1
|
Ghazi R, Ibrahim TK, Nasir JA, Gai S, Ali G, Boukhris I, Rehman Z. Iron oxide based magnetic nanoparticles for hyperthermia, MRI and drug delivery applications: a review. RSC Adv 2025; 15:11587-11616. [PMID: 40230636 PMCID: PMC11995399 DOI: 10.1039/d5ra00728c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
Iron-oxide nanoparticles (IONPs) have garnered substantial attention in both research and technological domains due to their exceptional chemical and physical properties. These nanoparticles have mitigated the adverse effects of conventional treatment procedures by facilitating advanced theranostic approaches in integration with biomedicine. These IONPs have been extensively utilized in MRI (as contrast agents in diagnosis), drug delivery (as drug carriers), and hyperthermia (treatment), demonstrating promising results with potential for further enhancement. This study elucidates the operational principles of these NPs during diagnosis, drug delivery, and treatment, and emphasizes their precision and efficacy in transporting therapeutic agents to targeted sites without drug loss. It also analyses various challenges associated with the application of these IONPs in this field, such as biocompatibility, agglomeration, and toxicity. Furthermore, diverse strategies have been delineated to address these challenges. Overall, this review provides a comprehensive overview of the applications of IONPs in the field of biomedicine and treatment, along with the associated challenges. It offers significant assistance to researchers, professionals, and clinicians in the field of biomedicine.
Collapse
Affiliation(s)
- Rizwana Ghazi
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-(051)90642241 +92-(051)90642245
| | - Talib K Ibrahim
- Department of Petroleum Engineering, College of Engineering, Knowledge University Erbil Iraq
- Department of Petroleum Engineering, Al-Kitab University Altun Kupri Iraq
| | - Jamal Abdul Nasir
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-(051)90642241 +92-(051)90642245
| | - Shili Gai
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University Harbin 150001 P. R. China
| | - Ghafar Ali
- Nanomaterials Research Group (NRG), Physics Division, PINSTECH Nilore Islamabad Pakistan
| | - Imed Boukhris
- Department of Physics, Faculty of Science, King Khalid University P. O. Box 9004 Abha Saudi Arabia
| | - Ziaur Rehman
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-(051)90642241 +92-(051)90642245
| |
Collapse
|
2
|
Aknine N, Pelletier R, Klymchenko AS. Lipid-Directed Covalent Labeling of Plasma Membranes for Long-Term Imaging, Barcoding and Manipulation of Cells. JACS AU 2025; 5:922-936. [PMID: 40017781 PMCID: PMC11863151 DOI: 10.1021/jacsau.4c01134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 03/01/2025]
Abstract
Fluorescent probes for cell plasma membranes (PM) generally exploit a noncovalent labeling mechanism, which constitutes a fundamental limitation in multiple bioimaging applications. Here, we report a concept of lipid-directed covalent labeling of PM, which exploits transient binding to the lipid membrane surface generating a high local dye concentration, thus favoring covalent ligation to random proximal membrane proteins. This concept yielded fluorescent probes for PM called MemGraft, which are built of a dye (cyanine Cy3 or Cy5) bearing a low-affinity membrane anchor and a reactive group: an activated ester or a maleimide. In contrast to specially designed control dyes and commercial Cy3-based labels of amino or thiol groups, MemGraft probes stain efficiently PM, revealing the crucial role of the membrane anchor combined with optimal reactivity of the activated ester or the maleimide. MemGraft probes overcome existing limitations of noncovalent probes, which makes them compatible with cell fixation, permeabilization, trypsinization, and the presence of serum. The latter allows long-term cell tracking and video imaging of cell PM dynamics without the signs of phototoxicity. The covalent strategy also enables staining and long-term tracking of cocultured cells labeled in different colors without exchange of probes. Moreover, the combination of MemGraft-Cy3 and MemGraft-Cy5 probes at different ratios enabled long-term cell barcoding in at least 5 color codes, important for tracking and visualizing multiple populations of cells. Ultimately, we found that the MemGraft strategy enables efficient biotinylation of the cell surface, opening the path to cell surface engineering and cell manipulation.
Collapse
Affiliation(s)
- Nathan Aknine
- Laboratoire de Bioimagerie
et Pathologies, UMR 7021 CNRS, ITI SysChem-Chimie des Systèmes
Complexes, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Remi Pelletier
- Laboratoire de Bioimagerie
et Pathologies, UMR 7021 CNRS, ITI SysChem-Chimie des Systèmes
Complexes, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Andrey S. Klymchenko
- Laboratoire de Bioimagerie
et Pathologies, UMR 7021 CNRS, ITI SysChem-Chimie des Systèmes
Complexes, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| |
Collapse
|
3
|
Rao Z, Cao D, Geng F, Huang H, Kang Y. Determination of the Localized Surface Plasmon Resonance Alteration of AgNPs via Multiwavelength Evanescent Scattering Microscopy for Pb(II) Detection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37981-37993. [PMID: 39007740 DOI: 10.1021/acsami.4c05900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
We developed multiwavelength evanescent scattering microscopy (MWESM), which can acquire plasmonic nanoparticle images at the particle level using the evanescent field as the incident source and distinguish different LSPR (localized surface plasmon resonance) spectral peaks among four wavelengths. Our microscope could be easily and simply built by modifying a commercial total internal reflection fluorescence microscope (TIRFM) with the substitution of a beamsplitter and the addition of a semicircular stop. The ultrathin depth of illumination and rejection of the reflected incident source together contribute to the high sensitivity and contrast of single nanoparticle imaging. We first validated the capability of our imaging system in distinguishing plasmonic nanoparticles bearing different LSPR spectral peaks, and the results were consistent with the scattering spectra results of hyperspectral imaging. Moreover, we demonstrated high imaging quality from the aspects of the signal/noise ratio and point spread function of the single-particle images. Meaningfully, the system can be utilized in rapidly determining the concentration of toxic lead ions in environmental and biological samples with good linearity and sensitivity, based on single-particle evanescent scattering imaging through the detection of the alteration of the LSPR of silver nanoparticles. This system holds the potential to advance the field of nanoparticle imaging and foster the application of nanomaterials as sensors.
Collapse
Affiliation(s)
- Ziyu Rao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, People's Republic of China
| | - Dong Cao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, People's Republic of China
| | - Fanglan Geng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, People's Republic of China
| | - Honglin Huang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, People's Republic of China
| | - Yuehui Kang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, People's Republic of China
| |
Collapse
|
4
|
Wang X, Song B, Wang Z, Qin L, Liang W. The innovative design of a delivery and real-time tracer system for anti-encephalitis drugs that can penetrate the blood-brain barrier. J Control Release 2023; 363:136-148. [PMID: 37742845 DOI: 10.1016/j.jconrel.2023.09.043] [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: 07/05/2023] [Revised: 09/09/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
As a "wall" between blood flow and brain cells, the blood-brain barrier (BBB) makes it really difficult for drugs to cross this barrier and work. This is particularly the case for pharmaceuticals of acute encephalitis therapies, largely excluded from the brain following systemic administration. Herein we report an advanced drug delivery system that can cross the BBB and target acute inflammation based on the controlled release of macrophage-camouflaged glow nanoparticles via a Trojan horse strategy. Benefiting from afterglow imaging that eliminates background interference and RAW 264.7 cells (RAW) with special immune homing and long-term tracking capabilities, polydopamine (PDA)-modified afterglow nanoparticles (ANPs) as near-infrared photo-responsive drug carriers in a controlled delivery system camouflaged by macrophages can penetrate the BBB by crossing the intercellular space and trigger the anti-inflammatory drug by photothermal conversion in the brain parenchyma dexamethasone (Dex) release, exhibiting good acute inflammation recognition and healing ability. APD@RAW was monitored to cross the BBB and image deep brain inflamed areas in a model of acute brain inflammation. Meanwhile, the delivered Dex mitigated the brain damage caused by inflammatory cytokines secretion (IL-6, TNF-α, and IL-1β). Overall, this drug delivery system holds excellent potential for BBB penetrating and acute encephalitis therapies.
Collapse
Affiliation(s)
- Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan 250117, Shandong, China
| | - Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan 250117, Shandong, China
| | - Zixuan Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan 250117, Shandong, China
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan 250117, Shandong, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan 250117, Shandong, China.
| |
Collapse
|
5
|
Ouyang X, Jia N, Luo J, Li L, Xue J, Bu H, Xie G, Wan Y. DNA Nanoribbon-Assisted Intracellular Biosynthesis of Fluorescent Gold Nanoclusters for Cancer Cell Imaging. JACS AU 2023; 3:2566-2577. [PMID: 37772173 PMCID: PMC10523492 DOI: 10.1021/jacsau.3c00365] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 09/30/2023]
Abstract
Metal nanoclusters (NCs) have emerged as a promising class of fluorescent probes for cellular imaging due to their high resistance to photobleaching and low toxicity. Nevertheless, their widespread use in clinical diagnosis is limited by their unstable intracellular fluorescence. In this study, we develop an intracellularly biosynthesized fluorescent probe, DNA nanoribbon-gold NCs (DNR/AuNCs), for long-term cellular tracking. Our results show that DNR/AuNCs exhibit a 4-fold enhancement of intracellular fluorescence intensity compared to free AuNCs. We also investigated the mechanism underlying the fluorescence enhancement of AuNCs by DNRs. Our findings suggest that the higher synthesis efficiency and stability of AuNCs in the lysosome may contribute to their fluorescence enhancement, which enables long-term (up to 15 days) fluorescence imaging of cancer cells (enhancement of ∼60 times compared to free AuNCs). Furthermore, we observe similar results with other metal NCs, confirming the generality of the DNR-assisted biosynthesis approach for preparing highly bright and stable fluorescent metal NCs for cancer cell imaging.
