
8
|
Shakil MS, Hasan MA, Sarker SR. Iron Oxide Nanoparticles for Breast Cancer Theranostics. Curr Drug Metab 2020; 20:446-456. [PMID: 30465497 DOI: 10.2174/1389200220666181122105043] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Breast cancer is the second leading cause of death in women worldwide. The extremely fast rate of metastasis and ability to develop resistance mechanism to all the conventional drugs make them very difficult to treat which are the causes of high morbidity and mortality of breast cancer patients. Scientists throughout the world have been focusing on the early detection of breast tumor so that treatment can be started at the very early stage. Moreover, conventional treatment processes such as chemotherapy, radiotherapy, and local surgery suffer from various limitations including toxicity, genetic mutation of normal cells, and spreading of cancer cells to healthy tissues. Therefore, new treatment regimens with minimum toxicity to normal cells need to be urgently developed. METHODS Iron oxide nanoparticles have been widely used for targeting hyperthermia and imaging of breast cancer cells. They can be conjugated with drugs, proteins, enzymes, antibodies or nucleotides to deliver them to target organs, tissues or tumors using external magnetic field. RESULTS Iron oxide nanoparticles have been successfully used as theranostic agents for breast cancer both in vitro and in vivo. Furthermore, their functionalization with drugs or functional biomolecules enhance their drug delivery efficiency and reduces the systemic toxicity of drugs. CONCLUSION This review mainly focuses on the versatile applications of superparamagnetic iron oxide nanoparticles on the diagnosis, treatment, and detecting progress of breast cancer treatment. Their wide application is because of their excellent superparamagnetic, biocompatible and biodegradable properties.
Collapse
Affiliation(s)
- Md Salman Shakil
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Md Ashraful Hasan
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Satya Ranjan Sarker
- Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| |
Collapse
|
9
|
Ardelean IL, Ficai D, Sonmez M, Oprea O, Nechifor G, Andronescu E, Ficai A, Titu MA. Hybrid Magnetic Nanostructures For Cancer Diagnosis And Therapy. Anticancer Agents Med Chem 2019; 19:6-16. [PMID: 30411694 DOI: 10.2174/1871520618666181109112655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 03/17/2018] [Accepted: 10/23/2018] [Indexed: 12/24/2022]
Abstract
Cancer is the second disease in the world from the point of view of mortality. The conventional routes of treatment were found to be not sufficient and thus alternative ways are imposed. The use of hybrid, magnetic nanostructures is a promising way for simultaneous targeted diagnosis and treatment of various types of cancer. For this reason, the development of core@shell structures was found to be an efficient way to develop stable, biocompatible, non-toxic carriers with shell-dependent internalization capacity in cancer cells. So, the multicomponent approach can be the most suitable way to assure the multifunctionality of these nanostructures to achieve the desired/necessary properties. The in vivo stability is mostly assured by the coating of the magnetic core with various polymers (including polyethylene glycol, silica etc.), while the targeting capacity is mostly assured by the decoration of these nanostructures with folic acid. Unfortunately, there are also some limitations related to the multilayered approach. For instance, the increasing of the thickness of layers leads to a decrease the magnetic properties, (hyperthermia and guiding ability in the magnetic field, for instance), the outer shell should contain the targeting molecules (as well as the agents helping the internalization into the cancer cells), etc.
Collapse
Affiliation(s)
- Ioana L Ardelean
- University POLITEHNICA of Bucharest, Faculty of Applied Chemistry and Material Science; 1-7 Polizu Str., 011061 Bucharest, Romania
| | - Denisa Ficai
- University POLITEHNICA of Bucharest, Faculty of Applied Chemistry and Material Science; 1-7 Polizu Str., 011061 Bucharest, Romania
| | - Maria Sonmez
- Leather and Footwear Research Institute, Department of Rubber, 93 Ion Minulescu street, 031215, Bucharest, Romania
| | - Ovidiu Oprea
- University POLITEHNICA of Bucharest, Faculty of Applied Chemistry and Material Science; 1-7 Polizu Str., 011061 Bucharest, Romania
| | - Gheorghe Nechifor
- University POLITEHNICA of Bucharest, Faculty of Applied Chemistry and Material Science; 1-7 Polizu Str., 011061 Bucharest, Romania
| | - Ecaterina Andronescu
- University POLITEHNICA of Bucharest, Faculty of Applied Chemistry and Material Science; 1-7 Polizu Str., 011061 Bucharest, Romania
| | - Anton Ficai
- University POLITEHNICA of Bucharest, Faculty of Applied Chemistry and Material Science; 1-7 Polizu Str., 011061 Bucharest, Romania
| | - Mihail A Titu
- "Lucian Blaga" University of Sibiu, Faculty of Engineering, Industrial Engineering and Management Departament, Sibiu, Romania
| |
Collapse
|
10
|
Pandey S, Mishra A. Rational approaches for toxicological assessments of nanobiomaterials. J Biochem Mol Toxicol 2019; 33:e22335. [DOI: 10.1002/jbt.22335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/09/2019] [Accepted: 03/15/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Shalabh Pandey
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER‐R)Lucknow Uttar Pradesh India
| | - Awanish Mishra
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER‐R)Lucknow Uttar Pradesh India
| |
Collapse
|
11
|
Zhao Z, Ukidve A, Krishnan V, Mitragotri S. Effect of physicochemical and surface properties on in vivo fate of drug nanocarriers. Adv Drug Deliv Rev 2019; 143:3-21. [PMID: 30639257 DOI: 10.1016/j.addr.2019.01.002] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/07/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023]
Abstract
Over the years, a plethora of materials - natural and synthetic - have been engineered at a nanoscopic level and explored for drug delivery. Nanocarriers based on such materials could improve the payload's pharmacokinetics and achieve the desired pharmacological response at the target tissue. Despite the development of rationally designed drug nanocarriers, only a handful of such formulations have been successfully translated into the clinic. The physicochemical properties (size, shape, surface chemistry, porosity, elasticity, and many others) of these nanocarriers influence its biological identity, which in presence of biological barriers in vivo, could significantly modulate the therapeutic index of its cargo and alter the desired outcome. Further, complexities associated with developing effective drug nanocarriers have led to conflicting views of its safety, permeation of biological barriers and cellular uptake. Here, in this review, we emphasize the effect of physicochemical properties of nanocarriers on their interactions with the biological milieu. The review will discuss in depth, how modulating the physicochemical properties would influence a drug nanocarrier's behavior in vivo and the mechanisms underlying these effects. The goal of this review is to summarize the design considerations based on these properties and to provide a conceptual template for achieving improved therapeutic efficacy with enhanced patient compliance.
Collapse
|
12
|
Patil RM, Thorat ND, Shete PB, Bedge PA, Gavde S, Joshi MG, Tofail SA, Bohara RA. Comprehensive cytotoxicity studies of superparamagnetic iron oxide nanoparticles. Biochem Biophys Rep 2018; 13:63-72. [PMID: 29349357 PMCID: PMC5766481 DOI: 10.1016/j.bbrep.2017.12.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 11/20/2022] Open
Abstract
Recently lots of efforts have been taken to develop superparamagnetic iron oxide nanoparticles (SPIONs) for biomedical applications. So it is utmost necessary to have in depth knowledge of the toxicity occurred by this material. This article is designed in such way that it covers all the associated toxicity issues of SPIONs. It mainly emphasis on toxicity occurred at different levels including cellular alterations in the form of damage to nucleic acids due to oxidative stress and altered cellular response. In addition focus is been devoted for in vitro and in vivo toxicity of SPIONs, so that a better therapeutics can be designed. At the end the time dependent nature of toxicity and its ultimate faith inside the body is being discussed.
Collapse
Affiliation(s)
- Rakesh M. Patil
- Directorate of Forensic Science Laboratory, Govt. of Maharashtra Kalina, Mumbai, India
- Centre for Interdisciplinary Research, D.Y.Patil University, Kolhapur, India
| | - Nanasaheb D. Thorat
- Material and Surface Science Institute, Bernal Institute, University of Limerick, Ireland
| | - Prajkta B. Shete
- Centre for Interdisciplinary Research, D.Y.Patil University, Kolhapur, India
| | - Poonam A. Bedge
- Department of Stem Cells and Regenerative Medicine, D.Y.Patil University, Kolhapur, India
| | - Shambala Gavde
- Centre for Interdisciplinary Research, D.Y.Patil University, Kolhapur, India
| | - Meghnad G. Joshi
- Department of Stem Cells and Regenerative Medicine, D.Y.Patil University, Kolhapur, India
| | - Syed A.M. Tofail
- Material and Surface Science Institute, Bernal Institute, University of Limerick, Ireland
| | - Raghvendra A. Bohara
- Centre for Interdisciplinary Research, D.Y.Patil University, Kolhapur, India
- Department of Stem Cells and Regenerative Medicine, D.Y.Patil University, Kolhapur, India
- Research and Innovations for Comprehensive Health (RICH), Cell D.Y.Patil University, Kolhapur, India
| |
Collapse
|
13
|
Namestnikova D, Gubskiy I, Kholodenko I, Melnikov P, Sukhinich K, Gabashvili A, Vishnevskiy D, Soloveva A, Abakumov M, Vakhrushev I, Lupatov A, Chekhonin V, Gubsky L, Yarygin K. Methodological aspects of MRI of transplanted superparamagnetic iron oxide-labeled mesenchymal stem cells in live rat brain. PLoS One 2017; 12:e0186717. [PMID: 29049361 PMCID: PMC5648235 DOI: 10.1371/journal.pone.0186717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 10/08/2017] [Indexed: 12/22/2022] Open
Abstract
In vivo tracking of transplanted mesenchymal stem cells (MSCs) migration and homing is vital for understanding the mechanisms of beneficial effects of MSCs transplantation in animal models of diseases and in clinical trials. Transplanted cells can be labeled with superparamagnetic iron oxide (SPIO) particles and visualized in vivo using a number of iron sensitive MRI techniques. However, the applicability of those techniques for SPIO-labeled MSCs tracking in live brain has not been sufficiently investigated. The goal of this study was to estimate the efficiency of various MRI techniques of SPIO-labeled cell tracing in the brain. To achieve that goal, the precision and specificity of T2WI, T2*WI and SWI (Susceptibility-Weighted Imaging) techniques of SPIO-labeled MSCs tracing in vitro and in live rat brain were for the first time compared in the same experiment. We have shown that SWI presents the most sensitive pulse sequence for SPIO-labeled MSCs MR visualization. After intracerebral administration due to limitations caused by local micro-hemorrhages the visualization threshold was 102 cells, while after intra-arterial transplantation SWI permitted detection of several cells or even single cells. There is just one publication claiming detection of individual SPIO-labeled MSCs in live brain, while the other state much lower sensitivity, describe detection of different cell types or high resolution tracing of MSCs in other tissues. This study confirms the possibility of single cell tracing in live brain and outlines the necessary conditions. SWI is a method convenient for the detection of single SPIO labeled MSCs and small groups of SPIO labeled MSCs in brain tissue and can be appropriate for monitoring migration and homing of transplanted cells in basic and translational neuroscience.
