1
|
Sindeeva OA, Kozyreva ZV, Abdurashitov AS, Sukhorukov GB. Engineering colloidal systems for cell manipulation, delivery, and tracking. Adv Colloid Interface Sci 2025; 340:103462. [PMID: 40037017 DOI: 10.1016/j.cis.2025.103462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
Men-made colloidal systems are widely presented across various aspects of biomedical science. There is a strong demand for engineering colloids to tailor their functions and properties to meet the requirements of biological and medical tasks. These requirements are not only related to size, shape, capacity to carry bioactive compounds as drug delivery systems, and the ability to navigate via chemical and physical targeting. Today, the more challenging aspects of colloid design are how the colloidal particles interact with biological cells, undergo internalization by cells, how they reside in the cell interior, and whether we can explore cells with colloids, intervene with biochemical processes, and alter cell functionality. Cell tracking, exploitation of cells as natural transporters of internalized colloidal carriers loaded with drugs, and exploring physical methods as external triggers of cell functions are ongoing topics in the research agenda. In this review, we summarize recent advances in these areas, focusing on how colloidal particles interact and are taken up by mesenchymal stem cells, dendritic cells, neurons, macrophages, neutrophils and lymphocytes, red blood cells, and platelets. The engineering of colloidal vesicles with cell membrane fragments and exosomes facilitates their application. The perspectives of different approaches in colloid design, their limitations, and obstacles on the biological side are discussed.
Collapse
Affiliation(s)
- Olga A Sindeeva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia.
| | - Zhanna V Kozyreva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia
| | - Arkady S Abdurashitov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia; Life Improvement by Future Technologies (LIFT) Center, Bolshoy Boulevard 30, Moscow 121205, Russia
| | - Gleb B Sukhorukov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia.
| |
Collapse
|
2
|
Remmo A, Kosch O, Kampen L, Ludwig A, Wiekhorst F, Löwa N. Counting cells in motion by quantitative real-time magnetic particle imaging. Sci Rep 2024; 14:4253. [PMID: 38378785 PMCID: PMC10879211 DOI: 10.1038/s41598-024-54784-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Magnetic Particle Imaging (MPI) is an advanced and powerful imaging modality for visualization and quantitative real-time detection of magnetic nanoparticles (MNPs). This opens the possibility of tracking cells in vivo once they have been loaded by MNPs. Imaging modalities such as optical imaging, X-ray computed tomography (CT), positron emission tomography (PET), single photon emission computed tomography (SPECT), and magnetic resonance imaging (MRI) face limitations, from depth of penetration and radiation exposure to resolution and quantification accuracy. MPI addresses these challenges, enabling radiation-free tracking of MNP-loaded cells with precise quantification. However, the real-time tracking of MNP-loaded cells with MPI has not been demonstrated yet. This study establishes real-time quantitative tracking of MNP-loaded cells. Therefore, THP-1 monocytes were loaded with three different MNP systems, including the MPI gold standard Resovist and Synomag. The real-time MPI experiments reveal different MPI resolution behaviors of the three MNP systems after cellular uptake. Real-time quantitative imaging was achieved by time-resolved cell number determination and comparison with the number of inserted cells. About 95% of the inserted cells were successfully tracked in a controlled phantom environment. These results underline the potential of MPI for real-time investigation of cell migration and interaction with tissue in vivo.
Collapse
Affiliation(s)
- Amani Remmo
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587, Berlin, Germany.
| | - Olaf Kosch
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587, Berlin, Germany
| | - Lena Kampen
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätzu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Antje Ludwig
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätzu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Frank Wiekhorst
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587, Berlin, Germany
| | - Norbert Löwa
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587, Berlin, Germany
| |
Collapse
|
3
|
Remmo A, Löwa N, Kosch O, Eberbeck D, Ludwig A, Kampen L, Grüttner C, Wiekhorst F. Cell Tracking by Magnetic Particle Imaging: Methodology for Labeling THP-1 Monocytes with Magnetic Nanoparticles for Cellular Imaging. Cells 2022; 11:cells11182892. [PMID: 36139467 PMCID: PMC9496715 DOI: 10.3390/cells11182892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Magnetic particle imaging (MPI) is a noninvasive tomographic imaging modality for the quantitative visualization of magnetic nanoparticles (MNPs) with high temporal and spatial resolution. The general capability of MPI for cell tracking (e.g., monitoring living cells labeled with MNPs) has successfully been shown. MNPs in cell culture media are often subjected to structural and magnetic changes. In addition to the deteriorating reproducibility, this also complicates the systematic study of the relationship between the MNP properties and their cellular uptake for MPI. Here, we present a method for the preparation of magnetically labeled THP-1 (Tamm-Horsfall Protein-1) monocytes that are used in MPI cell tracking. The method development was performed using two different MPI tracers, which exhibited electrostatic and steric stabilizations, respectively. In the first step, the interaction between the MNPs and cell culture media was investigated and adjusted to ensure high structural and magnetic stability. Furthermore, the influences of the incubation time, MNP concentration used for cellular uptake, and individual preparation steps (e.g., the washing of cells) were systematically investigated. Finally, the success of the developed loading method was demonstrated by the MPI measurements. The presented systematic investigation of the factors that influence the MNP loading of cells will help to develop a reliable and reproducible method for MPI monocyte tracking for the early detection of inflammation in the future.