Collapse
Affiliation(s)
- Xiangyuan Ouyang
- Xi’an
Key Laboratory of Functional Supramolecular Structure and Materials,
Key Laboratory of Synthetic and Natural Functional Molecule of Ministry
of Education, College of Chemistry & Materials Science, Northwest University, Xi’an, Shaanxi 710127, P. R. China
| | - Nan Jia
- Xi’an
Key Laboratory of Functional Supramolecular Structure and Materials,
Key Laboratory of Synthetic and Natural Functional Molecule of Ministry
of Education, College of Chemistry & Materials Science, Northwest University, Xi’an, Shaanxi 710127, P. R. China
| | - Jing Luo
- Key
Laboratory of Resource Biology and Biotechnology in Western China
(Ministry of Education), College of Life Sciences, Northwest University, Xi’an, Shaanxi 710069, PR China
| | - Le Li
- Xi’an
Key Laboratory of Functional Supramolecular Structure and Materials,
Key Laboratory of Synthetic and Natural Functional Molecule of Ministry
of Education, College of Chemistry & Materials Science, Northwest University, Xi’an, Shaanxi 710127, P. R. China
| | - Jiangshan Xue
- Key
Laboratory of Resource Biology and Biotechnology in Western China
(Ministry of Education), College of Life Sciences, Northwest University, Xi’an, Shaanxi 710069, PR China
| | - Huaiyu Bu
- Key
Laboratory of Resource Biology and Biotechnology in Western China
(Ministry of Education), College of Life Sciences, Northwest University, Xi’an, Shaanxi 710069, PR China
| | - Gang Xie
- Xi’an
Key Laboratory of Functional Supramolecular Structure and Materials,
Key Laboratory of Synthetic and Natural Functional Molecule of Ministry
of Education, College of Chemistry & Materials Science, Northwest University, Xi’an, Shaanxi 710127, P. R. China
| | - Ying Wan
- School
of Mechanical Engineering, Nanjing University
of Science and Technology, Nanjing 210094, China
| |
Collapse
|
6
|
Kommineni N, Chaudhari R, Conde J, Tamburaci S, Cecen B, Chandra P, Prasad R. Engineered Liposomes in Interventional Theranostics of Solid Tumors. ACS Biomater Sci Eng 2023; 9:4527-4557. [PMID: 37450683 DOI: 10.1021/acsbiomaterials.3c00510] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Engineered liposomal nanoparticles have unique characteristics as cargo carriers in cancer care and therapeutics. Liposomal theranostics have shown significant progress in preclinical and clinical cancer models in the past few years. Liposomal hybrid systems have not only been approved by the FDA but have also reached the market level. Nanosized liposomes are clinically proven systems for delivering multiple therapeutic as well as imaging agents to the target sites in (i) cancer theranostics of solid tumors, (ii) image-guided therapeutics, and (iii) combination therapeutic applications. The choice of diagnostics and therapeutics can intervene in the theranostics property of the engineered system. However, integrating imaging and therapeutics probes within lipid self-assembly "liposome" may compromise their overall theranostics performance. On the other hand, liposomal systems suffer from their fragile nature, site-selective tumor targeting, specific biodistribution and premature leakage of loaded cargo molecules before reaching the target site. Various engineering approaches, viz., grafting, conjugation, encapsulations, etc., have been investigated to overcome the aforementioned issues. It has been studied that surface-engineered liposomes demonstrate better tumor selectivity and improved therapeutic activity and retention in cells/or solid tumors. It should be noted that several other parameters like reproducibility, stability, smooth circulation, toxicity of vital organs, patient compliance, etc. must be addressed before using liposomal theranostics agents in solid tumors or clinical models. Herein, we have reviewed the importance and challenges of liposomal medicines in targeted cancer theranostics with their preclinical and clinical progress and a translational overview.
Collapse
Affiliation(s)
- Nagavendra Kommineni
- Center for Biomedical Research, Population Council, New York, New York 10065, United States
| | - Ruchita Chaudhari
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa; Lisboa 1169-056, Portugal
| | - Sedef Tamburaci
- Department of Chemical Engineering, Izmir Institute of Technology, Gulbahce Campus, Izmir 35430, Turkey
| | - Berivan Cecen
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rajendra Prasad
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| |
Collapse
|
7
|
Zheng J, Jiang X, Li Y, Gao J. Inorganic nanoparticle-integrated mesenchymal stem cells: A potential biological agent for multifaceted applications. MedComm (Beijing) 2023; 4:e313. [PMID: 37533768 PMCID: PMC10390757 DOI: 10.1002/mco2.313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 08/04/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based therapies are flourishing. MSCs could be used as potential therapeutic agents for regenerative medicine due to their own repair function. Meanwhile, the natural predisposition toward inflammation or injury sites makes them promising carriers for targeted drug delivery. Inorganic nanoparticles (INPs) are greatly favored for their unique properties and potential applications in biomedical fields. Current research has integrated INPs with MSCs to enhance their regenerative or antitumor functions. This model also allows the in vivo fate tracking of MSCs in multiple imaging modalities, as many INPs are also excellent contrast agents. Thus, INP-integrated MSCs would be a multifunctional biologic agent with great potential. In this review, the current roles performed by the integration of INPs with MSCs, including (i) enhancing their repair and regeneration capacity via the improvement of migration, survival, paracrine, or differentiation properties, (ii) empowering tumor-killing ability through agent loaded or hyperthermia, and (iii) conferring traceability are summarized. An introduction of INP-integrated MSCs for simultaneous treatment and tracking is also included. The promising applications of INP-integrated MSCs in future treatments are emphasized and the challenges to their clinical translation are discussed.
Collapse
Affiliation(s)
- Juan‐Juan Zheng
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Xin‐Chi Jiang
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Yao‐Sheng Li
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Jian‐Qing Gao
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Hangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
8
|
Abstract
Cancer is still a serious health problem globally. Conventional therapies have adverse effects, which affect human life quality. Tumor microenvironment (TME), also known as surrounding stroma, has a contributory role in cancer development. Understanding the interaction between TME and cancer progression is a challenge and helps to develop new therapeutic strategies that neutralize the tracks taken by cancer cells to grow, spread, and resist therapy. Therefore, targeting TME components may be effective in improving tumor therapy. Using nanotechnology for drug delivery is of great interest, where it overcomes some obstacles such as solubility and absorption of drugs and delivering them to the appropriate place of action. The main target of nanotechnology for drug delivery is the ability to differentiate between normal and cancer cells. It can be concluded that TME is an important complementary strategy for the development of anticancer drugs. Multitargeted therapy has better efficient potential than individual therapy against cancer.
Collapse
|
9
|
Bunzen H, Jirák D. Recent Advances in Metal-Organic Frameworks for Applications in Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50445-50462. [PMID: 36239348 PMCID: PMC10749454 DOI: 10.1021/acsami.2c10272] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
Diagnostics is an important part of medical practice. The information required for diagnosis is typically collected by performing diagnostic tests, some of which include imaging. Magnetic resonance imaging (MRI) is one of the most widely used and effective imaging techniques. To improve the sensitivity and specificity of MRI, contrast agents are used. In this review, the usage of metal-organic frameworks (MOFs) and composite materials based on them as contrast agents for MRI is discussed. MOFs are crystalline porous coordination polymers. Due to their huge design variety and high density of metal ions, they have been studied as a highly promising class of materials for developing MRI contrast agents. This review highlights the most important studies and focuses on the progress of the field over the last five years. The materials are classified based on their design and structural properties into three groups: MRI-active MOFs, composite materials based on MOFs, and MRI-active compounds loaded in MOFs. Moreover, an overview of MOF-based materials for heteronuclear MRI including 129Xe and 19F MRI is given.
Collapse
Affiliation(s)
- Hana Bunzen
- Chair
of Solid State and Materials Chemistry, Institute of Physics, University of Augsburg, Universitätsstraße 1, D-86159 Augsburg, Germany
| | - Daniel Jirák
- Department
of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Vídeňská1958/9, 140 21 Prague 4, Czech Republic
| |
Collapse
|
10
|
Clases D, Gonzalez de Vega R. Facets of ICP-MS and their potential in the medical sciences-Part 2: nanomedicine, immunochemistry, mass cytometry, and bioassays. Anal Bioanal Chem 2022; 414:7363-7386. [PMID: 36042038 PMCID: PMC9427439 DOI: 10.1007/s00216-022-04260-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022]
Abstract
Inductively coupled-plasma mass spectrometry (ICP-MS) has transformed our knowledge on the role of trace and major elements in biology and has emerged as the most versatile technique in elemental mass spectrometry. The scope of ICP-MS has dramatically changed since its inception, and nowadays, it is a mature platform technology that is compatible with chromatographic and laser ablation (LA) systems. Over the last decades, it kept pace with various technological advances and was inspired by interdisciplinary approaches which endorsed new areas of applications. While the first part of this review was dedicated to fundamentals in ICP-MS, its hyphenated techniques and the application in biomonitoring, isotope ratio analysis, elemental speciation analysis, and elemental bioimaging, this second part will introduce relatively current directions in ICP-MS and their potential to provide novel perspectives in the medical sciences. In this context, current directions for the characterisation of novel nanomaterials which are considered for biomedical applications like drug delivery and imaging platforms will be discussed while considering different facets of ICP-MS including single event analysis and dedicated hyphenated techniques. Subsequently, immunochemistry techniques will be reviewed in their capability to expand the scope of ICP-MS enabling analysis of a large range of biomolecules alongside elements. These methods inspired mass cytometry and imaging mass cytometry and have the potential to transform diagnostics and treatment by offering new paradigms for personalised medicine. Finally, the interlacing of immunochemistry methods, single event analysis, and functional nanomaterials has opened new horizons to design novel bioassays which promise potential as assets for clinical applications and larger screening programs and will be discussed in their capabilities to detect low-level proteins and nucleic acids.