Collapse
Affiliation(s)
| | - Ilya Gubskiy
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Pavel Melnikov
- Pirogov Russian National Research Medical University, Moscow, Russia.,Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | | | | | | | | | - Maxim Abakumov
- Pirogov Russian National Research Medical University, Moscow, Russia.,National University of Science and Technology, Moscow, Russia
| | | | | | - Vladimir Chekhonin
- Pirogov Russian National Research Medical University, Moscow, Russia.,Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Leonid Gubsky
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | |
Collapse
|
14
|
Li G, Bonamici N, Dey M, Lesniak MS, Balyasnikova IV. Intranasal delivery of stem cell-based therapies for the treatment of brain malignancies. Expert Opin Drug Deliv 2017; 15:163-172. [PMID: 28895435 DOI: 10.1080/17425247.2018.1378642] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most aggressive malignant brain cancer in adults, and its poor prognosis and resistance to the existing standard of care require the development of innovative therapeutic modalities. The local delivery of stem cells as therapeutic carriers against glioma has produced encouraging results, but encounters obstacles with regards to the repeatability and invasiveness of administration. Intranasal delivery of therapeutic stem cells could overcome these obstacles, among others, as a noninvasive and easily repeatable mode of administration. AREAS COVERED This review describes nasal anatomy, routes of stem cell migration, and factors affecting stem cell delivery to hard-to-reach tumors. Furthermore, this review discusses the molecular mechanisms underlying stem cell migration following delivery, as well as possible stem cell effector functions to be considered in combination with intranasal delivery. EXPERT OPINION Further research is necessary to elucidate the dynamics of stem cell effector functions in the context of intranasal delivery and optimize their therapeutic potency. Nonetheless, the technique represents a promising tool against brain cancer and has the potential to be expanded for use against other brain pathologies.
Collapse
Affiliation(s)
- Gina Li
- a Department of Neurological Surgery , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Nicolas Bonamici
- a Department of Neurological Surgery , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Mahua Dey
- b Department of Neurological Surgery , Indiana University , Indianapolis , IN , USA
| | - Maciej S Lesniak
- a Department of Neurological Surgery , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Irina V Balyasnikova
- a Department of Neurological Surgery , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| |
Collapse
|
15
|
Kavithaa K, Sumathi S, Padma PR. Intracellular Uptake of PEG-Funtionalized Baicalein Loaded Iron Oxide Nanoparticles Regulates Apoptotic Genes in Triple Negative Breast Cancer Cells: Mitochondrial Pathway Targeted Therapy for Breast Cancer. J CLUST SCI 2017. [DOI: 10.1007/s10876-017-1204-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
16
|
Mou Y, Lv S, Xiong F, Han Y, Zhao Y, Li J, Gu N, Zhou J. Effects of different doses of 2,3-dimercaptosuccinic acid-modified Fe 2 O 3 nanoparticles on intercalated discs in engineered cardiac tissues. J Biomed Mater Res B Appl Biomater 2016; 106:121-130. [PMID: 27889952 DOI: 10.1002/jbm.b.33757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/27/2016] [Accepted: 07/05/2016] [Indexed: 12/11/2022]
Abstract
Although iron oxide nanoparticles (IRONs) were applied in clinical magnetic resonance imaging in vivo and magnetic tissue engineering in vitro widely, the underlying effects of IRONs on the development of cardiomyocytes especially the intercellular junctions, intercalated discs (IDs), remain an unknown issue. Given the critical role of three-dimensional (3D) engineered cardiac tissues (ECTs) in evaluation of nanoparticles toxicology, it remained necessary to understand the effects of IRONs on IDs assembly of cardiomyocytes in 3D environment. In this study, we first reconstituted collagen/Matrigel based ECTs in vitro and prepared IRONs with 2,3-dimercaptosuccinic acid (DMSA-IRONs). We found that the internalization of DMSA-IRONs by cardiac cells in dose-dependent manner was not associated with the cell distribution in 3D environment by determination of Prussian blue staining and transmission electronic microscopy. Significantly, through determination of western blotting and immunofluorescence of connexin 43, N-cadherin, desmoplakin, and plakoglobin, DMSA-IRONs enhanced the assembly of gap junctions, decreased mechanical junctions (adherens junctions and desmosomes) of cardiac cells but not in dose-dependent manner in ECTs at seventh day. In addition, DMSA-IRONs increased the vesicles secretion of cardiac cells in ECTs apparently compared to control groups. Overall, we conclude that the internalization of DMSA-IRONs by cardiac cells in dose-dependent manner enhanced the assembly of electrochemical junctions and decreased the mechanical related microstructures. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 121-130, 2018.
Collapse
Affiliation(s)
- Yongchao Mou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China.,School of Life Science and Technology, Harbin Institute of Technology, Harbin, People's Republic of China
| | - Shuanghong Lv
- Laboratory of Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Fei Xiong
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 People's Republic of China
| | - Yao Han
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Yuwei Zhao
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Junjie Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Ning Gu
- Laboratory of Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Jin Zhou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
17
|
Zhang Z, Li M, Chen F, Li L, Liu J, Li Z, Ji R, Zuo X, Li Y. Probe-Based Confocal Laser Endomicroscopy for Imaging TRAIL-Expressing Mesenchymal Stem Cells to Monitor Colon Xenograft Tumors In Vivo. PLoS One 2016; 11:e0162700. [PMID: 27617958 PMCID: PMC5019474 DOI: 10.1371/journal.pone.0162700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 08/27/2016] [Indexed: 12/13/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) can serve as vehicles for therapeutic genes. However, little is known about MSC behavior in vivo. Here, we demonstrated that probe-based confocal laser endomicroscopy (pCLE) can be used to track MSCs in vivo and individually monitor tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) gene expression within carcinomas. Methods Isolated BALB/c nu/nu mice MSCs (MSCs) were characterized and engineered to co-express the TRAIL and enhanced green fluorescent protein (EGFP) genes. The number of MSCs co-expressing EGFP and TRAIL (TRAIL-MSCs) at tumor sites was quantified with pCLE in vivo, while their presence was confirmed using immunofluorescence (IF) and quantitative polymerase chain reaction (qPCR). The therapeutic effects of TRAIL-MSCs were evaluated by measuring the volumes and weights of subcutaneous HT29-derived xenograft tumors. Results Intravital imaging of the subcutaneous xenograft tumors revealed that BALB/c mice treated with TRAIL-MSCs exhibited specific cellular signals, whereas no specific signals were observed in the control mice. The findings from the pCLE images were consistent with the IF and qPCR results. Conclusion The pCLE results indicated that endomicroscopy could effectively quantify injected MSCs that homed to subcutaneous xenograft tumor sites in vivo and correlated well with the therapeutic effects of the TRAIL gene. By applying pCLE for the in vivo monitoring of cellular trafficking, stem cell-based anticancer gene therapeutic approaches might be feasible and attractive options for individualized clinical treatments.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Ming Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Feixue Chen
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Lixiang Li
- Laboratory of Translational Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Jun Liu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Zhen Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Rui Ji
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Xiuli Zuo
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
- * E-mail:
| |
Collapse
|
18
|
Naseroleslami M, Parivar K, Khoei S, Aboutaleb N. Magnetic Resonance Imaging of Human-Derived Amniotic Membrane Stem Cells Using PEGylated Superparamagnetic Iron Oxide Nanoparticles. CELL JOURNAL 2016; 18:332-9. [PMID: 27602314 PMCID: PMC5011320 DOI: 10.22074/cellj.2016.4560] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/03/2015] [Indexed: 11/20/2022]
Abstract
OBJECTIVE The label and detection of cells injected into target tissues is an area of focus for researchers. Iron oxide nanoparticles can be used to label cells as they have special characteristics. The purpose of this study is to examine the effects of iron oxide nanoparticles on human-derived amniotic membrane stem cell (hAMCs) survival and to investigate the magnetic properties of these nanoparticles with increased contrast in magnetic resonance imaging (MRI). MATERIALS AND METHODS In this experimental study, we initially isolated mesenchymal stem cells from amniotic membranes and analyzed them by flow cytometry. In addition, we synthesized superparamagnetic iron oxide nanoparticles (SPIONs) and characterized them by various methods. The SPIONs were incubated with hAMCs at concentrations of 25-800 μg/mL. The cytotoxicity of nanoparticles on hAMCs was measured by the MTT assay. Next, we evaluated the effectiveness of the magnetic nanoparticles as MRI contrast agents. Solutions of SPION were prepared in water at different iron concentrations for relaxivity measurements by a 1.5 Tesla clinical MRI instrument. RESULTS The isolated cells showed an adherent spindle shaped morphology. Polyethylene glycol (PEG)-coated SPIONs exhibited a spherical morphology. The average particle size was 20 nm and magnetic saturation was 60 emu/g. Data analysis showed no significant reduction in the percentage of viable cells (97.86 ± 0.41%) after 72 hours at the 125 μg/ml concentration compared with the control. The relaxometry results of this SPION showed a transverse relaxivity of 6.966 (μg/ml.s)(-1). CONCLUSION SPIONs coated with PEG used in this study at suitable concentrations had excellent labeling efficiency and biocompatibility for hAMCs.