Collapse
Affiliation(s)
- Amani Remmo
- Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
- Correspondence:
| | - Norbert Löwa
- Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| | - Olaf Kosch
- Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| | - Dietmar Eberbeck
- Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| | - Antje Ludwig
- Charité, Center for Cardiovascular Research (CCR), Berlin, Hessische Straße 3-4, 10115 Berlin, Germany
| | - Lena Kampen
- Charité, Center for Cardiovascular Research (CCR), Berlin, Hessische Straße 3-4, 10115 Berlin, Germany
| | - Cordula Grüttner
- Micromod Partikeltechnologie GmbH, Schillingallee 68, 18057 Rostock, Germany
| | - Frank Wiekhorst
- Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587 Berlin, Germany
| |
Collapse
|
4
|
Nakamura M, Nakamura J, Mochizuki C, Kuroda C, Kato S, Haruta T, Kakefuda M, Sato S, Tamanoi F, Sugino N. Analysis of cell-nanoparticle interactions and imaging of in vitro labeled cells showing barcorded endosomes using fluorescent thiol-organosilica nanoparticles surface-functionalized with polyethyleneimine. NANOSCALE ADVANCES 2022; 4:2682-2703. [PMID: 36132282 PMCID: PMC9417756 DOI: 10.1039/d1na00839k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/18/2022] [Indexed: 06/15/2023]
Abstract
Biomedical imaging using cell labeling is an important technique to visualize cell dynamics in the body. To label cells, thiol-organosilica nanoparticles (thiol-OS) containing fluorescein (thiol-OS/Flu) and rhodamine B (thiol-OS/Rho) were surface-functionalized with polyethyleneimine (PEI) (OS/Flu-PEI and OS/Rho-PEI) with 4 molecular weights (MWs). We hypothesized PEI structures such as brush, bent brush, bent lie-down, and coiled types on the surface depending on MWs based on dynamic light scattering and thermal gravimetric analyses. The labeling efficacy of OS/Flu-PEIs was dependent on the PEI MW and the cell type. A dual-particle administration study using thiol-OS and OS-PEIs revealed differential endosomal sorting of the particles depending on the surface of the NPs. The endosomes in the labeled cells using OS/Flu-PEI and thiol-OS/Rho revealed various patterns of fluorescence termed barcoded endosomes. The cells labeled with OS-PEI in vitro were administrated to mice intraperitoneally after in situ labeling of peritoneal cells using thiol-OS/Rho. The in vitro labeled cells were detected and identified in cell aggregates in vivo seamlessly. The labeled cells with barcoded endosomes were also identified in cell aggregates. Biomedical imaging of in vitro OS-PEI-labeled cells combined with in situ labeled cells showed high potential for observation of cell dynamics.
Collapse
Affiliation(s)
- Michihiro Nakamura
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Junna Nakamura
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Chihiro Mochizuki
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Chika Kuroda
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Shigeki Kato
- Department of Organ Anatomy and Nanomedicine, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | | | - Mayu Kakefuda
- EM Application Group, EM Business Unit, JEOL Ltd. Japan
| | - Shun Sato
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| | - Fuyuhiko Tamanoi
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles CA 90095 USA
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University Yoshida-Honmachi, Sakyo-ku Kyoto 606-8501 Japan
| | - Norihiro Sugino
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Yamaguchi University 1-1-1 Minami-Kogushi Ube Yamaguchi 755-8505 Japan
| |
Collapse
|
5
|
Peserico A, Di Berardino C, Russo V, Capacchietti G, Di Giacinto O, Canciello A, Camerano Spelta Rapini C, Barboni B. Nanotechnology-Assisted Cell Tracking. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1414. [PMID: 35564123 PMCID: PMC9103829 DOI: 10.3390/nano12091414] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023]
Abstract
The usefulness of nanoparticles (NPs) in the diagnostic and/or therapeutic sector is derived from their aptitude for navigating intra- and extracellular barriers successfully and to be spatiotemporally targeted. In this context, the optimization of NP delivery platforms is technologically related to the exploitation of the mechanisms involved in the NP-cell interaction. This review provides a detailed overview of the available technologies focusing on cell-NP interaction/detection by describing their applications in the fields of cancer and regenerative medicine. Specifically, a literature survey has been performed to analyze the key nanocarrier-impacting elements, such as NP typology and functionalization, the ability to tune cell interaction mechanisms under in vitro and in vivo conditions by framing, and at the same time, the imaging devices supporting NP delivery assessment, and consideration of their specificity and sensitivity. Although the large amount of literature information on the designs and applications of cell membrane-coated NPs has reached the extent at which it could be considered a mature branch of nanomedicine ready to be translated to the clinic, the technology applied to the biomimetic functionalization strategy of the design of NPs for directing cell labelling and intracellular retention appears less advanced. These approaches, if properly scaled up, will present diverse biomedical applications and make a positive impact on human health.
Collapse
Affiliation(s)
- Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (C.D.B.); (V.R.); (G.C.); (O.D.G.); (A.C.); (C.C.S.R.); (B.B.)
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Khattabi AM, Mahmoud NN. Interaction of folate - Linked silica nanoparticles with HeLa cells: Analysis and investigation the effect of polymer length. Saudi Pharm J 2021; 29:1083-1089. [PMID: 34703361 PMCID: PMC8523324 DOI: 10.1016/j.jsps.2021.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 08/01/2021] [Indexed: 11/28/2022] Open
Abstract
This work is a continuance to our previous findings on silica nanoparticles (NPs) modified with diamine polymer, carboxymethyl-β-cyclodextrin (CM-β-CD) and folic acid (FA), respectively. When four different polymer lengths (D230, D400, D2000 and D4000) were analyzed, the release rate of anticancer agents was inversely related to the polymer length while the cell toxicity was directly related to the length. We investigate here the effect of polymer length on the extent of cellular interaction with HeLa cells. The mean particle size, the polydispersity (PD) and the zeta potential of the NPs were measured using dynamic light scattering (DLS), the quantitative analysis of the extent of NPs' interaction was studied using fluorescence microscopy and transmission electron microscopy (TEM) was used to qualitatively visualize them. The particle size increased by increasing the polymer length, the PD values were within the acceptable ranges (0.3−0.5) and the zeta potential was in the range of (−16 to −20 mV). A direct relation was observed between the fluorescence intensity and the length. All modified NPs were capable of entering the cells, however a greater number of NPs with long polymers was observed compared to short polymers. Thus, the direct relation of polymer length to the cell toxicity is due to the release rate behavior and the enhanced interaction of NPs which possess long polymers.