Collapse
Affiliation(s)
- David Clases
- Nano Mirco LAB, Institute of Chemistry, University of Graz, Graz, Austria.
| | | |
Collapse
|
11
|
Hidangmayum A, Debnath A, Guru A, Singh BN, Upadhyay SK, Dwivedi P. Mechanistic and recent updates in nano-bioremediation for developing green technology to alleviate agricultural contaminants. INTERNATIONAL JOURNAL OF ENVIRONMENTAL SCIENCE AND TECHNOLOGY : IJEST 2022; 20:1-26. [PMID: 36196301 PMCID: PMC9521565 DOI: 10.1007/s13762-022-04560-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/29/2022] [Accepted: 09/17/2022] [Indexed: 05/09/2023]
Abstract
The rise in environmental pollutant levels in recent years is mostly attributable to anthropogenic activities such as industrial, agricultural and other activities. Additionally, these activities may produce excessive levels of dangerous toxicants such as heavy metals, organic pollutants including pesticide and herbicide chemicals, and sewage discharges from residential and commercial sources. With a focus on environmentally friendly, sustainable technology, new technologies such as combined process of nanotechnology and bioremediation are urgently needed to accelerate the cost-effective remediation process to alleviate toxic contaminants than the conventional remediation methods. Numerous studies have shown that nanoparticles possess special qualities including improved catalysis and adsorption as well as increased reactivity. Currently, microorganisms and their extracts are being used as promising, environmentally friendly catalysts for engineered nanomaterial. In the long term, this combination of both technologies called nano-bioremediation may significantly alter the field of environmental remediation since it is more intelligent, safe, environmentally friendly, economical and green. This review provides an overview of soil and water remediation techniques as well as the use of nano-bioremediation, which is made from various living organisms. Additionally, current developments related to the mechanism, model and kinetic studies for remediation of agricultural contaminants have been discussed.
Collapse
Affiliation(s)
- A Hidangmayum
- Department of Plant Physiology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - A Debnath
- Department of Civil Engineering, Indian Institute of Technology (BHU), Varanasi, India
| | - A Guru
- Department of Plant Physiology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - B N Singh
- Department of Plant Physiology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - S K Upadhyay
- Department of Environmental Science, V.B.S. Purvanchal University, Jaunpur, India
| | - P Dwivedi
- Department of Plant Physiology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
12
|
Zhao Z, Li M, Zeng J, Huo L, Liu K, Wei R, Ni K, Gao J. Recent advances in engineering iron oxide nanoparticles for effective magnetic resonance imaging. Bioact Mater 2022; 12:214-245. [PMID: 35310380 PMCID: PMC8897217 DOI: 10.1016/j.bioactmat.2021.10.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/27/2021] [Accepted: 10/10/2021] [Indexed: 02/09/2023] Open
Abstract
Iron oxide nanoparticle (IONP) with unique magnetic property and high biocompatibility have been widely used as magnetic resonance imaging (MRI) contrast agent (CA) for long time. However, a review which comprehensively summarizes the recent development of IONP as traditional T2 CA and its new application for different modality of MRI, such as T1 imaging, simultaneous T2/T1 or MRI/other imaging modality, and as environment responsive CA is rare. This review starts with an investigation of direction on the development of high-performance MRI CA in both T2 and T1 modal based on quantum mechanical outer sphere and Solomon–Bloembergen–Morgan (SBM) theory. Recent rational attempts to increase the MRI contrast of IONP by adjusting the key parameters, including magnetization, size, effective radius, inhomogeneity of surrounding generated magnetic field, crystal phase, coordination number of water, electronic relaxation time, and surface modification are summarized. Besides the strategies to improve r2 or r1 values, strategies to increase the in vivo contrast efficiency of IONP have been reviewed from three different aspects, those are introducing second imaging modality to increase the imaging accuracy, endowing IONP with environment response capacity to elevate the signal difference between lesion and normal tissue, and optimizing the interface structure to improve the accumulation amount of IONP in lesion. This detailed review provides a deep understanding of recent researches on the development of high-performance IONP based MRI CAs. It is hoped to trigger deep thinking for design of next generation MRI CAs for early and accurate diagnosis. T2 contrast capacity of iron oxide nanoparticles (IONPs) could be improved based on quantum mechanical outer sphere theory. IONPs could be expand to be used as effective T1 CAs by improving q value, extending τs, and optimizing interface structure. Environment responsive MRI CAs have been developed to improve the diagnosis accuracy. Introducing other imaging contrast moiety into IONPs could increase the contrast efficiency. Optimizing in vivo behavior of IONPs have been proved to enlarge the signal difference between normal tissue and lesion.
Collapse
|
13
|
Wu K, Liu J, Chugh VK, Liang S, Saha R, Krishna VD, Cheeran MCJ, Wang JP. Magnetic nanoparticles and magnetic particle spectroscopy-based bioassays: a 15 year recap. NANO FUTURES 2022; 6:022001. [PMID: 36199556 PMCID: PMC9531898 DOI: 10.1088/2399-1984/ac5cd1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Magnetic nanoparticles (MNPs) have unique physical and chemical properties, such as high surface area to volume ratio and size-related magnetism, which are completely different from their bulk materials. Benefiting from the facile synthesis and chemical modification strategies, MNPs have been widely studied for applications in nanomedicine. Herein, we firstly summarized the designs of MNPs from the perspectives of materials and physicochemical properties tailored for biomedical applications. Magnetic particle spectroscopy (MPS), first reported in 2006, has flourished as an independent platform for many biological and biomedical applications. It has been extensively reported as a versatile platform for a variety of bioassays along with the artificially designed MNPs, where the MNPs serve as magnetic nanoprobes to specifically probe target analytes from fluid samples. In this review, the mechanisms and theories of different MPS platforms realizing volumetric- and surface-based bioassays are discussed. Some representative works of MPS platforms for applications such as disease diagnosis, food safety and plant pathology monitoring, drug screening, thrombus maturity assessments are reviewed. At the end of this review, we commented on the rapid growth and booming of MPS-based bioassays in its first 15 years. We also prospected opportunities and challenges that portable MPS devices face in the rapidly growing demand for fast, inexpensive, and easy-to-use biometric techniques.
Collapse
Affiliation(s)
- Kai Wu
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Jinming Liu
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Vinit Kumar Chugh
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Shuang Liang
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Renata Saha
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Venkatramana D Krishna
- Department of Veterinary Population Medicine, University of Minnesota, St Paul, MN 55108, United States of America
| | - Maxim C-J Cheeran
- Department of Veterinary Population Medicine, University of Minnesota, St Paul, MN 55108, United States of America
| | - Jian-Ping Wang
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, United States of America
| |
Collapse
|
14
|
Zhang Z, Wu Y, Lin N, Yin S, Meng Z. Monitoring Clinical-Pathological Grading of Hepatocellular Carcinoma Using MicroRNA-Guided Semiconducting Polymer Dots. ACS APPLIED MATERIALS & INTERFACES 2022; 14:7717-7730. [PMID: 35112844 DOI: 10.1021/acsami.1c24191] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
MicroRNAs (miRNAs) are a class of small, noncoding RNAs involved in nearly all genetic central dogma processes and human biological behavior, which also play a significant role in the pathological activity of tumors, such as gene transcription, protein translation, and exosome secretion. Therefore, through the navigation of certain specific miRNAs, we can trace the specific physiological processes or image some specific tissues. Designing and accurately positioning microRNA (miRNA)-sensitive fluorescent nanoprobes with benign specificity and recognition in cells or tissues are a challenging research field. To solve the difficulties, we introduce four semiconducting polymer dots (Pdots) as nanoprobes linked by specific miRNA antisense sequences for monitoring the pathological grading by the variation in miRNA expression. Based on the base pairing principle, these miRNA-sensitive Pdots could bind to specific miRNAs within the cancerous cells. As impacted by the background of different pathology gradings, the proportions of the four hepatocellular carcinoma (HCC)-specific miRNAs within the cancerous cell are different, and the pathological grading of the patient tissues can be determined by comparing the palette combinations. The short single-stranded RNA-functionalized Pdots, which have excellent microRNA sensitivity, are observed in an experimental cell model and a series of tissue specimens from HCC patients for the first time. Using the Förster (or fluorescence) resonance energy transfer (FRET) model of Pdots and Cy3dt tag to simulate in vivo miRNA detection, the superior sensitivity and specificity of these nanoprobes are verified. The interference of subjective factors in traditional single/bis-dye emission intensity detection is abandoned, and multiple label staining is used to enhance sensitivity further and reduce the false-positive rate. The feasibility exhibited by this novel staining method is verified in normal hepatocellular HCC cell lines and 16 frozen ultrathin tissue sections, which are employed to quantify pathological grading-related color presentation systems for clinical doctors and pathologists' use. The intelligently designed miRNA-guided Pdots will emerge as an ideal platform with promising biological imaging.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, No. 126 Xiantai Street, Changchun, Jilin 130000, P. R. China
| | - Yuyang Wu
- State Key Laboratory of Integrated Optoelectronic, College of Electronic Science and Engineering, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Nan Lin
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Shengyan Yin
- State Key Laboratory of Integrated Optoelectronic, College of Electronic Science and Engineering, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Zihui Meng
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, No. 126 Xiantai Street, Changchun, Jilin 130000, P. R. China
| |
Collapse
|
15
|
Wang X, Yuan W, Xu M, Su X, Li F. Visualization of Acute Inflammation through a Macrophage-Camouflaged Afterglow Nanocomplex. ACS APPLIED MATERIALS & INTERFACES 2022; 14:259-267. [PMID: 34957836 DOI: 10.1021/acsami.1c19388] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Acute inflammation is a basic innate, immediate, and stereotyped immune response to injury, which is characterized by rapid recruitment of immune cells to the vasculature and extravasation into the damaged parenchyma. Visualization of acute inflammation plays an important role in monitoring the disease course and understanding pathogenesis, which lacks specific targeted and observing tools in vivo. Here, we report a Trojan horse strategy of a macrophage-camouflaged afterglow nanocomplex (UCANPs@RAW) to specifically visualize acute inflammation. Due to the advantages of optical antibackground interference elimination, as well as particular immune homing and long-term tracking capacity, UCANPs@RAW demonstrates an excellent acute inflammatory recognition ability. In an arthritis model, previously intravenously injected UCANPs@RAW could directionally migrate from the liver to the inflammation site as soon as 3 h after the model was induced, which could be continuously lighted for at least 36 h with the highest imaging signal-to-background ratio (SBR) as 382 at the time point of 9 h. Additionally, UCANPs@RAW is observed to penetrate the blood-brain barrier and image the deep brain inflamed region covered by the thick skull in an acute brain inflammation model with an SBRmax of 258, which is based on the strong recruiting ability of macrophages to immune response. In view of this smart nanocomplex, our strategy holds great potential for inflammatory detection and treatments.