Collapse
Affiliation(s)
- Maryam Naseroleslami
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Kazem Parivar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Samideh Khoei
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Aboutaleb
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Yuan H, Gomez JA, Chien JS, Zhang L, Wilson CM, Li S, Fales AM, Liu Y, Grant GA, Mirotsou M, Dzau VJ, Vo-Dinh T. Tracking mesenchymal stromal cells using an ultra-bright TAT-functionalized plasmonic-active nanoplatform. JOURNAL OF BIOPHOTONICS 2016; 9:406-413. [PMID: 27095616 PMCID: PMC5645019 DOI: 10.1002/jbio.201500173] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 08/11/2015] [Accepted: 08/24/2015] [Indexed: 06/05/2023]
Abstract
High-resolution tracking of stem cells remains a challenging task. An ultra-bright contrast agent with extended intracellular retention is suitable for in vivo high-resolution tracking of stem cells following the implantation. Here, a plasmonic-active nanoplatform was developed for tracking mesenchymal stromal cells (MSCs) in mice. The nanoplatform consisted of TAT peptide-functionalized gold nanostars (TAT-GNS) that emit ultra-bright two-photon photoluminescence capable of tracking MSCs under high-resolution optical imaging. In vitro experiment showed TAT-GNS-labeled MSCs retained a similar differentiability to that of non-labeled MSCs controls. Due to their star shape, TAT-GNS exhibited greater intracellular retention than that of commercial Q-Tracker. In vivo imaging of TAT-GNS-labeled MSCs five days following intra-arterial injections in mice kidneys showed possible MSCs implantation in juxta-glomerular (JG) regions, but non-specifically in glomeruli and afferent arterioles as well. With future design to optimize GNS labeling specificity and clearance, plasmonic-active nanoplatforms may be a useful intracellular tracking tool for stem cell research. An ultra-bright intracellular contrast agent is developed using TAT peptide-functionalized gold nanostars (TAT-GNS). It poses minimal influence on the stem cell differentiability. It exhibits stronger two-photon photoluminescence and superior labeling efficiency than commercial Q-Tracker. Following renal implantation, some TAT-GNS-labeled MSCs permeate blood vessels and migrate to the juxta-glomerular region.
Collapse
Affiliation(s)
- Hsiangkuo Yuan
- Department of Biomedical Engineering, Fitzpatrick Institute for Photonics, Duke University, Durham, NC 27708, USA
| | - Jose A Gomez
- Department of Medicine, Duke University Medical Center and Mandel Center for Hypertension and Atherosclerosis Research, Durham, NC 27710, USA
| | - Jennifer S Chien
- Department of Medicine, Duke University Medical Center and Mandel Center for Hypertension and Atherosclerosis Research, Durham, NC 27710, USA
| | - Lunan Zhang
- Department of Medicine, Duke University Medical Center and Mandel Center for Hypertension and Atherosclerosis Research, Durham, NC 27710, USA
| | - Christy M Wilson
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Shuqin Li
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Andrew M Fales
- Department of Biomedical Engineering, Fitzpatrick Institute for Photonics, Duke University, Durham, NC 27708, USA
| | - Yang Liu
- Department of Biomedical Engineering, Fitzpatrick Institute for Photonics, Duke University, Durham, NC 27708, USA
- Department of Chemistry, Duke University, NC 27708, USA
| | - Gerald A Grant
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Maria Mirotsou
- Department of Medicine, Duke University Medical Center and Mandel Center for Hypertension and Atherosclerosis Research, Durham, NC 27710, USA
| | - Victor J Dzau
- Department of Medicine, Duke University Medical Center and Mandel Center for Hypertension and Atherosclerosis Research, Durham, NC 27710, USA
| | - Tuan Vo-Dinh
- Department of Biomedical Engineering, Fitzpatrick Institute for Photonics, Duke University, Durham, NC 27708, USA.
- Department of Chemistry, Duke University, NC 27708, USA.
| |
Collapse
|
20
|
Cheng SH, Yu D, Tsai HM, Morshed RA, Kanojia D, Lo LW, Leoni L, Govind Y, Zhang L, Aboody KS, Lesniak MS, Chen CT, Balyasnikova IV. Dynamic In Vivo SPECT Imaging of Neural Stem Cells Functionalized with Radiolabeled Nanoparticles for Tracking of Glioblastoma. J Nucl Med 2015; 57:279-84. [PMID: 26564318 DOI: 10.2967/jnumed.115.163006] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/09/2015] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED There is strong clinical interest in using neural stem cells (NSCs) as carriers for targeted delivery of therapeutics to glioblastoma. Multimodal dynamic in vivo imaging of NSC behaviors in the brain is necessary for developing such tailored therapies; however, such technology is lacking. Here we report a novel strategy for mesoporous silica nanoparticle (MSN)-facilitated NSC tracking in the brain via SPECT. METHODS (111)In was conjugated to MSNs, taking advantage of the large surface area of their unique porous feature. A series of nanomaterial characterization assays was performed to assess the modified MSN. Loading efficiency and viability of NSCs with (111)In-MSN complex were optimized. Radiolabeled NSCs were administered to glioma-bearing mice via either intracranial or systemic injection. SPECT imaging and bioluminescence imaging were performed daily up to 48 h after NSC injection. Histology and immunocytochemistry were used to confirm the findings. RESULTS (111)In-MSN complexes show minimal toxicity to NSCs and robust in vitro and in vivo stability. Phantom studies demonstrate feasibility of this platform for NSC imaging. Of significance, we discovered that decayed (111)In-MSN complexes exhibit strong fluorescent profiles in preloaded NSCs, allowing for ex vivo validation of the in vivo data. In vivo, SPECT visualizes actively migrating NSCs toward glioma xenografts in real time after both intracranial and systemic administrations. This is in agreement with bioluminescence live imaging, confocal microscopy, and histology. CONCLUSION These advancements warrant further development and integration of this technology with MRI for multimodal noninvasive tracking of therapeutic NSCs toward various brain malignancies.
Collapse
Affiliation(s)
- Shih-Hsun Cheng
- Department of Radiology, The University of Chicago, Chicago, Illinois
| | - Dou Yu
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois
| | - Hsiu-Ming Tsai
- Department of Radiology, The University of Chicago, Chicago, Illinois
| | - Ramin A Morshed
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois
| | - Deepak Kanojia
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois
| | - Leu-Wei Lo
- Department of Radiology, The University of Chicago, Chicago, Illinois Institute of Biomedical Engineering and Nanomedicine, National Health Research Institute(s), Taiwan; and
| | - Lara Leoni
- Department of Radiology, The University of Chicago, Chicago, Illinois
| | - Yureve Govind
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois
| | - Lingjiao Zhang
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois
| | - Karen S Aboody
- Department of Neuroscience, City of Hope National Medical Center and Beckman Research Institute, Duarte, California
| | - Maciej S Lesniak
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois
| | - Chin-Tu Chen
- Department of Radiology, The University of Chicago, Chicago, Illinois
| | | |
Collapse
|
21
|
Connell JJ, Patrick PS, Yu Y, Lythgoe MF, Kalber TL. Advanced cell therapies: targeting, tracking and actuation of cells with magnetic particles. Regen Med 2015; 10:757-72. [PMID: 26390317 DOI: 10.2217/rme.15.36] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Regenerative medicine would greatly benefit from a new platform technology that enabled measurable, controllable and targeting of stem cells to a site of disease or injury in the body. Superparamagnetic iron-oxide nanoparticles offer attractive possibilities in biomedicine and can be incorporated into cells, affording a safe and reliable means of tagging. This review describes three current and emerging methods to enhance regenerative medicine using magnetic particles to guide therapeutic cells to a target organ; track the cells using MRI and assess their spatial localization with high precision and influence the behavior of the cell using magnetic actuation. This approach is complementary to the systemic injection of cell therapies, thus expanding the horizon of stem cell therapeutics.
Collapse
Affiliation(s)
- John J Connell
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - P Stephen Patrick
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Yichao Yu
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Mark F Lythgoe
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Tammy L Kalber
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| |
Collapse
|
22
|
Bahadar H, Maqbool F, Niaz K, Abdollahi M. Toxicity of Nanoparticles and an Overview of Current Experimental Models. IRANIAN BIOMEDICAL JOURNAL 2015; 20:1-11. [PMID: 26286636 PMCID: PMC4689276 DOI: 10.7508/ibj.2016.01.001] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Nanotechnology is a rapidly growing field having potential applications in many areas. Nanoparticles (NPs) have been studied for cell toxicity, immunotoxicity, and genotoxicity. Tetrazolium-based assays such as MTT, MTS, and WST-1 are used to determine cell viability. Cell inflammatory response induced by NPs is checked by measuring inflammatory biomarkers, such as IL-8, IL-6, and tumor necrosis factor, using ELISA. Lactate dehydrogenase (LDH) assay is used for cell membrane integrity. Different types of cell cultures, including cancer cell lines have been employed as in vitro toxicity models. It has been generally agreed that NPs interfere with either assay materials or with detection systems. So far, toxicity data generated by employing such models are conflicting and inconsistent. Therefore, on the basis of available experimental models, it may be difficult to judge and list some of the more valuable NPs as more toxic to biological systems and vice versa. Considering the potential applications of NPs in many fields and the growing apprehensions of FDA about the toxic potential of nanoproducts, it is the need of the hour to look for new internationally agreed free of bias toxicological models by focusing more on in vivo studies.