Collapse
Affiliation(s)
- Areen M Khattabi
- Department of Pharmaceutical Sciences and Pharmaceutics, Applied Science Private University, Amman, Jordan
| | - Nouf N Mahmoud
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| |
Collapse
|
7
|
Moonshi SS, Wu Y, Ta HT. Visualizing stem cells in vivo using magnetic resonance imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1760. [PMID: 34651465 DOI: 10.1002/wnan.1760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/18/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022]
Abstract
Stem cell (SC) therapies displayed encouraging efficacy and clinical outcome in various disorders. Despite this huge hype, clinical translation of SC therapy has been disheartening due to contradictory results from clinical trials. The ability to monitor migration and engraftment of cells in vivo represents an ideal strategy in cell therapy. Therefore, suitable imaging approach to track MSCs would allow understanding of migratory and homing efficiency, optimal route of delivery and engraftment of cells at targeted location. Hence, longitudinal tracking of SCs is crucial for the optimization of treatment parameters, leading to improved clinical outcome and translation. Magnetic resonance imaging (MRI) represents a suitable imaging modality to observe cells non-invasively and repeatedly. Tracking is achieved when cells are incubated prior to implantation with appropriate contrast agents (CA) or tracers which can then be detected in an MRI scan. This review explores and emphasizes the importance of monitoring the distribution and fate of SCs post-implantation using current contrast agents, such as positive CAs including paramagnetic metals (gadolinium), negative contrast agents such as superparamagnetic iron oxides and 19 F containing tracers, specifically for the in vivo tracking of MSCs using MRI. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Shehzahdi Shebbrin Moonshi
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia
| | - Yuao Wu
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia
| | - Hang Thu Ta
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia.,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland, Australia.,School of Environment and Science, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
8
|
Stueber DD, Villanova J, Aponte I, Xiao Z, Colvin VL. Magnetic Nanoparticles in Biology and Medicine: Past, Present, and Future Trends. Pharmaceutics 2021; 13:943. [PMID: 34202604 PMCID: PMC8309177 DOI: 10.3390/pharmaceutics13070943] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/27/2021] [Accepted: 06/16/2021] [Indexed: 12/23/2022] Open
Abstract
The use of magnetism in medicine has changed dramatically since its first application by the ancient Greeks in 624 BC. Now, by leveraging magnetic nanoparticles, investigators have developed a range of modern applications that use external magnetic fields to manipulate biological systems. Drug delivery systems that incorporate these particles can target therapeutics to specific tissues without the need for biological or chemical cues. Once precisely located within an organism, magnetic nanoparticles can be heated by oscillating magnetic fields, which results in localized inductive heating that can be used for thermal ablation or more subtle cellular manipulation. Biological imaging can also be improved using magnetic nanoparticles as contrast agents; several types of iron oxide nanoparticles are US Food and Drug Administration (FDA)-approved for use in magnetic resonance imaging (MRI) as contrast agents that can improve image resolution and information content. New imaging modalities, such as magnetic particle imaging (MPI), directly detect magnetic nanoparticles within organisms, allowing for background-free imaging of magnetic particle transport and collection. "Lab-on-a-chip" technology benefits from the increased control that magnetic nanoparticles provide over separation, leading to improved cellular separation. Magnetic separation is also becoming important in next-generation immunoassays, in which particles are used to both increase sensitivity and enable multiple analyte detection. More recently, the ability to manipulate material motion with external fields has been applied in magnetically actuated soft robotics that are designed for biomedical interventions. In this review article, the origins of these various areas are introduced, followed by a discussion of current clinical applications, as well as emerging trends in the study and application of these materials.
Collapse
Affiliation(s)
- Deanna D. Stueber
- Center for Biomedical Engineering, School of Engineering, Brown University, 171 Meeting Street, Providence, RI 02912, USA; (D.D.S.); (J.V.); (I.A.)
| | - Jake Villanova
- Center for Biomedical Engineering, School of Engineering, Brown University, 171 Meeting Street, Providence, RI 02912, USA; (D.D.S.); (J.V.); (I.A.)
- Department of Chemistry, Brown University, 324 Brook Street, Providence, RI 02912, USA;
| | - Itzel Aponte
- Center for Biomedical Engineering, School of Engineering, Brown University, 171 Meeting Street, Providence, RI 02912, USA; (D.D.S.); (J.V.); (I.A.)
| | - Zhen Xiao
- Department of Chemistry, Brown University, 324 Brook Street, Providence, RI 02912, USA;
| | - Vicki L. Colvin
- Center for Biomedical Engineering, School of Engineering, Brown University, 171 Meeting Street, Providence, RI 02912, USA; (D.D.S.); (J.V.); (I.A.)
- Department of Chemistry, Brown University, 324 Brook Street, Providence, RI 02912, USA;
| |
Collapse
|
9
|
Ghosh G, Panicker L. Protein-nanoparticle interactions and a new insight. SOFT MATTER 2021; 17:3855-3875. [PMID: 33885450 DOI: 10.1039/d0sm02050h] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The study of protein-nanoparticle interactions provides knowledge about the bio-reactivity of nanoparticles, and creates a database of nanoparticles for applications in nanomedicine, nanodiagnosis, and nanotherapy. The problem arises when nanoparticles come in contact with physiological fluids such as plasma or serum, wherein they interact with the proteins (or other biomolecules). This interaction leads to the coating of proteins on the nanoparticle surface, mostly due to the electrostatic interaction, called 'corona'. These proteins are usually partially unfolded. The protein corona can deter nanoparticles from their targeted functionalities, such as drug/DNA delivery at the site and fluorescence tagging of diseased tissues. The protein corona also has many repercussions on cellular intake, inflammation, accumulation, degradation, and clearance of the nanoparticles from the body depending on the exposed part of the proteins. Hence, the protein-nanoparticle interaction and the configuration of the bound-proteins on the nanosurface need thorough investigation and understanding. Several techniques such as DLS and zeta potential measurement, UV-vis spectroscopy, fluorescence spectroscopy, circular dichroism, FTIR, and DSC provide valuable information in the protein-nanoparticle interaction study. Besides, theoretical simulations also provide additional understanding. Despite a lot of research publications, the fundamental question remained unresolved. Can we aim for the application of functional nanoparticles in medicine? A new insight, given by us, in this article assumes a reasonable solution to this crucial question.
Collapse
Affiliation(s)
- Goutam Ghosh
- UGC-DAE Consortium for Scientific Research, Mumbai Centre, Mumbai 400 085, India.
| | | |
Collapse
|
10
|
Kiraga Ł, Kucharzewska P, Strzemecki D, Rygiel TP, Król M. Non-radioactive imaging strategies for in vivo immune cell tracking. PHYSICAL SCIENCES REVIEWS 2021; 8:385-403. [PMID: 36975764 PMCID: PMC10037928 DOI: 10.1515/psr-2020-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
In vivo tracking of administered cells chosen for specific disease treatment may be conducted by diagnostic imaging techniques preceded by cell labeling with special contrast agents. The most commonly used agents are those with radioactive properties, however their use in research is often impossible. This review paper focuses on the essential aspect of cell tracking with the exclusion of radioisotope tracers, therefore we compare application of different types of non-radioactive contrast agents (cell tracers), methods of cell labeling and application of various techniques for cell tracking, which are commonly used in preclinical or clinical studies. We discuss diagnostic imaging methods belonging to three groups: (1) Contrast-enhanced X-ray imaging, (2) Magnetic resonance imaging, and (3) Optical imaging. In addition, we present some interesting data from our own research on tracking immune cell with the use of discussed methods. Finally, we introduce an algorithm which may be useful for researchers planning leukocyte targeting studies, which may help to choose the appropriate cell type, contrast agent and diagnostic technique for particular disease study.