Collapse
Affiliation(s)
- Xiu Wang
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Wei Yuan
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Ming Xu
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Xianlong Su
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Fuyou Li
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| |
Collapse
|
16
|
Yu C, Chen Z, Li X, Bao H, Wang Y, Zhang B, Huang J, Zhang Z. pH-Triggered Aggregation of Gold Nanoparticles for Enhanced Labeling and Long-Term CT Imaging Tracking of Stem Cells in Pulmonary Fibrosis Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101861. [PMID: 34235846 DOI: 10.1002/smll.202101861] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/11/2021] [Indexed: 06/13/2023]
Abstract
Gold nanoparticles (AuNPs) pose a great challenge in the development of nanotracers that can self-adaptively alter their properties in response to certain cellular environments for long-term stem cell tracking. Herein, pH-sensitive Au nanotracers (CPP-PSD@Au) are fabricated by sequential coupling of AuNPs with sulfonamide-based polymer (PSD) and cell-penetrating peptide (CPP), which can be efficiently internalized by mesenchymal stem cells (MSCs) and undergo pH-induced self-assembly in endosomes, facilitating long-term computed tomography (CT) imaging tracking MSCs in a murine model of idiopathic pulmonary fibrosis (IPF). Using the CPP-PSD@Au, the transplanted MSCs for the first time can be monitored with CT imaging for up to 35 days after transplantation into the lung of IPF mice, clearly elucidating the migration process of MSCs in vivo. Moreover, we preliminarily explored the mechanism of the CPP-PSD@Au labeled MSCs in the alleviation of IPF, including recovery of alveolar integrity, decrease of collagen deposition, as well as down-regulation of relevant cytokine level. This work facilitates our understanding of the behavior and effect of MSCs in the therapy of IPF, thereby providing an important insight into the stem cell-based treatment of lung diseases.
Collapse
Affiliation(s)
- Chenggong Yu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Zhongjin Chen
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xiaodi Li
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Hongying Bao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yujie Wang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Bo Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jie Huang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Zhijun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
17
|
Carnovale C, Guarnieri D, Di Cristo L, De Angelis I, Veronesi G, Scarpellini A, Malvindi MA, Barone F, Pompa PP, Sabella S. Biotransformation of Silver Nanoparticles into Oro-Gastrointestinal Tract by Integrated In Vitro Testing Assay: Generation of Exposure-Dependent Physical Descriptors for Nanomaterial Grouping. NANOMATERIALS 2021; 11:nano11061587. [PMID: 34204296 PMCID: PMC8233905 DOI: 10.3390/nano11061587] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/28/2021] [Accepted: 06/09/2021] [Indexed: 12/28/2022]
Abstract
Grouping approaches of nanomaterials have the potential to facilitate high throughput and cost effective nanomaterial screening. However, an effective grouping of nanomaterials hinges on the application of suitable physicochemical descriptors to identify similarities. To address the problem, we developed an integrated testing approach coupling acellular and cellular phases, to study the full life cycle of ingested silver nanoparticles (NPs) and silver salts in the oro-gastrointestinal (OGI) tract including their impact on cellular uptake and integrity. This approach enables the derivation of exposure-dependent physical descriptors (EDPDs) upon biotransformation of undigested nanoparticles, digested nanoparticles and digested silver salts. These descriptors are identified in: size, crystallinity, chemistry of the core material, dissolution, high and low molecular weight Ag-biomolecule soluble complexes, and are compared in terms of similarities in a grouping hypothesis. Experimental results indicate that digested silver nanoparticles are neither similar to pristine nanoparticles nor completely similar to digested silver salts, due to the presence of different chemical nanoforms (silver and silver chloride nanocrystals), which were characterized in terms of their interactions with the digestive matrices. Interestingly, the cellular responses observed in the cellular phase of the integrated assay (uptake and inflammation) are also similar for the digested samples, clearly indicating a possible role of the soluble fraction of silver complexes. This study highlights the importance of quantifying exposure-related physical descriptors to advance grouping of NPs based on structural similarities.
Collapse
Affiliation(s)
- Catherine Carnovale
- Istituto Italiano Di Tecnologia, Nanoregulatory Platform, Drug Discovery and Development Department, 16163 Genova, Italy; (C.C.); (L.D.C.)
| | - Daniela Guarnieri
- Dipartimento di Chimica e Biologia “A. Zambelli”, Università di Salerno, Via Giovanni Paolo II 132, Fisciano, 84084 Salerno, Italy;
- Research Centre for Biomaterials BIONAM, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084 Salerno, Italy
| | - Luisana Di Cristo
- Istituto Italiano Di Tecnologia, Nanoregulatory Platform, Drug Discovery and Development Department, 16163 Genova, Italy; (C.C.); (L.D.C.)
| | | | - Giulia Veronesi
- Laboratory of Chemistry and Biology of Metals (CBM), University Grenoble Alpes/CNRS/CEA, 38000 Grenoble, France;
- ESRF, the European Synchrotron, 71 Av. des Martyrs, 38000 Grenoble, France
| | - Alice Scarpellini
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy;
| | | | - Flavia Barone
- Istituto Superiore di Sanità (ISS), 00161 Rome, Italy; (I.D.A.); (F.B.)
| | - Pier Paolo Pompa
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy;
| | - Stefania Sabella
- Istituto Italiano Di Tecnologia, Nanoregulatory Platform, Drug Discovery and Development Department, 16163 Genova, Italy; (C.C.); (L.D.C.)
- Correspondence:
| |
Collapse
|
18
|
Ho LWC, Yin B, Dai G, Choi CHJ. Effect of Surface Modification with Hydrocarbyl Groups on the Exocytosis of Nanoparticles. Biochemistry 2021; 60:1019-1030. [PMID: 33169977 DOI: 10.1021/acs.biochem.0c00631] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Designing nanoparticles (NPs) with desirable cell type-specific exocytosis properties, say promoting their exocytosis from scavenging cell types (e.g., macrophages and endothelial cells) or suppressing their exocytosis from target disease cell types (e.g., cancer cells), improves the application of nanomedicines. However, the design parameters available for tuning the exocytosis of NPs remain scarce in the "nano-cell" literature. Here, we demonstrate that surface modification of NPs with hydrocarbyl functional groups, commonly found in biomolecules and NP-based drug carriers, is a critical parameter for tuning the exocytosis of NPs from RAW264.7 macrophages, C166 endothelial cells, and HeLa epithelial cancer cells. To exclude the effect of hydrophobicity, we prepare a collection of hydrophilic NPs that bear a gold NP (AuNP) core, a dense polyethylene glycol (PEG) shell, and different types of hydrocarbyl groups (X) that are attached to the distal end of the PEG strands (termed "Au@PEG-X NPs"). For all three cell types tested, modification of NPs with straight-chain dodecane leads to a >10-fold increase in the level of cellular uptake, drastically higher than those of all other types of X tested. However, the probability of exocytosis of NPs significantly depends on the types of cell and X. Notably, NPs modified with cyclododecanes are most likely to be exocytosed by RAW264.7 and C166 cells (but not HeLa cells), accompanied by the release of intralumenal vesicles to the extracellular milieu. These data suggest a reductionist approach for rationally assembling bionanomaterials for nanomedicine applications by using hydrocarbyl functional groups as building blocks.