Collapse
Affiliation(s)
- Haji Bahadar
- Dept. of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Faheem Maqbool
- Dept. of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamal Niaz
- Dept. of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Dept. of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, International Campus, Tehran University of Medical Sciences, Tehran, Iran.,Endocrinology and Metabolism Research Center, Institute of Clinical Endocrine Sciences, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Shim J, Kwak BK, Jung J, Park S. Evaluation of engraftment of superparamagnetic iron oxide-labeled mesenchymal stem cells using three-dimensional reconstruction of magnetic resonance imaging in photothrombotic cerebral infarction models of rats. Korean J Radiol 2015; 16:575-85. [PMID: 25995687 PMCID: PMC4435988 DOI: 10.3348/kjr.2015.16.3.575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 02/21/2015] [Indexed: 12/28/2022] Open
Abstract
Objective To evaluate engraftment by visualizing the location of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) three-dimensionally in photothrombotic cerebral infarction (PTCI) models of rats. Materials and Methods Magnetic resonance imaging (MRI) of an agarose block containing superparamagnetic iron oxide (SPIO)-labeled hBM-MSCs was performed using a 3.0-T MRI, T2-(T2WI), T2*-(T2*WI), and susceptibility-weighted images (SWI). PTCI was induced in 6 rats, and 2.5 × 105 SPIO-labeled hBM-MSCs were infused through the ipsilateral internal carotid artery (ICA group) or tail vein (IV group). MRI was performed on days 1, 3, 7, and 14 after stem cell injection. Dark signal regions were confirmed using histology. Three-dimensional MRI reconstruction was performed using the clinical workflow solution to evaluate the engraftment of hBM-MSCs. Volumetric analysis of the engraftment was also performed. Results The volumes of SPIO-labeled hBM-MSCs in the phantom MRI were 129.3, 68.4, and 25.9 µL using SWI, T2*WI, and T2WI, respectively. SPIO-labeled hBM-MSCs appeared on day 1 after injection, encircling the cerebral infarction from the ventral side. Dark signal regions matched iron positive cells and human origin (positive) cells. The volume of the engraftment was larger in the ICA group on days 1, 3, and 7, after stem cell injection (p < 0.05 on SWI). SWI was the most sensitive MRI pulse sequence (p < 0.05). The volume of infarction decreased until day 14. Conclusion The engraftment of SPIO-labeled hBM-MSCs can be visualized and evaluated three-dimensionally in PTCI models of rats. The engraftment volume was larger in the ICA group than IV group on early stage within one week.
Collapse
Affiliation(s)
- Jaehyun Shim
- Department of Convergence Medicine and Pharmaceutical Biosciences, Chung-Ang University, Seoul 156-756, Korea
| | - Byung Kook Kwak
- Department of Radiology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 156-755, Korea
| | - Jisung Jung
- Department of Radiology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 156-755, Korea
| | - Serah Park
- Department of Convergence Medicine and Pharmaceutical Biosciences, Chung-Ang University, Seoul 156-756, Korea. ; Major of Biomedical Science, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| |
Collapse
|
24
|
Arutyunyan I, Elchaninov A, Fatkhudinov T, Makarov A, Kananykhina E, Usman N, Bolshakova G, Glinkina V, Goldshtein D, Sukhikh G. Elimination of allogeneic multipotent stromal cells by host macrophages in different models of regeneration. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:4469-80. [PMID: 26191137 PMCID: PMC4503009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/26/2015] [Indexed: 02/07/2023]
Abstract
Allogeneic multipotent stromal cells were previously thought to be poorly recognized by host immune system; the prolonged survival in host environments was explained by their immune privileged status. As long as the concept is currently reconsidered, the routes of elimination of allogeneic multipotent stromal cells by host immunity must be taken into account. This is necessary for correct comprehension of their therapeutic action. The study was focused upon survival of umbilical cord-derived allogeneic multipotent stromal cells in different rat models of tissue regeneration induced by partial hepatectomy or by critical limb ischemia. The observations were carried out by means of vital labeling of the cells with PKH26 prior to injection, in combination with differential immunostaining of host macrophages with anti-CD68 antibody. According to the results, allogeneic multipotent stromal cells are specifically eliminated by host immune system; the efficacy can reach 100%. Massive clearance of transplanted cells by host macrophages is accompanied by appropriation of the label by the latter, and this is a pronounced case of misleading presentation of exogenous label by host cells. The study emphasizes the role of macrophages in host response and also the need of additional criteria for correct data interpretation.
Collapse
Affiliation(s)
- Irina Arutyunyan
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of The Russian Federation4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology3 Tsurupa Street, Moscow 117418, Russia
| | - Andrey Elchaninov
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of The Russian Federation4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology3 Tsurupa Street, Moscow 117418, Russia
- Pirogov Russian National Research Medical University, Ministry of Healthcare of The Russian Federation1 Ostrovitianov Street, Moscow 117997, Russia
| | - Timur Fatkhudinov
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of The Russian Federation4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology3 Tsurupa Street, Moscow 117418, Russia
- Pirogov Russian National Research Medical University, Ministry of Healthcare of The Russian Federation1 Ostrovitianov Street, Moscow 117997, Russia
| | - Andrey Makarov
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of The Russian Federation4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology3 Tsurupa Street, Moscow 117418, Russia
- Pirogov Russian National Research Medical University, Ministry of Healthcare of The Russian Federation1 Ostrovitianov Street, Moscow 117997, Russia
| | - Evgeniya Kananykhina
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of The Russian Federation4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology3 Tsurupa Street, Moscow 117418, Russia
| | - Natalia Usman
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of The Russian Federation4 Oparina Street, Moscow 117997, Russia
- Scientific Research Institute of Human Morphology3 Tsurupa Street, Moscow 117418, Russia
- Pirogov Russian National Research Medical University, Ministry of Healthcare of The Russian Federation1 Ostrovitianov Street, Moscow 117997, Russia
| | - Galina Bolshakova
- Scientific Research Institute of Human Morphology3 Tsurupa Street, Moscow 117418, Russia
| | - Valeria Glinkina
- Pirogov Russian National Research Medical University, Ministry of Healthcare of The Russian Federation1 Ostrovitianov Street, Moscow 117997, Russia
| | - Dmitry Goldshtein
- Research Centre of Medical Genetics1 Moskvorechie Street, Moscow 115478, Russia
| | - Gennady Sukhikh
- Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of The Russian Federation4 Oparina Street, Moscow 117997, Russia
| |
Collapse
|
25
|
Ramos-Gómez M, Seiz EG, Martínez-Serrano A. Optimization of the magnetic labeling of human neural stem cells and MRI visualization in the hemiparkinsonian rat brain. J Nanobiotechnology 2015; 13:20. [PMID: 25890124 PMCID: PMC4416262 DOI: 10.1186/s12951-015-0078-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/02/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Magnetic resonance imaging is the ideal modality for non-invasive in vivo cell tracking allowing for longitudinal studies over time. Cells labeled with superparamagnetic iron oxide nanoparticles have been shown to induce sufficient contrast for in vivo magnetic resonance imaging enabling the in vivo analysis of the final location of the transplanted cells. For magnetic nanoparticles to be useful, a high internalization efficiency of the particles is required without compromising cell function, as well as validation of the magnetic nanoparticles behaviour inside the cells. RESULTS In this work, we report the development, optimization and validation of an efficient procedure to label human neural stem cells with commercial nanoparticles in the absence of transfection agents. Magnetic nanoparticles used here do not affect cell viability, cell morphology, cell differentiation or cell cycle dynamics. Moreover, human neural stem cells progeny labeled with magnetic nanoparticles are easily and non-invasively detected long time after transplantation in a rat model of Parkinson's disease (up to 5 months post-grafting) by magnetic resonance imaging. CONCLUSIONS These findings support the use of commercial MNPs to track cells for short- and mid-term periods after transplantation for studies of brain cell replacement therapy. Nevertheless, long-term MR images should be interpreted with caution due to the possibility that some MNPs may be expelled from the transplanted cells and internalized by host microglial cells.