Collapse
Affiliation(s)
- Łukasz Kiraga
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
- Cellis AG, 80002 Zurich, Switzerland
| | - Paulina Kucharzewska
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
- Cellis AG, 80002 Zurich, Switzerland
| | | | - Tomasz P. Rygiel
- Cellis AG, 80002 Zurich, Switzerland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Magdalena Król
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
- Cellis AG, 80002 Zurich, Switzerland
| |
Collapse
|
11
|
Stephen ZR, Zhang M. Recent Progress in the Synergistic Combination of Nanoparticle-Mediated Hyperthermia and Immunotherapy for Treatment of Cancer. Adv Healthc Mater 2021; 10:e2001415. [PMID: 33236511 PMCID: PMC8034553 DOI: 10.1002/adhm.202001415] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has demonstrated great clinical success in certain cancers, driven primarily by immune checkpoint blockade and adoptive cell therapies. Immunotherapy can elicit strong, durable responses in some patients, but others do not respond, and to date immunotherapy has demonstrated success in only a limited number of cancers. To address this limitation, combinatorial approaches with chemo- and radiotherapy have been applied in the clinic. Extensive preclinical evidence suggests that hyperthermia therapy (HT) has considerable potential to augment immunotherapy with minimal toxicity. This progress report will provide a brief overview of immunotherapy and HT approaches and highlight recent progress in the application of nanoparticle (NP)-based HT in combination with immunotherapy. NPs allow for tumor-specific targeting of deep tissue tumors while potentially providing more even heating. NP-based HT increases tumor immunogenicity and tumor permeability, which improves immune cell infiltration and creates an environment more responsive to immunotherapy, particularly in solid tumors.
Collapse
Affiliation(s)
- Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, Department of Neurological Surgery, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
12
|
Chandrasekaran R, Madheswaran T, Tharmalingam N, Bose RJ, Park H, Ha DH. Labeling and tracking cells with gold nanoparticles. Drug Discov Today 2020; 26:94-105. [PMID: 33130336 DOI: 10.1016/j.drudis.2020.10.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/03/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022]
Abstract
Gold nanoparticles (AuNPs) have garnered much attention as contrast agents for computerized tomography (CT) because of their facile synthesis and surface functionalization, in addition to their significant X-ray attenuation and minimal cytotoxicity. Cell labeling using AuNPs and tracking of the labeled cells using CT has become a time-efficient and cost-effective method. Actively targeted AuNPs can enhance CT contrast and sensitivity, and further reduce the radiation dosage needed during CT imaging. In this review, we summarize the state-of-the-art use of AuNPs in CT for cell tracking, including the precautionary steps necessary for their use and the difficulty in translating the process into clinical use.
Collapse
Affiliation(s)
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, No. 126 Jalan Jalil Perkasa 19, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Nagendran Tharmalingam
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Rajendran Jc Bose
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea; Masonic Medical Research Institute, Utica, NY, USA
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea.
| | - Don-Hyung Ha
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
13
|
McGinley LM, Willsey MS, Kashlan ON, Chen KS, Hayes JM, Bergin IL, Mason SN, Stebbins AW, Kwentus JF, Pacut C, Kollmer J, Sakowski SA, Bell CB, Chestek CA, Murphy GG, Patil PG, Feldman EL. Magnetic resonance imaging of human neural stem cells in rodent and primate brain. Stem Cells Transl Med 2020; 10:83-97. [PMID: 32841522 PMCID: PMC7780819 DOI: 10.1002/sctm.20-0126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Stem cell transplantation therapies are currently under investigation for central nervous system disorders. Although preclinical models show benefit, clinical translation is somewhat limited by the absence of reliable noninvasive methods to confirm targeting and monitor transplanted cells in vivo. Here, we assess a novel magnetic resonance imaging (MRI) contrast agent derived from magnetotactic bacteria, magneto‐endosymbionts (MEs), as a translatable methodology for in vivo tracking of stem cells after intracranial transplantation. We show that ME labeling provides robust MRI contrast without impairment of cell viability or other important therapeutic features. Labeled cells were visualized immediately post‐transplantation and over time by serial MRI in nonhuman primate and mouse brain. Postmortem tissue analysis confirmed on‐target grft location, and linear correlations were observed between MRI signal, cell engraftment, and tissue ME levels, suggesting that MEs may be useful for determining graft survival or rejection. Overall, these findings indicate that MEs are an effective tool for in vivo tracking and monitoring of cell transplantation therapies with potential relevance to many cellular therapy applications.
Collapse
Affiliation(s)
- Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew S Willsey
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Osama N Kashlan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin S Chen
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - John M Hayes
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ingrid L Bergin
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shayna N Mason
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Aaron W Stebbins
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Crystal Pacut
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer Kollmer
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Caleb B Bell
- Bell Biosystems, San Francisco, California, USA.,G4S Capital & Ikigai Accelerator, Santa Clara, California, USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Electrical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Neuroscience and Robotics Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Geoffrey G Murphy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Parag G Patil
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Cuccione E, Chhour P, Si-Mohamed S, Dumot C, Kim J, Hubert V, Da Silva CC, Vandamme M, Chereul E, Balegamire J, Chevalier Y, Berthezène Y, Boussel L, Douek P, Cormode DP, Wiart M. Multicolor spectral photon counting CT monitors and quantifies therapeutic cells and their encapsulating scaffold in a model of brain damage. Nanotheranostics 2020; 4:129-141. [PMID: 32483519 PMCID: PMC7256015 DOI: 10.7150/ntno.45354] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 04/04/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale & aim: Various types of cell therapies are currently under investigation for the treatment of ischemic stroke patients. To bridge the gap between cell administration and therapeutic outcome, there is a need for non-invasive monitoring of these innovative therapeutic approaches. Spectral photon counting computed tomography (SPCCT) is a new imaging modality that may be suitable for cell tracking. SPCCT is the next generation of clinical CT that allows the selective visualization and quantification of multiple contrast agents. The aims of this study are: (i) to demonstrate the feasibility of using SPCCT to longitudinally monitor and quantify therapeutic cells, i.e. bone marrow-derived M2-polarized macrophages transplanted in rats with brain damage; and (ii) to evaluate the potential of this approach to discriminate M2-polarized macrophages from their encapsulating scaffold. Methods: Twenty one rats received an intralesional transplantation of bone marrow-derived M2-polarized macrophages. In the first set of experiments, cells were labeled with gold nanoparticles and tracked for up to two weeks post-injection in a monocolor study via gold K-edge imaging. In the second set of experiments, the same protocol was repeated for a bicolor study, in which the labeled cells are embedded in iodine nanoparticle-labeled scaffold. The amount of gold in the brain was longitudinally quantified using gold K-edge images reconstructed from SPCCT acquisition. Animals were sacrificed at different time points post-injection, and ICP-OES was used to validate the accuracy of gold quantification from SPCCT imaging. Results: The feasibility of therapeutic cell tracking was successfully demonstrated in brain-damaged rats with SPCCT imaging. The imaging modality enabled cell monitoring for up to 2 weeks post-injection, in a specific and quantitative manner. Differentiation of labeled cells and their embedding scaffold was also feasible with SPCCT imaging, with a detection limit as low as 5,000 cells in a voxel of 250 × 250 × 250 µm in dimension in vivo. Conclusion: Multicolor SPCCT is an innovative translational imaging tool that allows monitoring and quantification of therapeutic cells and their encapsulating scaffold transplanted in the damaged rat brain.