Collapse
|
19
|
Pisonero J, Traub H, Cappella B, Álvarez-Llamas C, Méndez A, Richter S, Encinar JR, Costa-Fernandez JM, Bordel N. Exploring quantitative cellular bioimaging and assessment of CdSe/ZnS quantum dots cellular uptake in single cells, using ns-LA-ICP-SFMS. Talanta 2021; 227:122162. [PMID: 33714466 DOI: 10.1016/j.talanta.2021.122162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/12/2021] [Accepted: 01/25/2021] [Indexed: 01/25/2023]
Abstract
Quantitative bioimaging of Quantum Dots (QDs) uptake in single cells by laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) is a challenging task due to the high sensitivity and high spatial resolution required, and to the lack of matrix-matched reference materials. In this work, high spatially resolved quantitative bioimaging of CdSe/ZnS QDs uptake in single HT22 mouse hippocampal neuronal cells and in single HeLa human cervical carcinoma cells is novelty investigated combining: (a) the use of a ns-LA-ICP-Sector Field (SF)MS unit with mono-elemental fast and sensitive single pulse response for 114Cd+; and (b) the spatially resolved analysis of dried pL-droplets from a solution with a known concentration of these QDs to obtain a response factor that allows quantification of elemental bioimages. Single cells and dried pL-droplets are morphologically characterized by Atomic Force Microscopy (AFM) to determine their volume and thickness distribution. Moreover, operating conditions (e.g. spot size, energy per laser pulse, etc.) are optimized to completely ablate the cells and pL droplets at high spatial resolution. Constant operating conditions for the analysis of the single cells and calibrating samples is employed to reduce potential fractionation effects related to mass load effects in the ICP. A number concentration of CdSe/ZnS QDs between 3.5 104 and 48 104 is estimated to be uptaken by several selected single HT22 and HeLa cells, after being incubated in the presence of a QDs suspension added to a standard cell culture medium. Mono-elemental bioimaging at subcellular resolution seems to show a higher number concentration of the CdSe/ZnS QDs in the cytosol around the cell nucleus.
Collapse
Affiliation(s)
- J Pisonero
- Department of Physics, University of Oviedo, C/ Federico García Lorca, Nº18, 33007, Oviedo, Spain.
| | - H Traub
- Bundesanstalt für Materialforschung und -prüfung, (BAM), Unter Den Eichen 87, 12205, Berlin, Germany
| | - B Cappella
- Bundesanstalt für Materialforschung und -prüfung, (BAM), Unter Den Eichen 87, 12205, Berlin, Germany
| | - C Álvarez-Llamas
- Department of Analytical Chemistry, University of Malaga, 29071, Málaga, Spain
| | - A Méndez
- Department of Physics, University of Oviedo, C/ Federico García Lorca, Nº18, 33007, Oviedo, Spain
| | - S Richter
- Bundesanstalt für Materialforschung und -prüfung, (BAM), Unter Den Eichen 87, 12205, Berlin, Germany
| | - J Ruiz Encinar
- Department of Physical and Analytical Chemistry, University of Oviedo, Avda. Julian Claveria, 8, 33006, Oviedo, Spain
| | - J M Costa-Fernandez
- Department of Physical and Analytical Chemistry, University of Oviedo, Avda. Julian Claveria, 8, 33006, Oviedo, Spain
| | - N Bordel
- Department of Physics, University of Oviedo, C/ Federico García Lorca, Nº18, 33007, Oviedo, Spain
| |
Collapse
|
20
|
Zhao H, Lv J, Li F, Zhang Z, Zhang C, Gu Z, Yang D. Enzymatical biomineralization of DNA nanoflowers mediated by manganese ions for tumor site activated magnetic resonance imaging. Biomaterials 2020; 268:120591. [PMID: 33338933 DOI: 10.1016/j.biomaterials.2020.120591] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/12/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
DNA nanoflower has been demonstrated as a promising DNA nanostructure for therapeutics and bioimaging primarily because of the programmable DNA sequence and unique structure. Herein, we report manganese ions mediated enzymatic biomineralization to prepare DNA-Mn hybrid nanoflower (DMNF). Paramagnetic Mn2+ was explored as the co-factor of DNA polymerase for the extension of long strand DNA. The biomimetic synthesis of DMNF was performed using the long strand DNA as template via nucleation and growth of Mn2PPi. The morphology and size of DMNF were controllable by tuning reaction time and Mn2+ concentration. The aptamer sequence was encoded into circle template to achieve tumor-targeted DMNF, and cellular uptake assay demonstrated obvious aptamer-mediated internalization. DMNF showed enhanced T1-weighted magnetic resonance (MR) imaging effect in acid environment for high tumor-specific MR imaging, and high spatial resolution imaging of kidneys and liver. Our work provides a facile enzymatically biomineral strategy to integrate multifunctional modules into one DNA structure and promotes the development of DNA nanostructure for precision medicine.
Collapse
Affiliation(s)
- Huaixin Zhao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Jigang Lv
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Feng Li
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Zhili Zhang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Chunzhi Zhang
- Department of Radiation Oncology, Tianjin Hospital, Tianjin, 300211, PR China
| | - Zi Gu
- School of Chemical Engineering and Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Dayong Yang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China.
| |
Collapse
|
21
|
Mehta N, Sahu SP, Shaik S, Devireddy R, Gartia MR. Dark-field hyperspectral imaging for label free detection of nano-bio-materials. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1661. [PMID: 32755036 DOI: 10.1002/wnan.1661] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 05/21/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022]
Abstract
Nanomaterials are playing an increasingly important role in cancer diagnosis and treatment. Nanoparticle (NP)-based technologies have been utilized for targeted drug delivery during chemotherapies, photodynamic therapy, and immunotherapy. Another active area of research is the toxicity studies of these nanomaterials to understand the cellular uptake and transport of these materials in cells, tissues, and environment. Traditional techniques such as transmission electron microscopy, and mass spectrometry to analyze NP-based cellular transport or toxicity effect are expensive, require extensive sample preparation, and are low-throughput. Dark-field hyperspectral imaging (DF-HSI), an integration of spectroscopy and microscopy/imaging, provides the ability to investigate cellular transport of these NPs and to quantify the distribution of them within bio-materials. DF-HSI also offers versatility in non-invasively monitoring microorganisms, single cell, and proteins. DF-HSI is a low-cost, label-free technique that is minimally invasive and is a viable choice for obtaining high-throughput quantitative molecular analyses. Multimodal imaging modalities such as Fourier transform infrared and Raman spectroscopy are also being integrated with HSI systems to enable chemical imaging of the samples. HSI technology is being applied in surgeries to obtain molecular information about the tissues in real-time. This article provides brief overview of fundamental principles of DF-HSI and its application for nanomaterials, protein-detection, single-cell analysis, microbiology, surgical procedures along with technical challenges and future integrative approach with other imaging and measurement modalities. This article is categorized under: Diagnostic Tools > in vitro Nanoparticle-Based Sensing Diagnostic Tools > in vivo Nanodiagnostics and Imaging Implantable Materials and Surgical Technologies > Nanoscale Tools and Techniques in Surgery.
Collapse
Affiliation(s)
- Nishir Mehta
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Sushant P Sahu
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Shahensha Shaik
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Ram Devireddy
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
22
|
Abstract
Contaminants, organic or inorganic, represent a threat for the environment and human health and in recent years their presence and persistence has increased rapidly. For this reason, several technologies including bioremediation in combination with nanotechnology have been explored to identify more systemic approaches for their removal from environmental matrices. Understanding the interaction between the contaminant, the microorganism, and the nanomaterials (NMs) is of crucial importance since positive and negative effects may be produced. For example, some nanomaterials are stimulants for microorganisms, while others are toxic. Thus, proper selection is of paramount importance. The main objective of this review was to analyze the principles of bioremediation assisted by nanomaterials, nanoparticles (NPs) included, and their interaction with environmental matrices. It also analyzed the response of living organisms employed to remediate the contaminants in the presence of nanomaterials. Besides, we discuss the international regulatory frame applicable to these technologies and how they might contribute to sustainability.
Collapse
|
23
|
Kapara A, Brunton V, Graham D, Faulds K. Investigation of cellular uptake mechanism of functionalised gold nanoparticles into breast cancer using SERS. Chem Sci 2020; 11:5819-5829. [PMID: 34094083 PMCID: PMC8159335 DOI: 10.1039/d0sc01255f] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/20/2020] [Indexed: 01/04/2023] Open
Abstract
Gold nanoparticles (AuNPs) are widely used in various applications such as cancer imaging and drug delivery. The functionalisation of AuNPs has been shown to affect their cellular internalisation, accumulation and targeting efficiency. The mechanism of cellular uptake of functionalised AuNPs by different cancer cells is not well understood. Therefore, a detailed understanding of the molecular processes is necessary to improve AuNPs for their selective uptake and fate in specific cellular systems. This knowledge can greatly help in designing nanotags with higher cellular uptake for more selective and specific targeting capabilities with less off-target effects. Here, we demonstrate for the first time a straightforward and non-destructive 3D surface enhanced Raman spectroscopy (SERS) imaging approach to track the cellular uptake and localisation of AuNPs functionalised with an anti-ERα (estrogen receptor alpha) antibody in MCF-7 ERα-positive human breast cancer cells under different conditions including temperature and dynamin inhibition. 3D SERS enabled information rich monitoring of the intracellular internalisation of the SERS nanotags. It was found that ERα-AuNPs were internalised by MCF-7 cells in a temperature-dependent manner suggesting an active endocytosis-dependent mechanism. 3D SERS cell mapping also indicated that the nanotags entered MCF-7 cells using dynamin dependent endocytosis, since dynamin inhibition resulted in the SERS signal being obtained from, or close to, the cell surface rather than inside the cells. Finally, ERα-AuNPs were found to enter MCF-7 cells using an ERα receptor-mediated endocytosis process. This study addresses the role of functionalisation of SERS nanotags in biological environments and highlights the benefits of using 3D SERS for the investigation of cellular uptake processes.