Collapse
Affiliation(s)
- Milagros Ramos-Gómez
- Centre for Biomedical Technology, Polytechnic University of Madrid, 28223, Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| | - Emma G Seiz
- Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa", Autonomous University of Madrid-C.S.I.C, 28049, Madrid, Spain.
| | - Alberto Martínez-Serrano
- Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa", Autonomous University of Madrid-C.S.I.C, 28049, Madrid, Spain.
| |
Collapse
|
26
|
Bone Marrow-Derived Mesenchymal Stem Cells Repair Necrotic Pancreatic Tissue and Promote Angiogenesis by Secreting Cellular Growth Factors Involved in the SDF-1 α /CXCR4 Axis in Rats. Stem Cells Int 2015; 2015:306836. [PMID: 25810724 PMCID: PMC4355908 DOI: 10.1155/2015/306836] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/29/2014] [Accepted: 01/08/2015] [Indexed: 01/08/2023] Open
Abstract
Acute pancreatitis (AP), a common acute abdominal disease, 10%-20% of which can evolve into severe acute pancreatitis (SAP), is of significant morbidity and mortality. Bone marrow-derived mesenchymal stem cells (BMSCs) have been reported to have a potential therapeutic role on SAP, but the specific mechanism is unclear. Therefore, we conducted this experiment to shed light on the probable mechanism. We validated that SDF-1α significantly stimulated the expressions of VEGF, ANG-1, HGF, TGF-β, and CXCR4 in BMSCs, which were inhibited by its receptor agonist, AMD3100. The capacities of proliferation, migration, and repair of human umbilical vein endothelial cells were enhanced by BMSCs supernatant. Meanwhile, BMSCs supernatant could also promote angiogenesis, especially after the stimulation with SDF-1α. In vivo, the migration of BMSCs was regulated by SDF-1α/CXCR4 axis. Moreover, transplanted BMSCs could significantly alleviate SAP, reduce the systematic inflammation (TNF-α↓, IL-1β↓, IL-6↓, IL-4↑, IL-10↑, and TGF-β↑), and promote tissue repair and angiogenesis (VEGF↑, ANG-1↑, HGF↑, TGF-β↑, and CD31↑), compared with the SAP and anti-CXCR4 groups. Taken together, the results showed that BMSCs ameliorated SAP and the SDF-1α/CXCR4 axis was involved in the repair and regeneration process.
Collapse
|
27
|
Agrawal H, Shang H, Sattah AP, Yang N, Peirce SM, Katz AJ. Human adipose-derived stromal/stem cells demonstrate short-lived persistence after implantation in both an immunocompetent and an immunocompromised murine model. Stem Cell Res Ther 2014; 5:142. [PMID: 25523792 PMCID: PMC4445497 DOI: 10.1186/scrt532] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 12/08/2014] [Indexed: 01/16/2023] Open
Abstract
Introduction Mesenchymal cells are emerging as a promising cell platform for regenerative therapies. However, the fate of cells after transplantation in many different disease settings and tissue beds remains unclear. Methods In this study, human adipose-derived stromal/stem (ASCs) cells were fluorescently labeled with a membrane dye and injected into both immunocompetent and immunocompromised mouse strains. Cells were injected either as single cell suspensions, or as self-assembling spheroids. In parallel, cells were purposefully devitalized prior to injection and then implanted in the opposite side in a randomized fashion. These ‘control’ groups were included to determine whether the fluorescent membrane dye would remain localized at the injection site despite the use of nonviable cells. Cell implants and the surrounding tissues were harvested on days 3, 10 and 21 after in vivo delivery and evaluated in a blinded manner. Injection sites were analyzed by fluorescent microscopy, and human cell numbers were quantified using PCR detection of a human-specific endogenous retrovirus (ERV-3). Host response was evaluated by immunofluorescent staining of macrophages. Results ERV-3 quantification showed that 95% of the human cells that were viable when they were injected were undetectable at the three-week time-point. Although fluorescent signal persisted for the entire study period, further analysis revealed that much of this signal was located within host macrophages. Conclusions These results suggest that human ASCs survive for less than three weeks after injection into even immunocompromised mice, and call into question the notion that human ASCs are immuno-privileged and capable of surviving for extended periods in xenogeneic and/or allogeneic models.
Collapse
|
28
|
Moudgil R, Dick AJ. Regenerative Cell Imaging in Cardiac Repair. Can J Cardiol 2014; 30:1323-34. [DOI: 10.1016/j.cjca.2014.08.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/29/2014] [Accepted: 08/29/2014] [Indexed: 01/03/2023] Open
|
29
|
Naumova AV, Modo M, Moore A, Murry CE, Frank JA. Clinical imaging in regenerative medicine. Nat Biotechnol 2014; 32:804-18. [PMID: 25093889 PMCID: PMC4164232 DOI: 10.1038/nbt.2993] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/15/2014] [Indexed: 01/09/2023]
Abstract
In regenerative medicine, clinical imaging is indispensable for characterizing damaged tissue and for measuring the safety and efficacy of therapy. However, the ability to track the fate and function of transplanted cells with current technologies is limited. Exogenous contrast labels such as nanoparticles give a strong signal in the short term but are unreliable long term. Genetically encoded labels are good both short- and long-term in animals, but in the human setting they raise regulatory issues related to the safety of genomic integration and potential immunogenicity of reporter proteins. Imaging studies in brain, heart and islets share a common set of challenges, including developing novel labeling approaches to improve detection thresholds and early delineation of toxicity and function. Key areas for future research include addressing safety concerns associated with genetic labels and developing methods to follow cell survival, differentiation and integration with host tissue. Imaging may bridge the gap between cell therapies and health outcomes by elucidating mechanisms of action through longitudinal monitoring.
Collapse
Affiliation(s)
- Anna V Naumova
- 1] Department of Radiology, University of Washington, Seattle, Washington, USA. [2] Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA. [3] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Michel Modo
- 1] McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [2] Centre for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [3] Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [4] Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anna Moore
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Charles E Murry
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA. [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA. [3] Department of Pathology, University of Washington, Seattle, Washington, USA. [4] Department of Bioengineering, University of Washington, Seattle, Washington, USA. [5] Department of Medicine/Cardiology, University of Washington, Seattle, Washington, USA
| | - Joseph A Frank
- 1] Radiology and Imaging Sciences, Clinical, National Institutes of Health, Bethesda, Maryland, USA. [2] National Institutes of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
30
|
Cianciaruso C, Pagani A, Martelli C, Bacigaluppi M, Squadrito ML, Lo Dico A, De Palma M, Furlan R, Lucignani G, Falini A, Biffi A, Ottobrini L, Politi LS. Cellular magnetic resonance with iron oxide nanoparticles: long-term persistence of SPIO signal in the CNS after transplanted cell death. Nanomedicine (Lond) 2014; 9:1457-74. [PMID: 24823433 DOI: 10.2217/nnm.14.84] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIM To study the specificity of cellular MRI based on superparamagnetic iron oxide particles (SPIOs), especially within the CNS. MATERIALS & METHODS A microglial cell line was engineered for the expression of a suicide gene, the receptor of diphtheria toxin (DT), and two reporter genes, green fluorescent protein and luciferase, in order to induce, in a controlled manner, cell death and test it through bioluminescence. SPIO-labeled DT-sensitive and control DT-insensitive cells were transplanted into the brains of mice, which underwent serial MRI and bioluminescence studies before and up to 90 days after DT-induced cell death. RESULTS No variations in SPIO signal voids were detected along longitudinal monitoring in brain hemispheres transplanted with DT-sensitive cells. Ex vivo analyses showed persistence of iron nanoparticle deposits at transplantation sites. CONCLUSION Due to the long-term persistence of signal after transplanted cell death, caution is advised when SPIOs are employed for cell tracking.
Collapse
Affiliation(s)
- Chiara Cianciaruso
- Neuroradiology Department & Neuroradiology Research Group, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Anal sphincter repair with muscle progenitor cell transplantation: serial assessment with iron oxide-enhanced MRI. AJR Am J Roentgenol 2014; 202:619-25. [PMID: 24555600 DOI: 10.2214/ajr.13.11146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The purpose of this study was to assess homing of ultrasmall superparamagnetic iron oxide (USPIO)-labeled muscle progenitor cells in an experimental rabbit model of anal sphincter repair using MRI. MATERIALS AND METHODS Twelve rabbits underwent external anal sphincterotomy and randomly received injection of either autologous muscle progenitor cells labeled with USPIO at a concentration of 4 mg/10(6) cells (experimental group) or saline (control group) at the site of sphincter damage. In vivo MRI, electromyography, and manometry were performed before, 1 hour after, and 1, 2, and 4 weeks after the injection. At the end time point, anal sphincter sections were obtained for histologic analysis. Semiquantitative analysis of fibrosis, desmin, iron, CD3, and CD68 was performed using two microscopic fields in two distinct regions of the sphincter according to either presence (zone I) or absence (zone II) of signal loss on the corresponding MR images. RESULTS Labeling efficiency was 88.67% and did not influence cell viability. On follow-up images of the cell-transplanted rabbits, significant influence was reported at 1 hour, 1 and 2 weeks after transplantation. The maximum signal loss was detected at 1 hour (75.7%). Regenerating myofibers stained positively for desmin and mainly correlated to zone I on MR images. Clusters of iron-positive particles were detectable in the myofibers located mainly at the site of injection, which correlated well to zone I. Significant signal loss and Perls Prussian blue-positive area were not detected in the control group. Functional studies showed significant improvement in sphincter pressure and electrical activity in the experimental group after 4 weeks (p<0.001). CONCLUSION Our results support the potential of iron oxide-enhanced MRI for serial monitoring of transplanted cells in an animal model of anal sphincter repair.
Collapse
|
32
|
Sibov TT, Pavon LF, Miyaki LA, Mamani JB, Nucci LP, Alvarim LT, Silveira PH, Marti LC, Gamarra L. Umbilical cord mesenchymal stem cells labeled with multimodal iron oxide nanoparticles with fluorescent and magnetic properties: application for in vivo cell tracking. Int J Nanomedicine 2014; 9:337-50. [PMID: 24531365 PMCID: PMC3891565 DOI: 10.2147/ijn.s53299] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Here we describe multimodal iron oxide nanoparticles conjugated to Rhodamine-B (MION-Rh), their stability in culture medium, and subsequent validation of an in vitro protocol to label mesenchymal stem cells from umbilical cord blood (UC-MSC) with MION-Rh. These cells showed robust labeling in vitro without impairment of their functional properties, the viability of which were evaluated by proliferation kinetic and ultrastructural analyzes. Thus, labeled cells were infused into striatum of adult male rats of animal model that mimic late onset of Parkinson’s disease and, after 15 days, it was observed that cells migrated along the medial forebrain bundle to the substantia nigra as hypointense spots in T2 magnetic resonance imaging. These data were supported by short-term magnetic resonance imaging. Studies were performed in vivo, which showed that about 5 × 105 cells could be efficiently detected in the short term following infusion. Our results indicate that these labeled cells can be efficiently tracked in a neurodegenerative disease model.