Collapse
Affiliation(s)
- Elisa Cuccione
- CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale U1060, INRA U1397, Université Lyon 1, INSA Lyon, F-69600 Oullins, France
- VOXCAN, 1 avenue Bourgelat, 69280 Marcy l'Etoile, France
| | - Peter Chhour
- Department of Radiology, University of Pennsylvania, Pennsylvania, United States
| | - Salim Si-Mohamed
- CREATIS, CNRS UMR 5220 - INSERM U1206 - University of Lyon 1 - INSA Lyon, Lyon, France
- Hospices Civils de Lyon, Radiology Department, Lyon, France
| | - Chloé Dumot
- CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale U1060, INRA U1397, Université Lyon 1, INSA Lyon, F-69600 Oullins, France
| | - Johoon Kim
- Department of Radiology, University of Pennsylvania, Pennsylvania, United States
| | - Violaine Hubert
- CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale U1060, INRA U1397, Université Lyon 1, INSA Lyon, F-69600 Oullins, France
| | - Claire Crola Da Silva
- CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale U1060, INRA U1397, Université Lyon 1, INSA Lyon, F-69600 Oullins, France
| | - Marc Vandamme
- VOXCAN, 1 avenue Bourgelat, 69280 Marcy l'Etoile, France
| | | | - Joëlle Balegamire
- LAGEPP, University of Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre, 69622 Villeurbanne, France
| | - Yves Chevalier
- LAGEPP, University of Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre, 69622 Villeurbanne, France
| | - Yves Berthezène
- CREATIS, CNRS UMR 5220 - INSERM U1206 - University of Lyon 1 - INSA Lyon, Lyon, France
- Hospices Civils de Lyon, Radiology Department, Lyon, France
| | - Loïc Boussel
- CREATIS, CNRS UMR 5220 - INSERM U1206 - University of Lyon 1 - INSA Lyon, Lyon, France
- Hospices Civils de Lyon, Radiology Department, Lyon, France
| | - Philippe Douek
- CREATIS, CNRS UMR 5220 - INSERM U1206 - University of Lyon 1 - INSA Lyon, Lyon, France
- Hospices Civils de Lyon, Radiology Department, Lyon, France
| | - David P. Cormode
- Department of Radiology, University of Pennsylvania, Pennsylvania, United States
| | - Marlène Wiart
- CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale U1060, INRA U1397, Université Lyon 1, INSA Lyon, F-69600 Oullins, France
| |
Collapse
|
15
|
Wang F, Wang Z, Wang F, Dong K, Zhang J, Sun YJ, Liu CF, Xing MJ, Cheng X, Wei S, Zheng JW, Zhao XF, Wang XM, Fu J, Song HF. Comparative strategies for stem cell biodistribution in a preclinical study. Acta Pharmacol Sin 2020; 41:572-580. [PMID: 31705124 PMCID: PMC7470780 DOI: 10.1038/s41401-019-0313-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
Abstract
Stem cell therapy represents the potential alternative effective strategy for some diseases that lack effective treatment currently. Correspondingly, it is crucial to establish high-sensitive and reliable quantification assay for tracing exogenous cell migration. In the present study, we first used both bioluminescence imaging (BLI) indirect labeling (human norepinephrine transporter-luciferase reporter system) and 89zirconium (89Zr)-hNSCs direct labeling combined with positron emission tomography/computer tomography (PET/CT) system for tracking human neural stem cells (hNSCs) migration into the brain via nasal administration in preclinical study. But the above two methods failed to give the biodistribution profile due to their low sensitivity. Considering its superior sensitivity and absolute quantitation capability, we developed and validated the droplet digital PCR (ddPCR) targeting species-specific gene in frozen and paraffin sections, slices, and whole blood with the sensitivity of 100–200 hNSCs. Accurate and high throughput quantification could be performed using ddPCR with the coefficient of variation (CVs) of lower quality control (LQC) below 30%. In combination with immunohistochemistry and ddPCR, we confirmed the migration of hNSCs into the brain via nasal administration, which supported the efficacy of hNSCs in MPTP-treated mice, an animal model of Parkinson’s disease. In conclusion, the present study is the first to report the application of ddPCR in the pharmacokinetics profile description of tracking of hNSCs in preclinical studies.
Collapse
|
16
|
Ali AAA, Shahror RA, Chen KY. Efficient Labeling Of Mesenchymal Stem Cells For High Sensitivity Long-Term MRI Monitoring In Live Mice Brains. Int J Nanomedicine 2020; 15:97-114. [PMID: 32021167 PMCID: PMC6955624 DOI: 10.2147/ijn.s211205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 11/08/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Regenerative medicine field is still lagging due to the lack of adequate knowledge regarding the homing of therapeutic cells towards disease sites, tracking of cells during treatment, and monitoring the biodistribution and fate of cells. Such necessities require labeling of cells with imaging agents that do not alter their biological characteristics, and development of suitable non-invasive imaging modalities. PURPOSE We aimed to develop, characterize, and standardize a facile labeling strategy for engineered mesenchymal stem cells without altering their viability, secretion of FGF21 protein (neuroprotective), and differentiation capabilities for non-invasive longitudinal MRI monitoring in live mice brains with high sensitivity. METHODS We compared the labeling efficiency of different commercial iron oxide nanoparticles towards our stem cells and determined the optimum labeling conditions using prussian blue staining, confocal microscopy, transmission electron microscopy, and flow cytometry. To investigate any change in biological characteristics of labeled cells, we tested their viability by WST-1 assay, expression of FGF21 by Western blot, and adipogenic and osteogenic differentiation capabilities. MRI contrast-enhancing properties of labeled cells were investigated in vitro using cell-agarose phantoms and in mice brains transplanted with the therapeutic stem cells. RESULTS We determined the nanoparticles that showed best labeling efficiency and least extracellular aggregation. We further optimized their labeling conditions (nanoparticles concentration and media supplementation) to achieve high cellular uptake and minimal extracellular aggregation of nanoparticles. Cell viability, expression of FGF21 protein, and differentiation capabilities were not impeded by nanoparticles labeling. Low number of labeled cells produced strong MRI signal decay in phantoms and in live mice brains which were visible for 4 weeks post transplantation. CONCLUSION We established a standardized magnetic nanoparticle labeling platform for stem cells that were monitored longitudinally with high sensitivity in mice brains using MRI for regenerative medicine applications.