Collapse
Affiliation(s)
- Anastasia Kapara
- Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde 99 George Street Glasgow Scotland G1 1RD UK
- Edinburgh Cancer Research UK Centre, University of Edinburgh Crewe Road South Edinburgh Scotland EH4 2XU UK
| | - Valerie Brunton
- Edinburgh Cancer Research UK Centre, University of Edinburgh Crewe Road South Edinburgh Scotland EH4 2XU UK
| | - Duncan Graham
- Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde 99 George Street Glasgow Scotland G1 1RD UK
| | - Karen Faulds
- Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde 99 George Street Glasgow Scotland G1 1RD UK
| |
Collapse
|
24
|
Ghiassian S, Yu L, Gobbo P, Nazemi A, Romagnoli T, Luo W, Luyt LG, Workentin MS. Nitrone-Modified Gold Nanoparticles: Synthesis, Characterization, and Their Potential as 18F-Labeled Positron Emission Tomography Probes via I-SPANC. ACS OMEGA 2019; 4:19106-19115. [PMID: 31763533 PMCID: PMC6868604 DOI: 10.1021/acsomega.9b02322] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/28/2019] [Indexed: 05/11/2023]
Abstract
A novel bioorthogonal gold nanoparticle (AuNP) template displaying interfacial nitrone functional groups for bioorthogonal interfacial strain-promoted alkyne-nitrone cycloaddition reactions has been synthesized. These nitrone-AuNPs were characterized in detail using 1H nuclear magnetic resonance spectroscopy, transmission electron microscopy, thermogravimetric analysis, and X-ray photoelectron spectroscopy, and a nanoparticle raw formula was calculated. The ability to control the conjugation of molecules of interest at the molecular level onto the nitrone-AuNP template allowed us to create a novel methodology for the synthesis of AuNP-based radiolabeled probes.
Collapse
Affiliation(s)
- Sara Ghiassian
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Lihai Yu
- London
Regional Cancer Program, 800 Commissioners Rd. E., London N6A 5W9, Ontario, Canada
| | - Pierangelo Gobbo
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Ali Nazemi
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Tommaso Romagnoli
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Wilson Luo
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Leonard G. Luyt
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
- London
Regional Cancer Program, 800 Commissioners Rd. E., London N6A 5W9, Ontario, Canada
| | - Mark S. Workentin
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| |
Collapse
|
25
|
Yuan H, Li X, Tang J, Zhou M, Liu F. Local application of doxorubicin- loaded Iron oxid nanoparticles and the vascular disrupting agent via the hepatic artery: chemoembolization-photothermal ablation treatment of hepatocellular carcinoma in rats. Cancer Imaging 2019; 19:71. [PMID: 31685015 PMCID: PMC6829940 DOI: 10.1186/s40644-019-0257-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/25/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES This study investigates the effectiveness of local application of doxorubicin(Dox)-loaded, polydopamine (PDA)- coated single crystal hematite (α- Fe2O3) nanocubes (Fe2O3-PDA-Dox) and combretastatin A-4 phosphate disodium(CA4P)in treating hepatocellular carcinoma (HCC) in rats. METHODS The magnetic characteristics and photothermal effects of the nanoparticles were determined in vitro. Tumor-bearing Sprague-Dawley rats were divided into 3 groups of 8 according to treatment: controls, transarterial chemoembolization-photothermal ablation (pTACE) (Lipidol+Fe2O3-PDA-Dox + NIR), and CA4P + pTACE (CA4P+ Lipidol+Fe2O3-PDA-Dox + NIR). Drugs were administered through the hepatic artery, and the tumors exposed to 808-nm near-infrared radiation. The Fe content of tumors was assessed using neutron activation analysis. Treatment effectiveness was assessed using heating curves, magnetic resonance imaging, pathology results, and immunohistochemical analysis. RESULTS The mean tumor Fe content was greater in rats treated with CA4P + pTACE (1 h, 23.72 ± 12.45 μg/g; 24 h, 14.61 ± 8.23 μg/g) than in those treated with pTACE alone (1 h, 5.66 ± 4.29 μg/g; 24 h, 2.76 ± 1.33 μg/g). The tumor T2 imaging signal was lower in rats treated with CA4P + pTACE. Following laser irradiation, the tumor temperature increased, with higher temperatures reached in the CA4P + pTACE group (62 °C vs 55 °C). Tumor cells exhibited necrosis, apoptosis, and proliferation inhibition, with greater effects in the CA4P + pTACE group. Transient liver and kidney toxicity were observed on day 3, with more severe effects after CA4P + pTACE. CONCLUSIONS Fe2O3-PDA-Dox nanoparticles are effective for TACE-PTA. Pretreatment with CA4P increases nanoparticle uptake by tumors, increasing the treatment effectiveness without increasing hepatorenal toxicity.
Collapse
Affiliation(s)
- Hongjun Yuan
- Department of Interventional Radiology, The First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Xin Li
- Department of Interventional Radiology, The First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Jing Tang
- Department of Interventional Radiology, The First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Min Zhou
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Fengyong Liu
- Department of Interventional Radiology, The First Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
26
|
Abstract
Nanoparticulate materials displaying enzyme-like properties, so-called nanozymes, are explored as substitutes for natural enzymes in several industrial, energy-related, and biomedical applications. Outstanding high stability, enhanced catalytic activities, low cost, and availability at industrial scale are some of the fascinating features of nanozymes. Furthermore, nanozymes can also be equipped with the unique attributes of nanomaterials such as magnetic or optical properties. Due to the impressive development of nanozymes during the last decade, their potential in the context of tissue engineering and regenerative medicine also started to be explored. To highlight the progress, in this review, we discuss the two most representative nanozymes, namely, cerium- and iron-oxide nanomaterials, since they are the most widely studied. Special focus is placed on their applications ranging from cardioprotection to therapeutic angiogenesis, bone tissue engineering, and wound healing. Finally, current challenges and future directions are discussed.
Collapse
|
27
|
Chen LJ, Zhao X, Yan XP. Cell-Penetrating Peptide-Functionalized Persistent Luminescence Nanoparticles for Tracking J774A.1 Macrophages Homing to Inflamed Tissues. ACS APPLIED MATERIALS & INTERFACES 2019; 11:19894-19901. [PMID: 31081614 DOI: 10.1021/acsami.9b05870] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Recruitment of leukocytes exhibiting arguable and ambiguous processes is involved in the inflammatory response, a specific reaction of organisms to tissue damage. Tracking leukocytes is of great importance for understanding the recruitment of leukocytes. Here, we report the fabrication of carboxyl silane and TAT cell-penetrating peptide-functionalized near-infrared-emitting persistent luminescence nanoparticles (PLNP-TAT) with deep tissue penetration in bioimaging for autofluorescence-free tracking of J774A.1 macrophages homing to inflamed tissues. The PLNP-TAT enables effective labeling of the J774A.1 macrophages and the tracking of the migration of cells to the simulated endothelial inflammatory microenvironment in vitro. Moreover, the PLNP-TAT also allows the tracking of J774A.1 macrophages homing to inflamed tissues in vivo under discontinuous illumination with a red light-emitting diode light. The PLNP-TAT allows in vivo tracking of leukocytes without the need for conventional continuous excitation and offers great potential in cell-tracking and diagnostic applications without the autofluorescence background and thermal damages brought about by continuous excitation.
Collapse
|
28
|
Liu X, Yang Z, Sun J, Ma T, Hua F, Shen Z. A brief review of cytotoxicity of nanoparticles on mesenchymal stem cells in regenerative medicine. Int J Nanomedicine 2019; 14:3875-3892. [PMID: 31213807 PMCID: PMC6539172 DOI: 10.2147/ijn.s205574] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/21/2019] [Indexed: 12/30/2022] Open
Abstract
Multipotent mesenchymal stem cells have shown great promise for application in regenerative medicine owing to their particular therapeutic effects, such as significant self-renewability, low immunogenicity, and ability to differentiate into a variety of specialized cells. However, there remain certain complicated and unavoidable problems that limit their further development and application. One of the challenges is to noninvasively monitor the delivery and biodistribution of transplanted stem cells during treatment without relying on behavioral endpoints or tissue histology, and it is important to explore the potential mechanisms to clarify how stem cells work in vivo. To solve these problems, various nanoparticles (NPs) and their corresponding imaging methods have been developed recently and have made great progress. In this review, we mainly discuss NPs used to label stem cells and their toxic effects on the latter, the imaging techniques to detect such NPs, and the current existing challenges in this field.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, People's Republic of China
| | - Ziying Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, People's Republic of China
| | - Jiacheng Sun
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, People's Republic of China
| | - Teng Ma
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, People's Republic of China
| | - Fei Hua
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, People's Republic of China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
29
|
Fu W, Yan C, Zhang Y, Ma Y, Guo Z, Zhu WH. Near-Infrared Aggregation-Induced Emission-Active Probe Enables in situ and Long-Term Tracking of Endogenous β-Galactosidase Activity. Front Chem 2019; 7:291. [PMID: 31139612 PMCID: PMC6527754 DOI: 10.3389/fchem.2019.00291] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/09/2019] [Indexed: 01/08/2023] Open
Abstract
High-fidelity tracking of specific enzyme activities is critical for the early diagnosis of diseases such as cancers. However, most of the available fluorescent probes are difficult to obtain in situ information because of tending to facile diffusion or inevitably suffering from aggregation-caused quenching (ACQ) effect. In this work, we developed an elaborated near-infrared (NIR) aggregation-induced emission (AIE)-active fluorescent probe, which is composed of a hydrophobic 2-(2-hydroxyphenyl) benzothiazole (HBT) moiety for extending into the NIR wavelength, and a hydrophilic β-galactosidase (β-gal) triggered unit for improving miscibility and guaranteeing its non-emission in aqueous media. This probe is virtually activated by β-gal, and then specific enzymatic turnover would liberate hydrophobic AIE luminogen (AIEgen) QM-HBT-OH. Simultaneously, brightness NIR fluorescent nanoaggregates are in situ generated as a result of the AIE-active process, making on-site the detection of endogenous β-gal activity in living cells. By virtue of the NIR AIE-active performance of enzyme-catalyzed nanoaggregates, QM-HBT-βgal is capable of affording a localizable fluorescence signal and long-term tracking of endogenous β-gal activity. All results demonstrate that the probe QM-HBT-βgal has potential to be a powerful molecular tool to evaluate the biological activity of β-gal, attaining high-fidelity information in preclinical applications.