Collapse
Affiliation(s)
- Tatiana T Sibov
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Departamento de Neurologia e Neurociências, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lorena F Pavon
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Departamento de Neurologia e Neurociências, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Liza A Miyaki
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Leopoldo P Nucci
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Departamento de Neurologia e Neurociências, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Larissa T Alvarim
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | | | | | - Lf Gamarra
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Departamento de Neurologia e Neurociências, Universidade Federal de São Paulo, São Paulo, Brazil ; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| |
Collapse
|
33
|
Achyut BR, Varma NRS, Arbab AS. Application of Umbilical Cord Blood Derived Stem Cells in Diseases of the Nervous System. ACTA ACUST UNITED AC 2014; 4. [PMID: 25599002 DOI: 10.4172/2157-7633.1000202] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Umbilical cord blood (UCB) derived multipotent stem cells are capable of giving rise hematopoietic, epithelial, endothelial and neural progenitor cells. Thus suggested to significantly improve graft-versus-host disease and represent the distinctive therapeutic option for several malignant and non-malignant diseases. Recent advances in strategies to isolate, expand and shorten the timing of UCB stem cells engraftment have tremendously improved the efficacy of transplantations. Nervous system has limited regenerative potential in disease conditions such as cancer, neurodegeneration, stroke, and several neural injuries. This review focuses on application of UCB derived stem/progenitor cells in aforementioned pathological conditions. We have discussed the possible attempts to make use of UCB therapies to generate neural cells and tissues with developmental and functional similarities to neuronal cells. In addition, emerging applications of UCB derived AC133+ (CD133+) endothelial progenitor cells (EPCs) as imaging probe, regenerative agent, and gene delivery vehicle are mentioned that will further improve the understanding of use of UCB cells in therapeutic modalities. However, safe and effective protocols for cell transplantations are still required for therapeutic efficacy.
Collapse
Affiliation(s)
- Bhagelu R Achyut
- Tumor Angiogenesis Lab, Cancer Center, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Ali S Arbab
- Tumor Angiogenesis Lab, Cancer Center, Georgia Regents University, Augusta, GA 30912, USA
| |
Collapse
|
34
|
Kolosnjaj-Tabi J, Wilhelm C, Clément O, Gazeau F. Cell labeling with magnetic nanoparticles: opportunity for magnetic cell imaging and cell manipulation. J Nanobiotechnology 2013; 11 Suppl 1:S7. [PMID: 24564857 PMCID: PMC4029272 DOI: 10.1186/1477-3155-11-s1-s7] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This tutorial describes a method of controlled cell labeling with citrate-coated ultra small superparamagnetic iron oxide nanoparticles. This method may provide basically all kinds of cells with sufficient magnetization to allow cell detection by high-resolution magnetic resonance imaging (MRI) and to enable potential magnetic manipulation. In order to efficiently exploit labeled cells, quantify the magnetic load and deliver or follow-up magnetic cells, we herein describe the main requirements that should be applied during the labeling procedure. Moreover we present some recommendations for cell detection and quantification by MRI and detail magnetic guiding on some real-case studies in vitro and in vivo.
Collapse
|
35
|
Bone mesenchymal stem cells contributed to the neointimal formation after arterial injury. PLoS One 2013; 8:e82743. [PMID: 24349351 PMCID: PMC3857273 DOI: 10.1371/journal.pone.0082743] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/28/2013] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Recent findings suggest that in response to repair-to-injury bone marrow mesenchymal stem cells (BMSCs) participate in the process of angiogenesis. It is unclear what role BMSCs play in the structure of the vessel wall. In present study, we aimed to determine whether BMSCs had the capacity of endothelial cells (ECs). METHODS BMSCs were separated and cultured. FACS and RT-PCR analysis confirmed the gene expression phenotype. The capacity of migration and adhesion and the ultrastructure of BMSCs were examined. The effect of BMSCs transplantation on the vascular repair was investigated in a murine carotid artery-injured model. RESULTS BMSCs could express some markers and form the tube-like structure. The migration and adhesion capacity of BMSCs increased significantly after stimulated. In addition, BMSCs had the intact cell junction. In vivo the local transfer of BMSCs differentiated into neo-endothelial cells in the injury model for carotid artery and contributed to the vascular remodeling. CONCLUSION These results showed that BMSCs could contribute to neointimal formation for vascular lesion and might be associated with the differentiation into ECs, which indicated the important therapeutic implications for vascular diseases.
Collapse
|
36
|
Hachani R, Lowdell M, Birchall M, Thanh NTK. Tracking stem cells in tissue-engineered organs using magnetic nanoparticles. NANOSCALE 2013; 5:11362-11373. [PMID: 24108444 DOI: 10.1039/c3nr03861k] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The use of human stem cells (SCs) in tissue engineering holds promise in revolutionising the treatment of numerous diseases. There is a pressing need to comprehend the distribution, movement and role of SCs once implanted onto scaffolds. Nanotechnology has provided a platform to investigate this through the development of inorganic magnetic nanoparticles (MNPs). MNPs can be used to label and track SCs by magnetic resonance imaging (MRI) since this clinically available imaging modality has high spatial resolution. In this review, we highlight recent applications of iron oxide and gadolinium based MNPs in SC labelling and MRI; and offer novel considerations for their future development.
Collapse
Affiliation(s)
- Roxanne Hachani
- Department of Physics & Astronomy, University College London, Gower Street, London WC1E 6BT, UK.
| | | | | | | |
Collapse
|
37
|
Panizzo RA, Gadian DG, Sowden JC, Wells JA, Lythgoe MF, Ferretti P. Monitoring ferumoxide-labelled neural progenitor cells and lesion evolution by magnetic resonance imaging in a model of cell transplantation in cerebral ischaemia. F1000Res 2013; 2:252. [PMID: 24715962 PMCID: PMC3962009 DOI: 10.12688/f1000research.2-252.v2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2014] [Indexed: 01/05/2023] Open
Abstract
Efficacy of neural stem/progenitor cell (NPC) therapies after cerebral ischaemia could be better evaluated by monitoring
in vivo migration and distribution of cells post-engraftment in parallel with analysis of lesion volume and functional recovery. Magnetic resonance imaging (MRI) is ideally placed to achieve this, but still poses several challenges. We show that combining the ferumoxide MRI contrast agent Endorem with protamine sulphate (FePro) improves iron oxide uptake in cells compared to Endorem alone and is non-toxic. Hence FePro complex is a better contrast agent than Endorem for monitoring NPCs. FePro complex-labelled NPCs proliferated and differentiated normally
in vitro, and upon grafting into the brain 48 hours post-ischaemia they were detected
in vivo by MRI. Imaging over four weeks showed the development of a confounding endogenous hypointense contrast evolution at later timepoints within the lesioned tissue. This was at least partly due to accumulation within the lesion of macrophages and endogenous iron. Neither significant NPC migration, assessed by MRI and histologically, nor a reduction in the ischaemic lesion volume was observed in NPC-grafted brains. Crucially, while MRI provides reliable information on engrafted cell location early after an ischaemic insult, pathophysiological changes to ischaemic lesions can interfere with cellular imaging at later timepoints.
Collapse
Affiliation(s)
- Rachael A Panizzo
- Developmental Biology Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK ; Imaging and Biophysics Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK ; UCL Centre for Advanced Biomedical Imaging, Department of Medicine, University College London, London, WC1E 6DD, UK
| | - David G Gadian
- Imaging and Biophysics Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Jane C Sowden
- Developmental Biology Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Jack A Wells
- UCL Centre for Advanced Biomedical Imaging, Department of Medicine, University College London, London, WC1E 6DD, UK
| | - Mark F Lythgoe
- Imaging and Biophysics Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK ; UCL Centre for Advanced Biomedical Imaging, Department of Medicine, University College London, London, WC1E 6DD, UK
| | - Patrizia Ferretti
- Developmental Biology Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK
| |
Collapse
|
38
|
Douvaras P, Mort RL, Edwards D, Ramaesh K, Dhillon B, Morley SD, Hill RE, West JD. Increased corneal epithelial turnover contributes to abnormal homeostasis in the Pax6(+/-) mouse model of aniridia. PLoS One 2013; 8:e71117. [PMID: 23967157 PMCID: PMC3742784 DOI: 10.1371/journal.pone.0071117] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/24/2013] [Indexed: 01/09/2023] Open
Abstract
We aimed to test previous predictions that limbal epithelial stem cells (LESCs) are quantitatively deficient or qualitatively defective in Pax6+/− mice and decline with age in wild-type (WT) mice. Consistent with previous studies, corneal epithelial stripe patterns coarsened with age in WT mosaics. Mosaic patterns were also coarser in Pax6+/− mosaics than WT at 15 weeks but not at 3 weeks, which excludes a developmental explanation and strengthens the prediction that Pax6+/− mice have a LESC-deficiency. To investigate how Pax6 genotype and age affected corneal homeostasis, we compared corneal epithelial cell turnover and label-retaining cells (LRCs; putative LESCs) in Pax6+/− and WT mice at 15 and 30 weeks. Limbal BrdU-LRC numbers were not reduced in the older WT mice, so this analysis failed to support the predicted age-related decline in slow-cycling LESC numbers in WT corneas. Similarly, limbal BrdU-LRC numbers were not reduced in Pax6+/− heterozygotes but BrdU-LRCs were also present in Pax6+/− corneas. It seems likely that Pax6+/− LRCs are not exclusively stem cells and some may be terminally differentiated CD31-positive blood vessel cells, which invade the Pax6+/− cornea. It was not, therefore, possible to use this approach to test the prediction that Pax6+/− corneas had fewer LESCs than WT. However, short-term BrdU labelling showed that basal to suprabasal movement (leading to cell loss) occurred more rapidly in Pax6+/− than WT mice. This implies that epithelial cell loss is higher in Pax6+/− mice. If increased corneal epithelial cell loss exceeds the cell production capacity it could cause corneal homeostasis to become unstable, resulting in progressive corneal deterioration. Although it remains unclear whether Pax6+/− mice have LESC-deficiency, we suggest that features of corneal deterioration, that are often taken as evidence of LESC-deficiency, might occur in the absence of stem cell deficiency if corneal homeostasis is destabilised by excessive cell loss.