Collapse
Affiliation(s)
- Ahmed Atef Ahmed Ali
- TMU Neuroscience Research Center – NeuroImage, College of Medicine, Taipei Medical University, Taipei110, Taiwan,Correspondence: Ahmed Atef Ahmed Ali Taipei Medical University, No. 250, Wuxing Street, Xinyi District, Taipei110, Taiwan ROCTel +886-2-2736-1661 ext 3215 Email
| | - Rami Ahmad Shahror
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei110, Taiwan,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei110, Taiwan
| | - Kai-Yun Chen
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei110, Taiwan,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei110, Taiwan
| |
Collapse
|
17
|
Fath-Bayati L, Vasei M, Sharif-Paghaleh E. Optical fluorescence imaging with shortwave infrared light emitter nanomaterials for in vivo cell tracking in regenerative medicine. J Cell Mol Med 2019; 23:7905-7918. [PMID: 31559692 PMCID: PMC6850965 DOI: 10.1111/jcmm.14670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/13/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
In vivo tracking and monitoring of adoptive cell transfer has a distinct importance in cell‐based therapy. There are many imaging modalities for in vivo monitoring of biodistribution, viability and effectiveness of transferred cells. Some of these procedures are not applicable in the human body because of low sensitivity and high possibility of tissue damages. Shortwave infrared region (SWIR) imaging is a relatively new technique by which deep biological tissues can be potentially visualized with high resolution at cellular level. Indeed, scanning of the electromagnetic spectrum (beyond 1000 nm) of SWIR has a great potential to increase sensitivity and resolution of in vivo imaging for various human tissues. In this review, molecular imaging modalities used for monitoring of biodistribution and fate of administered cells with focusing on the application of non‐invasive optical imaging at shortwave infrared region are discussed in detail.
Collapse
Affiliation(s)
- Leyla Fath-Bayati
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Department of Tissue Engineering, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Vasei
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Cell-based Therapies Research Institute, Digestive Disease Research Institute (DDRI), Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Ehsan Sharif-Paghaleh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Imaging Chemistry and Biology, Faculty of Life Sciences and Medicine, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
18
|
Bergenheim F, Seidelin JB, Pedersen MT, Mead BE, Jensen KB, Karp JM, Nielsen OH. Fluorescence-based tracing of transplanted intestinal epithelial cells using confocal laser endomicroscopy. Stem Cell Res Ther 2019; 10:148. [PMID: 31133056 PMCID: PMC6537188 DOI: 10.1186/s13287-019-1246-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022] Open
Abstract
Background Intestinal stem cell transplantation has been shown to promote mucosal healing and to engender fully functional epithelium in experimental colitis. Hence, stem cell therapies may provide an innovative approach to accomplish mucosal healing in patients with debilitating conditions such as inflammatory bowel disease. However, an approach to label and trace transplanted cells, in order to assess engraftment efficiency and to monitor wound healing, is a key hurdle to overcome prior to initiating human studies. Genetic engineering is commonly employed in animal studies, but may be problematic in humans due to potential off-target and long-term adverse effects. Methods We investigated the applicability of a panel of fluorescent dyes and nanoparticles to label intestinal organoids for visualization using the clinically approved imaging modality, confocal laser endomicroscopy (CLE). Staining homogeneity, durability, cell viability, differentiation capacity, and organoid forming efficiency were evaluated, together with visualization of labeled organoids in vitro and ex vivo using CLE. Results 5-Chloromethylfluorescein diacetate (CMFDA) proved to be suitable as it efficiently stained all organoids without transfer to unstained organoids in co-cultures. No noticeable adverse effects on viability, organoid growth, or stem cell differentiation capacity were observed, although single-cell reseeding revealed a dose-dependent reduction in organoid forming efficiency. Labeled organoids were easily identified in vitro using CLE for a duration of at least 3 days and could additionally be detected ex vivo following transplantation into murine experimental colitis. Conclusions It is highly feasible to use fluorescent dye-based labeling in combination with CLE to trace intestinal organoids following transplantation to confirm implantation at the intestinal target site. Electronic supplementary material The online version of this article (10.1186/s13287-019-1246-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fredrik Bergenheim
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark.
| | - Jakob B Seidelin
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark
| | | | - Benjamin E Mead
- Broad Institute of Massachusetts, Institute of Technology and Harvard University, Cambridge, MA, 02139, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kim B Jensen
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, DK-2200, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Medical and Health, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Jeffrey M Karp
- Broad Institute of Massachusetts, Institute of Technology and Harvard University, Cambridge, MA, 02139, USA.,Engineering in Medicine, Department of Medicine, Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, 02115, Boston, MA, USA.,Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, 02139, Cambridge, MA, USA
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark
| |
Collapse
|
19
|
Li W, Song W, Chen B, Matcher SJ. Superparamagnetic graphene quantum dot as a dual-modality contrast agent for confocal fluorescence microscopy and magnetomotive optical coherence tomography. JOURNAL OF BIOPHOTONICS 2019; 12:e201800219. [PMID: 30191684 DOI: 10.1002/jbio.201800219] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/04/2018] [Indexed: 05/03/2023]
Abstract
A magnetic graphene quantum dot (MGQD) nanoparticle, synthesized by hydrothermally reducing and cutting graphene oxide-iron oxide sheet, was demonstrated to possess the capabilities of simultaneous confocal fluorescence and magnetomotive optical coherence tomography (MMOCT) imaging. This MGQD shows low toxicity, significant tunable blue fluorescence and superparamagnetism, which can thus be used as a dual-modality contrast agent for confocal fluorescence microscopy (CFM) and MMOCT. The feasibility of applying MGQD as a tracer of cells is shown by imaging and visualizing MGQD labeled cells using CFM and our in-house MMOCT. Since MMOCT and CFM can offer anatomical structure and intracellular details, respectively, the MGQD for cell tracking could provide a more comprehensive diagnosis.