Collapse
Affiliation(s)
| | | | | | | | - Zhiqian Guo
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, Institute of Fine Chemicals, East China University of Science and Technology, Shanghai, China
| | | |
Collapse
|
30
|
Macrophage cell tracking PET imaging using mesoporous silica nanoparticles via in vivo bioorthogonal F-18 labeling. Biomaterials 2019; 199:32-39. [PMID: 30735894 DOI: 10.1016/j.biomaterials.2019.01.043] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/01/2019] [Accepted: 01/30/2019] [Indexed: 11/20/2022]
Abstract
We introduce an efficient cell tracking imaging protocol using positron emission tomography (PET). Since macrophages are known to home and accumulate in tumor tissues and atherosclerotic plaque, we design a PET imaging protocol for macrophage cell tracking using aza-dibenzocyclooctyne-tethered PEGylated mesoporous silica nanoparticles (DBCO-MSNs) with the short half-life F-18-labeled azide-radiotracer via an in vivo strain-promoted alkyne azide cycloaddition (SPAAC) covalent labeling reaction inside macrophage cells in vivo. This PET imaging protocol for in vivo cell tracking successfully visualizes the migration of macrophage cells into the tumor site by the bioorthogonal SPAAC reaction of DBCO-MSNs with [18F]fluoropentaethylene glycolic azide ([18F]2) to form 18F-labeled aza-dibenzocycloocta-triazolic MSNs (18F-DBCOT-MSNs) inside RAW 264.7 cells. The tissue radioactivity distribution results were consistent with PET imaging findings. In addition, PET images of atherosclerosis in ApoE-/- mice fed a western diet for 30 weeks were obtained using the devised macrophage cell-tracking protocol.
Collapse
|
31
|
Ashraf S, Taylor A, Sharkey J, Barrow M, Murray P, Wilm B, Poptani H, Rosseinsky MJ, Adams DJ, Lévy R. In vivo fate of free and encapsulated iron oxide nanoparticles after injection of labelled stem cells. NANOSCALE ADVANCES 2019; 1:367-377. [PMID: 36132463 PMCID: PMC9473218 DOI: 10.1039/c8na00098k] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/16/2018] [Indexed: 05/21/2023]
Abstract
Nanoparticle contrast agents are useful tools to label stem cells and monitor the in vivo bio-distribution of labeled cells in pre-clinical models of disease. In this context, understanding the in vivo fate of the particles after injection of labelled cells is important for their eventual clinical use as well as for the interpretation of imaging results. We examined how the formulation of superparamagnetic iron oxide nanoparticles (SPIONs) impacts the labelling efficiency, magnetic characteristics and fate of the particles by comparing individual SPIONs with polyelectrolyte multilayer capsules containing SPIONs. At low labelling concentration, encapsulated SPIONs served as an efficient labelling agent for stem cells. The bio-distribution after intra-cardiac injection of labelled cells was monitored longitudinally by MRI and as an endpoint by inductively coupled plasma-optical emission spectrometry. The results suggest that, after being released from labelled cells after cell death, both formulations of particles are initially stored in liver and spleen and are not completely cleared from these organs 2 weeks post-injection.
Collapse
Affiliation(s)
- Sumaira Ashraf
- Department of Biochemistry, Institute of Integrative Biology (IIB), University of Liverpool Liverpool UK
| | - Arthur Taylor
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
- Centre for Preclinical Imaging, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
| | - Jack Sharkey
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
- Centre for Preclinical Imaging, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
| | - Michael Barrow
- Department of Chemistry, University of Liverpool Liverpool UK
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
- Centre for Preclinical Imaging, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
| | - Bettina Wilm
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
- Centre for Preclinical Imaging, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
| | - Harish Poptani
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
- Centre for Preclinical Imaging, Institute of Translational Medicine (ITM), University of Liverpool Liverpool UK
| | | | - Dave J Adams
- Department of Chemistry, University of Liverpool Liverpool UK
- School of Chemistry, University of Glasgow Glasgow UK
| | - Raphaël Lévy
- Department of Biochemistry, Institute of Integrative Biology (IIB), University of Liverpool Liverpool UK
| |
Collapse
|
32
|
Gu K, Qiu W, Guo Z, Yan C, Zhu S, Yao D, Shi P, Tian H, Zhu WH. An enzyme-activatable probe liberating AIEgens: on-site sensing and long-term tracking of β-galactosidase in ovarian cancer cells. Chem Sci 2019; 10:398-405. [PMID: 30746088 PMCID: PMC6334664 DOI: 10.1039/c8sc04266g] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022] Open
Abstract
Development of fluorescent probes for on-site sensing and long-term tracking of specific biomarkers is particularly desirable for the early detection of diseases. However, available small-molecule probes tend to facilely diffuse across the cell membrane or remain at the activation site but always suffer from the aggregation-caused quenching (ACQ) effect. Here we report an enzyme-activatable aggregation-induced emission (AIE) probe QM-βgal, which is composed of a hydrophilic β-galactosidase (β-gal)-triggered galactose moiety and a hydrophobic AIE-active fluorophore QM-OH. The probe is virtually non-emissive in aqueous media, but when activated by β-gal, specific enzymatic turnover would liberate hydrophobic AIE luminogen (AIEgen) QM-OH, and then highly fluorescent nanoaggregates are in situ generated as a result of the AIE process, allowing for on-site sensing of endogenous β-gal activity in living cells. Notably, taking advantage of the improved intracellular retention of nanoaggregates, we further exemplify QM-βgal for long-term (∼12 h) visualization of β-gal-overexpressing ovarian cancer cells with high fidelity, which is essential for biomedicine and diagnostics. Thus, this enzyme-activatable AIE probe not only is a potent tool for elucidating the roles of β-gal in biological systems, but also offers an enzyme-regulated liberation strategy to exploit multifunctional probes for preclinical applications.
Collapse
Affiliation(s)
- Kaizhi Gu
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Advanced Materials and Institute of Fine Chemicals , Joint International Research Laboratory of Precision Chemistry and Molecular Engineering , Feringa Nobel Prize Scientist Joint Research Center , School of Chemistry and Molecular Engineering , East China University of Science & Technology , Shanghai 200237 , China .
| | - Wanshan Qiu
- Department of Cardiothoracic Surgery , Children's Hospital of Fudan University , Shanghai 201102 , China
| | - Zhiqian Guo
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Advanced Materials and Institute of Fine Chemicals , Joint International Research Laboratory of Precision Chemistry and Molecular Engineering , Feringa Nobel Prize Scientist Joint Research Center , School of Chemistry and Molecular Engineering , East China University of Science & Technology , Shanghai 200237 , China .
- State Key Laboratory of Bioreactor Engineering , East China University of Science & Technology , Shanghai 200237 , China
| | - Chenxu Yan
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Advanced Materials and Institute of Fine Chemicals , Joint International Research Laboratory of Precision Chemistry and Molecular Engineering , Feringa Nobel Prize Scientist Joint Research Center , School of Chemistry and Molecular Engineering , East China University of Science & Technology , Shanghai 200237 , China .
| | - Shiqin Zhu
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Advanced Materials and Institute of Fine Chemicals , Joint International Research Laboratory of Precision Chemistry and Molecular Engineering , Feringa Nobel Prize Scientist Joint Research Center , School of Chemistry and Molecular Engineering , East China University of Science & Technology , Shanghai 200237 , China .
| | - Defan Yao
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Advanced Materials and Institute of Fine Chemicals , Joint International Research Laboratory of Precision Chemistry and Molecular Engineering , Feringa Nobel Prize Scientist Joint Research Center , School of Chemistry and Molecular Engineering , East China University of Science & Technology , Shanghai 200237 , China .
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering , East China University of Science & Technology , Shanghai 200237 , China
| | - He Tian
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Advanced Materials and Institute of Fine Chemicals , Joint International Research Laboratory of Precision Chemistry and Molecular Engineering , Feringa Nobel Prize Scientist Joint Research Center , School of Chemistry and Molecular Engineering , East China University of Science & Technology , Shanghai 200237 , China .
| | - Wei-Hong Zhu
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Advanced Materials and Institute of Fine Chemicals , Joint International Research Laboratory of Precision Chemistry and Molecular Engineering , Feringa Nobel Prize Scientist Joint Research Center , School of Chemistry and Molecular Engineering , East China University of Science & Technology , Shanghai 200237 , China .
| |
Collapse
|
33
|
Adams CF, Delaney AM, Carwardine DR, Tickle J, Granger N, Chari DM. Nanoparticle-Based Imaging of Clinical Transplant Populations Encapsulated in Protective Polymer Matrices. Macromol Biosci 2018; 19:e1800389. [PMID: 30511815 DOI: 10.1002/mabi.201800389] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/18/2018] [Indexed: 01/26/2023]
Abstract
A recent clinical trial proves that autologous olfactory mucosal cell (OMC) transplantation improves locomotion in dogs with naturally occurring spinal injuries comparable to human lesions. However, not all dogs respond to the treatment, likely due to the transplantation procedures involving injections of cell suspensions that are associated with cell death, uneven cell distribution, and cell washout. Encapsulating cells in protective hydrogel matrices offers a tissue engineering solution to safely achieve 3D growth of viable transplant cells for implantation into injury sites, to improve regenerative outcomes. It is shown for the first time that canine OMCs (cOMCs) can be propagated with high viability in 3D collagen matrices. Further, a method to incorporate cOMCs pre-labeled with clinical-grade iron oxide nanoparticles into the constructs is described. Intraconstruct labeled cells are visualized using magnetic resonance imaging, offering substantial promise for in vivo tracking of cOMCs delivered in protective matrices.