Collapse
Affiliation(s)
- Panagiotis Douvaras
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard L. Mort
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Dominic Edwards
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Kanna Ramaesh
- Tennent Institute of Ophthalmology, Gartnaval General Hospital, Glasgow, United Kingdom
| | - Baljean Dhillon
- School of Clinical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Steven D. Morley
- Division of Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert E. Hill
- Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - John D. West
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
39
|
Martinez-Quintanilla J, Bhere D, Heidari P, He D, Mahmood U, Shah K. Therapeutic efficacy and fate of bimodal engineered stem cells in malignant brain tumors. Stem Cells 2013; 31:1706-14. [PMID: 23389839 PMCID: PMC3775922 DOI: 10.1002/stem.1355] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 12/31/2012] [Indexed: 12/28/2022]
Abstract
Therapeutically engineered stem cells (SC) are emerging as an effective tumor-targeted approach for different cancer types. However, the assessment of the long-term fate of therapeutic SC post-tumor treatment is critical if such promising therapies are to be translated into clinical practice. In this study, we have developed an efficient SC-based therapeutic strategy that simultaneously allows killing of tumor cells and assessment and eradication of SC after treatment of highly malignant glioblastoma multiforme (GBM). Mesenchymal stem cells (MSC) engineered to co-express the prodrug converting enzyme, herpes simplex virus thymidine kinase (HSV-TK) and a potent and secretable variant of tumor necrosis factor apoptosis-inducing ligand (S-TRAIL) induced caspase-mediated GBM cell death and showed selective MSC sensitization to the prodrug ganciclovir (GCV). A significant decrease in tumor growth and a subsequent increase in survival were observed when mice bearing highly aggressive GBM were treated with MSC coexpressing S-TRAIL and HSV-TK. Furthermore, the systemic administration of GCV post-tumor treatment selectively eliminated therapeutic MSC expressing HSV-TK in vitro and in vivo, which was monitored in real time by positron emission-computed tomography imaging using 18F-FHBG, a substrate for HSV-TK. These findings demonstrate the development and validation of a novel therapeutic strategy that has implications in translating SC-based therapies in cancer patients.
Collapse
Affiliation(s)
- Jordi Martinez-Quintanilla
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Deepak Bhere
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Pedram Heidari
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Derek He
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Umar Mahmood
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
| | - Khalid Shah
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138
| |
Collapse
|
40
|
Li L, Jiang W, Luo K, Song H, Lan F, Wu Y, Gu Z. Superparamagnetic iron oxide nanoparticles as MRI contrast agents for non-invasive stem cell labeling and tracking. Am J Cancer Res 2013; 3:595-615. [PMID: 23946825 PMCID: PMC3741608 DOI: 10.7150/thno.5366] [Citation(s) in RCA: 287] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 12/12/2012] [Indexed: 12/21/2022] Open
Abstract
Stem cells hold great promise for the treatment of multiple human diseases and disorders. Tracking and monitoring of stem cells in vivo after transplantation can supply important information for determining the efficacy of stem cell therapy. Magnetic resonance imaging (MRI) combined with contrast agents is believed to be the most effective and safest non-invasive technique for stem cell tracking in living bodies. Commercial superparamagnetic iron oxide nanoparticles (SPIONs) in the aid of transfection agents (TAs) have been applied to labeling stem cells. However, owing to the potential toxicity of TAs, more attentions have been paid to develop novel SPIONs with specific surface coating or functional moieties which facilitate effective cell internalization in the absence of TAs. This review aims to summarize the recent progress in the design and preparation of SPIONs as cellular MRI probes, to discuss their applications and current problems facing in stem cell labeling and tracking, and to offer perspectives and solutions for the future development of SPIONs in this field.
Collapse
|
41
|
Edmundson M, Thanh NTK, Song B. Nanoparticles based stem cell tracking in regenerative medicine. Theranostics 2013; 3:573-82. [PMID: 23946823 PMCID: PMC3741606 DOI: 10.7150/thno.5477] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 01/07/2013] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapies offer great potentials in the treatment for a wide range of diseases and conditions. With so many stem cell replacement therapies going through clinical trials currently, there is a great need to understand the mechanisms behind a successful therapy, and one of the critical points of discovering them is to track stem cell migration, proliferation and differentiation in vivo. To be of most use tracking methods should ideally be non-invasive, high resolution and allow tracking in three dimensions. Magnetic resonance imaging (MRI) is one of the ideal methods, but requires a suitable contrast agent to be loaded to the cells to be tracked, and one of the most wide-spread in stem cell tracking is a group of agents known as magnetic nanoparticles. This review will explore the current use of magnetic nanoparticles in developing and performing stem cell therapies, and will investigate their potential limitations and the future directions magnetic nanoparticle tracking is heading in.
Collapse
|
42
|
Liu G, Gao J, Ai H, Chen X. Applications and potential toxicity of magnetic iron oxide nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:1533-45. [PMID: 23019129 DOI: 10.1002/smll.201201531] [Citation(s) in RCA: 349] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Indexed: 05/22/2023]
Abstract
Owing to their unique physical and chemical properties, magnetic iron oxide nanoparticles have become a powerful platform in many diverse aspects of biomedicine, including magnetic resonance imaging, drug and gene delivery, biological sensing, and hyperthermia. However, the biomedical applications of magnetic iron oxide nanoparticles arouse serious concerns about their pharmacokinetics, metabolism, and toxicity. In this review, the updated research on the biomedical applications and potential toxicity of magnetic iron oxide nanoparticles is summarized. Much more effort is required to develop magnetic iron oxide nanoparticles with improved biocompatible surface engineering to achieve minimal toxicity, for various applications in biomedicine.
Collapse
Affiliation(s)
- Gang Liu
- Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361005, China.
| | | | | | | |
Collapse
|
43
|
Noad J, Gonzalez-Lara LE, Broughton HC, McFadden C, Chen Y, Hess DA, Foster PJ. MRI tracking of transplanted iron-labeled mesenchymal stromal cells in an immune-compromised mouse model of critical limb ischemia. NMR IN BIOMEDICINE 2013; 26:458-467. [PMID: 23165968 DOI: 10.1002/nbm.2884] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 08/30/2012] [Accepted: 09/29/2012] [Indexed: 06/01/2023]
Abstract
Peripheral arterial disease is a clinical problem in which mesenchymal stromal cell (MSC) transplantation may offer substantial benefit by promoting the generation of new blood vessels and improving limb ischemia and wound healing via their potent paracrine activities. MRI allows for the noninvasive tracking of cells over time using iron oxide contrast agents to label cells before they are injected or transplanted. However, a major limitation of the tracking of iron oxide-labeled cells with MRI is the possibility that dead or dying cells will transfer the iron oxide label to local bystander macrophages, making it very difficult to distinguish between viable transplanted cells and endogenous macrophages in the images. In this study, a severely immune-compromised mouse, with limited macrophage activity, was investigated to examine cell tracking in a system in which bystander cell uptake of dead, iron-labeled cells or free iron particles was minimized. MRI was used to track the fate of MSCs over 21 days after their intramuscular transplantation in mice with a femoral artery ligation. In all mice, a region of signal loss was observed at the injection site and the volume of signal hypointensity diminished over time. Fluorescence and light microscopy showed that iron-positive MSCs persisted at the transplant site and often appeared to be integrated in perivascular niches. This was compared with MSC transplantation in immune-competent mice with femoral artery ligation. In these mice, the regions of signal loss caused by iron-labeled MSC cleared more slowly, and histology revealed iron particles trapped at the site of cell transplantation and associated with areas of inflammation.