Collapse
Affiliation(s)
- Wei Li
- Department of Electronic and Electrical Engineering, The University of Sheffield, Sheffield, UK
| | - Wenxing Song
- Department of Chemical and Biological Engineering, The University of Sheffield, Sheffield, UK
| | - Biqiong Chen
- School of Mechanical and Aerospace Engineering, Queen's University Belfast, Belfast, UK
| | - Stephen J Matcher
- Department of Electronic and Electrical Engineering, The University of Sheffield, Sheffield, UK
| |
Collapse
|
20
|
Skachkov I, Luan Y, van Tiel ST, van der Steen AFW, de Jong N, Bernsen MR, Kooiman K. SPIO labeling of endothelial cells using ultrasound and targeted microbubbles at diagnostic pressures. PLoS One 2018; 13:e0204354. [PMID: 30235336 PMCID: PMC6147550 DOI: 10.1371/journal.pone.0204354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023] Open
Abstract
In vivo cell tracking of therapeutic, tumor, and endothelial cells is an emerging field and a promising technique for imaging cardiovascular disease and cancer development. Site-specific labeling of endothelial cells with the MRI contrast agent superparamagnetic iron oxide (SPIO) in the absence of toxic agents is challenging. Therefore, the aim of this in vitro study was to find optimal parameters for efficient and safe SPIO-labeling of endothelial cells using ultrasound-activated CD31-targeted microbubbles for future MRI tracking. Ultrasound at a frequency of 1 MHz (10,000 cycles, repetition rate of 20 Hz) was used for varying applied peak negative pressures (10–160 kPa, i.e. low mechanical index (MI) of 0.01–0.16), treatment durations (0–30 s), time of SPIO addition (-5 min– 15 min with respect to the start of the ultrasound), and incubation time after SPIO addition (5 min– 3 h). Iron specific Prussian Blue staining in combination with calcein-AM based cell viability assays were applied to define the most efficient and safe conditions for SPIO-labeling. Optimal SPIO labeling was observed when the ultrasound parameters were 40 kPa peak negative pressure (MI 0.04), applied for 30 s just before SPIO addition (0 min). Compared to the control, this resulted in an approximate 12 times increase of SPIO uptake in endothelial cells in vitro with 85% cell viability. Therefore, ultrasound-activated targeted ultrasound contrast agents show great potential for effective and safe labeling of endothelial cells with SPIO.
Collapse
Affiliation(s)
- Ilya Skachkov
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Ying Luan
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Sandra T. van Tiel
- Department of Radiology & Nucleair Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Antonius F. W. van der Steen
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Nico de Jong
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Monique R. Bernsen
- Department of Radiology & Nucleair Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- * E-mail:
| |
Collapse
|
21
|
Grunert B, Saatz J, Hoffmann K, Appler F, Lubjuhn D, Jakubowski N, Resch-Genger U, Emmerling F, Briel A. Multifunctional Rare-Earth Element Nanocrystals for Cell Labeling and Multimodal Imaging. ACS Biomater Sci Eng 2018; 4:3578-3587. [DOI: 10.1021/acsbiomaterials.8b00495] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
| | - Jessica Saatz
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany
| | - Katrin Hoffmann
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany
| | | | - Dominik Lubjuhn
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany
| | - Norbert Jakubowski
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany
| | - Ute Resch-Genger
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany
| | - Franziska Emmerling
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Straße 11, 12489 Berlin, Germany
| | | |
Collapse
|
22
|
Masthoff M, Gran S, Zhang X, Wachsmuth L, Bietenbeck M, Helfen A, Heindel W, Sorokin L, Roth J, Eisenblätter M, Wildgruber M, Faber C. Temporal window for detection of inflammatory disease using dynamic cell tracking with time-lapse MRI. Sci Rep 2018; 8:9563. [PMID: 29934611 PMCID: PMC6015069 DOI: 10.1038/s41598-018-27879-z] [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: 02/20/2018] [Accepted: 06/12/2018] [Indexed: 12/20/2022] Open
Abstract
Time-lapse MRI was implemented for dynamic non-invasive cell tracking of individual slowly moving intravascular immune cells. Repetitive MRI acquisition enabled dynamic observation of iron oxide nanoparticle (ION) labelled cells. Simulations of MRI contrast indicated that only cells moving slower than 1 µm/s were detectable. Time-lapse MRI of the brain was performed after either IONs or ION-labelled monocytes were injected intravenously into naïve and experimental autoimmune encephalomyelitis (EAE) bearing mice at a presymptomatic or symptomatic stage. EAE mice showed a reduced number of slow moving, i.e. patrolling cells before and after onset of symptoms as compared to naïve controls. This observation is consistent with the notion of altered cell dynamics, i.e. higher velocities of immune cells rolling along the endothelium in the inflamed condition. Thus, time-lapse MRI enables for assessing immune cell dynamics non-invasively in deep tissue and may serve as a tool for detection or monitoring of an inflammatory response.
Collapse
Affiliation(s)
- Max Masthoff
- Translational Research Imaging Center, Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| | - Sandra Gran
- Institute for Immunology, University of Muenster, Roentgenstraße 21, 48149, Muenster, Germany
| | - Xueli Zhang
- Institute for Physiological Chemistry and Pathobiochemistry, University of Muenster, Waldeyerstraße 15, 48149, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Waldeyerstraße 15, 48149, Muenster, Germany
| | - Lydia Wachsmuth
- Translational Research Imaging Center, Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| | - Michael Bietenbeck
- Translational Research Imaging Center, Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| | - Anne Helfen
- Translational Research Imaging Center, Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| | - Walter Heindel
- Translational Research Imaging Center, Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| | - Lydia Sorokin
- Institute for Physiological Chemistry and Pathobiochemistry, University of Muenster, Waldeyerstraße 15, 48149, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Waldeyerstraße 15, 48149, Muenster, Germany
| | - Johannes Roth
- Institute for Immunology, University of Muenster, Roentgenstraße 21, 48149, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Waldeyerstraße 15, 48149, Muenster, Germany
| | - Michel Eisenblätter
- Translational Research Imaging Center, Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany.,Division of Imaging Sciences & Biomedical Engineering, King's College London, London, UK
| | - Moritz Wildgruber
- Translational Research Imaging Center, Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Waldeyerstraße 15, 48149, Muenster, Germany
| | - Cornelius Faber
- Translational Research Imaging Center, Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany. .,Cells-in-Motion Cluster of Excellence, University of Muenster, Waldeyerstraße 15, 48149, Muenster, Germany.
| |
Collapse
|
23
|
Chhour P, Kim J, Benardo B, Tovar A, Mian S, Litt HI, Ferrari VA, Cormode DP. Effect of Gold Nanoparticle Size and Coating on Labeling Monocytes for CT Tracking. Bioconjug Chem 2017; 28:260-269. [PMID: 28095688 PMCID: PMC5462122 DOI: 10.1021/acs.bioconjchem.6b00566] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
With advances in cell therapies, interest in cell tracking techniques to monitor the migration, localization, and viability of these cells continues to grow. X-ray computed tomography (CT) is a cornerstone of medical imaging but has been limited in cell tracking applications due to its low sensitivity toward contrast media. In this study, we investigate the role of size and surface functionality of gold nanoparticles for monocyte uptake to optimize the labeling of these cells for tracking in CT. We synthesized gold nanoparticles (AuNP) that range from 15 to 150 nm in diameter and examined several capping ligands, generating 44 distinct AuNP formulations. In vitro cytotoxicity and uptake experiments were performed with the RAW 264.7 monocyte cell line. The majority of formulations at each size were found to be biocompatible, with only certain 150 nm PEG functionalized particles reducing viability at high concentrations. High uptake of AuNP was found using small capping ligands with distal carboxylic acids (11-MUA and 16-MHA). Similar uptake values were found with intermediate sizes (50 and 75 nm) of AuNP when coated with 2000 MW poly(ethylene-glycol) carboxylic acid ligands (PCOOH). Low uptake values were observed with 15, 25, 100, and 150 nm PCOOH AuNP, revealing interplay between size and surface functionality. Transmission electron microscopy (TEM) and CT performed on cells revealed similar patterns of high gold uptake for 50 nm PCOOH and 75 nm PCOOH AuNP. These results demonstrate that highly negatively charged carboxylic acid coatings for AuNP provide the greatest internalization of AuNP in monocytes, with a complex dependency on size.
Collapse
Affiliation(s)
- Peter Chhour
- Department of Radiology, ‡Department of Bioengineering, and §Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine of the University of Pennsylvania , 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Johoon Kim
- Department of Radiology, ‡Department of Bioengineering, and §Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine of the University of Pennsylvania , 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Barbara Benardo
- Department of Radiology, ‡Department of Bioengineering, and §Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine of the University of Pennsylvania , 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Alfredo Tovar
- Department of Radiology, ‡Department of Bioengineering, and §Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine of the University of Pennsylvania , 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Shaameen Mian
- Department of Radiology, ‡Department of Bioengineering, and §Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine of the University of Pennsylvania , 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Harold I Litt
- Department of Radiology, ‡Department of Bioengineering, and §Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine of the University of Pennsylvania , 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Victor A Ferrari
- Department of Radiology, ‡Department of Bioengineering, and §Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine of the University of Pennsylvania , 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - David P Cormode
- Department of Radiology, ‡Department of Bioengineering, and §Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine of the University of Pennsylvania , 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
24
|
Rhodamine bound maghemite as a long-term dual imaging nanoprobe of adipose tissue-derived mesenchymal stromal cells. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 46:433-444. [DOI: 10.1007/s00249-016-1187-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 10/25/2016] [Accepted: 11/02/2016] [Indexed: 01/09/2023]
|
25
|
Ishihara K, Chen W, Liu Y, Tsukamoto Y, Inoue Y. Cytocompatible and multifunctional polymeric nanoparticles for transportation of bioactive molecules into and within cells. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2016; 17:300-312. [PMID: 27877883 PMCID: PMC5111563 DOI: 10.1080/14686996.2016.1190257] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/21/2016] [Accepted: 05/12/2016] [Indexed: 05/23/2023]
Abstract
Multifunctional polymeric nanoparticles are materials with great potential for a wide range of biomedical applications. For progression in this area of research, unfavorable interactions of these nanoparticles with proteins and cells must be avoided in biological environments, for example, through treatment of the nanoparticle surfaces. Construction of an artificial cell membrane structure based on polymers bearing the zwitterionic phosphorylcholine group can prevent biological reactions at the surface effectively. In addition, certain bioactive molecules can be immobilized on the surface of the polymer to generate enough affinity to capture target biomolecules. Furthermore, entrapment of inorganic nanoparticles inside polymeric matrices enhances the nanoparticle functionality significantly. This review summarizes the preparation and characterization of cytocompatible and multifunctional polymeric nanoparticles; it analyzes the efficiency of their fluorescence function, the nature of the artificial cell membrane structure, and their performance as in-cell devices; and finally, it evaluates both their chemical reactivity and effects in cells.
Collapse
Affiliation(s)
- Kazuhiko Ishihara
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Weixin Chen
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yihua Liu
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yuriko Tsukamoto
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yuuki Inoue
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
26
|
Ramos-Gómez M, Martínez-Serrano A. Tracking of iron-labeled human neural stem cells by magnetic resonance imaging in cell replacement therapy for Parkinson's disease. Neural Regen Res 2016; 11:49-52. [PMID: 26981077 PMCID: PMC4774222 DOI: 10.4103/1673-5374.169628] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human neural stem cells (hNSCs) derived from the ventral mesencephalon are powerful research tools and candidates for cell therapies in Parkinson's disease. However, their clinical translation has not been fully realized due, in part, to the limited ability to track stem cell regional localization and survival over long periods of time after in vivo transplantation. Magnetic resonance imaging provides an excellent non-invasive method to study the fate of transplanted cells in vivo. For magnetic resonance imaging cell tracking, cells need to be labeled with a contrast agent, such as magnetic nanoparticles, at a concentration high enough to be easily detected by magnetic resonance imaging. Grafting of human neural stem cells labeled with magnetic nanoparticles allows cell tracking by magnetic resonance imaging without impairment of cell survival, proliferation, self-renewal, and multipotency. However, the results reviewed here suggest that in long term grafting, activated microglia and macrophages could contribute to magnetic resonance imaging signal by engulfing dead labeled cells or iron nanoparticles dispersed freely in the brain parenchyma over time.
Collapse
Affiliation(s)
- Milagros Ramos-Gómez
- Centre for Biomedical Technology, Polytechnic University of Madrid, Madrid, Spain; Biomedical Research Networking Center of Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Alberto Martínez-Serrano
- Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa", Autonomous University of Madrid-C.S.I.C, Madrid, Spain
| |
Collapse
|