Collapse
Affiliation(s)
- Jennifer Noad
- Robarts Research Institute, London, ON, Canada; Department of Medical Biophysics, The University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
44
|
Li XX, Li KA, Qin JB, Ye KC, Yang XR, Li WM, Xie QS, Jiang ME, Zhang GX, Lu XW. In vivo MRI tracking of iron oxide nanoparticle-labeled human mesenchymal stem cells in limb ischemia. Int J Nanomedicine 2013; 8:1063-73. [PMID: 23515426 PMCID: PMC3598527 DOI: 10.2147/ijn.s42578] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background Stem cell transplantation has been investigated for repairing damaged tissues in various injury models. Monitoring the safety and fate of transplanted cells using noninvasive methods is important to advance this technique into clinical applications. Methods In this study, lower-limb ischemia models were generated in nude mice by femoral artery ligation. As negative-contrast agents, positively charged magnetic iron oxide nanoparticles (aminopropyltriethoxysilane-coated Fe2O3) were investigated in terms of in vitro labeling efficiency, effects on human mesenchymal stromal cell (hMSC) proliferation, and in vivo magnetic resonance imaging (MRI) visualization. Ultimately, the mice were sacrificed for histological analysis three weeks after transplantation. Results With efficient labeling, aminopropyltriethoxysilane-modified magnetic iron oxide nanoparticles (APTS-MNPs) did not significantly affect hMSC proliferation. In vivo, APTS-MNP-labeled hMSCs could be monitored by clinical 3 Tesla MRI for at least three weeks. Histological examination detected numerous migrated Prussian blue-positive cells, which was consistent with the magnetic resonance images. Some migrated Prussian blue-positive cells were positive for mature endothelial cell markers of von Willebrand factor and anti-human proliferating cell nuclear antigen. In the test groups, Prussian blue-positive nanoparticles, which could not be found in other organs, were detected in the spleen. Conclusion APTS-MNPs could efficiently label hMSCs, and clinical 3 Tesla MRI could monitor the labeled stem cells in vivo, which may provide a new approach for the in vivo monitoring of implanted cells.
Collapse
Affiliation(s)
- Xiang-Xiang Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Molcanyi M, Bosche B, Kraitsy K, Patz S, Zivcak J, Riess P, El Majdoub F, Hescheler J, Goldbrunner R, Schäfer U. Pitfalls and fallacies interfering with correct identification of embryonic stem cells implanted into the brain after experimental traumatic injury. J Neurosci Methods 2013; 215:60-70. [PMID: 23454685 DOI: 10.1016/j.jneumeth.2013.02.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/13/2013] [Accepted: 02/14/2013] [Indexed: 11/26/2022]
Abstract
Cell-therapy was proposed to be a promising tool in case of death or impairment of specific cell types. Correct identification of implanted cells became crucial when evaluating the success of transplantation therapy. Various methods of cell labeling have been employed in previously published studies. The use of intrinsic signaling of green fluorescent protein (GFP) has led to a well known controversy in the field of cardiovascular research. We encountered similar methodological pitfalls after transplantation of GFP-transfected embryonic stem cells into rat brains following traumatic brain injury (TBI). As the identification of implanted graft by intrinsic autofluorescence failed, anti-GFP labeling coupled to fluorescent and conventional antibodies was needed to visualize the implanted cells. Furthermore, different cell types with strong intrinsic autofluorescence were found at the sites of injury and transplantation, thus mimicking the implanted stem cells. GFP-positive stem cells were correctly localized, using advanced histological techniques. The activation of microglia/macrophages, accompanying the transplantation post TBI, was shown to be a significant source of artefacts, interfering with correct identification of implanted stem cells. Dependent on the strategy of stem cell tracking, the phagocytosis of implanted cells as observed in this study, might also impede the interpretation of results. Critical appraisal of previously published data as well as a review of different histological techniques provide tools for a more accurate identification of transplanted stem cells.
Collapse
Affiliation(s)
- Marek Molcanyi
- Clinic of Neurosurgery, University of Cologne, Kerpener Strasse 62, 50937 Köln, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pichler R, Klima G, Richter E, Marksteiner R, Mayr V, Skradski V, Horninger W, Oswald J. Autologous fibroblast transplantation at the vesico-ureteral junction as potential reconstructive cell replacement in an animal model. World J Urol 2012; 31:169-74. [PMID: 22864402 DOI: 10.1007/s00345-012-0914-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 07/13/2012] [Indexed: 11/30/2022] Open
Abstract
PURPOSE To evaluate the cellular survival of donor fibroblasts after transplantation at the vesico-ureteral junction (VUJ) and to analyse their potential for reconstructive cell replacement in an animal model as autologous fibroblasts have been used as soft tissue augmentation material for scared and damaged tissue. METHODS Muscles biopsies were procured from the lower limb muscles of 4 pigs; cytoplasm of fibroblasts was labelled with nano-sized iron oxide particles. Six weeks after taking of the muscle biopsies, fibroblast transplantation was performed, 3 × 10(6) cells suspended in transplantation medium (in 1-ml syringes) were injected at the VUJ using the modified STING technique. Animals were killed 8 weeks later; seeded fibroblasts were identified using prussian blue staining protocol; histological evaluation and morphological analysis were performed by light microscopy (Mayer's haematoxylin-eosin staining); and bladders were scanned by MRI for visualization and localization of the iron-labelled donor cells. RESULTS Donor fibroblast cell colonization and cellular viability at the VUJ was demonstrated by MRI and histochemically indicating cellular uptake of iron particles at the VUJ. It was also evident that transplanted fibroblasts integrate into the extracellular matrix of the distal ureter augmenting ureteral host tissue. CONCLUSIONS Labelled implanted autologous fibroblasts were visualized by staining procedure as well as MRI scan demonstrating persistence at the VUJ, suggesting that in vitro expanded fibroblasts survived in vivo after transplantation.
Collapse
Affiliation(s)
- Renate Pichler
- Department of Urology, Medical University of Innsbruck, Anichstreet 35, 6020, Innsbruck, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Kim JH, Jin SM, Oh SH, Lee S, Oh BJ, Kim SK, Suh S, Lee JH, Jung HS, Lee MS, Lee MK, Kim KW. Counting small hypointense spots confounds the quantification of functional islet mass based on islet MRI. Am J Transplant 2012; 12:1303-12. [PMID: 22299723 DOI: 10.1111/j.1600-6143.2011.03941.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Iron-containing fragmented islets or free iron released from dying cells could confound the interpretation of MRI of iron nanoparticle-labeled islets. Exclusion of small hypointense spots could be a useful strategy to avoid such artifact. We investigated whether this strategy could improve the estimation of functioning islet mass after islet transplantation. Using a rat syngeneic intraportal islet transplantation model, we quantitatively assessed the relationships between total area, number of hypointense spots on MRI that belong to each size quartile and glycemic control of the recipients. The total area of hypointense spots on MRI was greater in the recipients that achieved diabetes reversal (p = 0.002), whereas the total number of hypointense spots was not different (p = 0.757). Exclusion of small hypointense spots improved the association between the number of hypointense spots and the blood glucose level of the recipients (p < 0.001). Ex-vivo imaging and histologic study confirmed that some small hypointense spots represent the phagocytosed free iron. Exclusion of small hypointense spots improved the quantification of the functional islet mass based on islet MRI. This would be a useful principle in the development of an algorithm to estimate functioning islet mass based on islet MRI.
Collapse
Affiliation(s)
- J H Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
During the past two decades, stem cells have created enthusiasm as a regenerative therapy for ischemic heart disease. Transplantation of bone marrow stem cells, skeletal myoblasts, and endothelial progenitor cells has shown to improve myocardial function after infarction. Recently, attention has focused on the potential use of embryonic stem cells and induced pluripotent stem cells because they possess the capacity to differentiate into various cell types, including cardiac and endothelial cells. Clinical trials have shown positive effects on the functional recovery of heart after myocardial infarction and have answered questions on timing, dosage, and cell delivery route of stem cells such as those derived from bone marrow. Despite the current advances in stem cell research, one main hurdle remains the lack of reliable information about the fate of cell engraftment, survival, and proliferation after transplantation. This review discusses the different cell types used in cardiac cell therapy as well as molecular imaging modalities relevant to survival issues.
Collapse
|
49
|
Perán M, García MA, López-Ruiz E, Bustamante M, Jiménez G, Madeddu R, Marchal JA. Functionalized nanostructures with application in regenerative medicine. Int J Mol Sci 2012; 13:3847-3886. [PMID: 22489186 PMCID: PMC3317746 DOI: 10.3390/ijms13033847] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Revised: 03/03/2012] [Accepted: 03/06/2012] [Indexed: 12/16/2022] Open
Abstract
In the last decade, both regenerative medicine and nanotechnology have been broadly developed leading important advances in biomedical research as well as in clinical practice. The manipulation on the molecular level and the use of several functionalized nanoscaled materials has application in various fields of regenerative medicine including tissue engineering, cell therapy, diagnosis and drug and gene delivery. The themes covered in this review include nanoparticle systems for tracking transplanted stem cells, self-assembling peptides, nanoparticles for gene delivery into stem cells and biomimetic scaffolds useful for 2D and 3D tissue cell cultures, transplantation and clinical application.
Collapse
Affiliation(s)
- Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén E-23071, Spain; E-Mails: (M.P.); (E.L.-R.)
| | - María A. García
- Research Unit, Hospital Universitario Virgen de las Nieves, Granada E-18014, Spain; E-Mail:
| | - Elena López-Ruiz
- Department of Health Sciences, University of Jaén, Jaén E-23071, Spain; E-Mails: (M.P.); (E.L.-R.)
| | - Milán Bustamante
- Biosciences Institute, University College Cork, Cork, Ireland; E-Mail:
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Biomedical Research Centre, University of Granada, Granada E-18100, Spain; E-Mail:
| | - Roberto Madeddu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; E-Mail:
| | - Juan A. Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Biomedical Research Centre, University of Granada, Granada E-18100, Spain; E-Mail:
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada E-18012, Spain
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34-958-249-321; Fax: +34-958-246-296
| |
Collapse
|
50
|
Kotek G, van Tiel ST, Wielopolski PA, Houston GC, Krestin GP, Bernsen MR. Cell quantification: evolution of compartmentalization and distribution of iron-oxide particles and labeled cells. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 7:195-203. [DOI: 10.1002/cmmi.481] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Gyula Kotek
- Department of Radiology; Erasmus MC; Rotterdam The Netherlands
| | | | | | - Gavin C. Houston
- Applied Science Laboratory; General Electric Healthcare; The Netherlands
| | | | | |
Collapse
|