Collapse
Affiliation(s)
- Christopher F Adams
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Alexander M Delaney
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | | | - Jacqueline Tickle
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Nicolas Granger
- The Royal Veterinary College, Hawkshead Lane, Hatfield, Hertfordshire, AL9 7TA, UK
| | - Divya M Chari
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| |
Collapse
|
34
|
Scarfe L, Taylor A, Sharkey J, Harwood R, Barrow M, Comenge J, Beeken L, Astley C, Santeramo I, Hutchinson C, Ressel L, Smythe J, Austin E, Levy R, Rosseinsky MJ, Adams DJ, Poptani H, Park BK, Murray P, Wilm B. Non-invasive imaging reveals conditions that impact distribution and persistence of cells after in vivo administration. Stem Cell Res Ther 2018; 9:332. [PMID: 30486897 PMCID: PMC6264053 DOI: 10.1186/s13287-018-1076-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/23/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022] Open
Abstract
Background Cell-based regenerative medicine therapies are now frequently tested in clinical trials. In many conditions, cell therapies are administered systemically, but there is little understanding of their fate, and adverse events are often under-reported. Currently, it is only possible to assess safety and fate of cell therapies in preclinical studies, specifically by monitoring animals longitudinally using multi-modal imaging approaches. Here, using a suite of in vivo imaging modalities to explore the fate of a range of human and murine cells, we investigate how route of administration, cell type and host immune status affect the fate of administered cells. Methods We applied a unique imaging platform combining bioluminescence, optoacoustic and magnetic resonance imaging modalities to assess the safety of different human and murine cell types by following their biodistribution and persistence in mice following administration into the venous or arterial system. Results Longitudinal imaging analyses (i) suggested that the intra-arterial route may be more hazardous than intravenous administration for certain cell types, (ii) revealed that the potential of a mouse mesenchymal stem/stromal cell (MSC) line to form tumours depended on administration route and mouse strain and (iii) indicated that clinically tested human umbilical cord (hUC)-derived MSCs can transiently and unexpectedly proliferate when administered intravenously to mice. Conclusions In order to perform an adequate safety assessment of potential cell-based therapies, a thorough understanding of cell biodistribution and fate post administration is required. The non-invasive imaging platform used here can expose not only the general organ distribution of these therapies, but also a detailed view of their presence within different organs and, importantly, tumourigenic potential. Our observation that the hUC-MSCs but not the human bone marrow (hBM)-derived MSCs persisted for a period in some animals suggests that therapies with these cells should proceed with caution. Electronic supplementary material The online version of this article (10.1186/s13287-018-1076-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lauren Scarfe
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Arthur Taylor
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Jack Sharkey
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Rachel Harwood
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Michael Barrow
- Department of Chemistry, University of Liverpool, Liverpool, UK
| | - Joan Comenge
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lydia Beeken
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Cai Astley
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Ilaria Santeramo
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Claire Hutchinson
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Lorenzo Ressel
- Department of Veterinary Pathology and Public Health, Institute of Veterinary Science, University of Liverpool, Liverpool, UK
| | | | | | - Raphael Levy
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | | | - Dave J Adams
- School of Chemistry, College of Science and Engineering, University of Glasgow, Glasgow, UK
| | - Harish Poptani
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Brian K Park
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK. .,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK. .,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| | - Bettina Wilm
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK. .,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK. .,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| |
Collapse
|
35
|
Prasad R, Chauhan DS, Yadav AS, Devrukhkar J, Singh B, Gorain M, Temgire M, Bellare J, Kundu GC, Srivastava R. A biodegradable fluorescent nanohybrid for photo-driven tumor diagnosis and tumor growth inhibition. NANOSCALE 2018; 10:19082-19091. [PMID: 30288516 DOI: 10.1039/c8nr05164j] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Specific targeting and phototriggered therapy in mouse model have recently emerged as the starting point of cancer theragnosis. Herein, we report a bioresponsive and degradable nanohybrid, a liposomal nanohybrid decorated with red emissive carbon dots, for localized tumor imaging and light-mediated tumor growth inhibition. Unsaturated carbon dots (C-dots) anchored to liposomes convert near-infrared (NIR) light into heat and also produce reactive oxygen species (ROS), demonstrating the capability of phototriggered cancer cell death and tumor regression. The photothermal and oxidative damage of breast tumor by the nonmetallic nanohybrid has also been demonstrated. Designed nanoparticles show excellent aqueous dispersibility, biocompatibility, light irradiated enhanced cellular uptake, release of reactive oxygen species, prolonged and specific tumor binding ability and good photothermal response (62 °C in 5 minutes). Safe and localized irradiation of 808 nm light demonstrates significant tumor growth inhibition and bioresponsive degradation of the fluorescent nanohybrid without affecting the surrounding healthy tissues.
Collapse
Affiliation(s)
- Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Naseroleslami M, Aboutaleb N, Parivar K. The effects of superparamagnetic iron oxide nanoparticles-labeled mesenchymal stem cells in the presence of a magnetic field on attenuation of injury after heart failure. Drug Deliv Transl Res 2018; 8:1214-1225. [PMID: 30128798 DOI: 10.1007/s13346-018-0567-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Migration of stem cells after transplantation reduces their therapeutic effects. In this study, we hypothesized that superparamagnetic iron oxide nanoparticles (SPION)-labeled mesenchymal stem cells (MSCs) in the presence of magnetic field may have a capability to increase regenerative ability after heart failure (HF). A rat model of ISO (isoproterenol)-HF was established to investigate the effects of SPION-labeled MSCs on tissue regeneration in the presence and absence of magnetic field. Hydrodynamic size, shape, and formation of chemical bonds between SPION and polyethylene glycol (PEG) were measured using dynamic light scattering (DLS), transmission electron microscopy (TEM), and Fourier-transform infrared spectroscopy (FTIR). The MRI was used to monitor SPION-labeled MSCs in vivo. Cell and tissue uptake of nanoparticles were determined by Prussian blue staining, atomic absorption spectroscopy (AAS), and inductively coupled plasma spectroscopy (ICP). Purity of the MSCs, heart function, myocardial fibrosis, and histologic damage were evaluated using flow-cytometry, echocardiography, Masson's trichrome, and H&E staining respectively. Various spectroscopic and microscopic analyses revealed that hydrodynamic size of SPION was 40 ± 2 and their shape was spherical. FTIR confirmed the presence of PEG on the surface of nanoparticles. The presence of magnetic field significantly increased cell homing. Highly purified MSCs population was detected by flow-cytometry. Using SPION-labeled MSCs in the presence of magnetic field markedly improved heart function and myocardial hypertrophy and reduced fibrosis (p < 0.05). Collectively, our results demonstrated that SPION-labeled MSCs in the presence of magnetic field might contribute to regeneration after HF.
Collapse
Affiliation(s)
- Maryam Naseroleslami
- Department of Cellular and Molecular Biology, Islamic Azad University Tehran 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.
| | - Kazem Parivar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
37
|
Peng Q, Li Y, Bo S, Yuan Y, Yang Z, Chen S, Zhou X, Jiang ZX. Paramagnetic nanoemulsions with unified signals for sensitive 19F MRI cell tracking. Chem Commun (Camb) 2018; 54:6000-6003. [PMID: 29796466 DOI: 10.1039/c8cc02938e] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As a promising cell tracking technology, 19F MRI suffers from low sensitivity. Here, fluorinated nanoemulsions with a unified 19F signal and paramagnetic relaxation enhancement were developed as 19F MRI cellular tracers with high stability, size controllability, biocompatibility, cellular uptake, and dual-modality for sensitive in vivo RAW264.7 cell tracking.
Collapse
Affiliation(s)
- Qiaoli Peng
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals and School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Lu M, Cheng X, Jiang J, Li T, Zhang Z, Tsauo C, Liu Y, Wang Z. Dual-modal photoacoustic and magnetic resonance tracking of tendon stem cells with PLGA/iron oxide microparticles in vitro. PLoS One 2018; 13:e0193362. [PMID: 29608568 PMCID: PMC5880337 DOI: 10.1371/journal.pone.0193362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/11/2018] [Indexed: 02/07/2023] Open
Abstract
Reliable cell tracking is essential to understand the fate of stem cells following implantation, and thus promote the clinical application of stem cell therapy. Dual or multiple modal imaging modalities mediated by different types of multifunctional contrast agent are generally needed for efficient cell tracking. Here, we created a new contrast agent-PLGA/iron oxide microparticles (PLGA/IO MPs) and characterized the morphology, structure and function of enhancing both photoacoustic (PA) and magnetic resonance imaging (MRI). Both PA and MRI signal increased with increased Fe concentration of PLGA/IO MPs. Fluorescent staining, Prussian blue staining and transmission electron microscope (TEM) certified that PLGA/IO MPs were successfully encapsulated in the labeled TSCs. The established PLGA/IO MPs demonstrated superior ability of dual-modal PA/MRI tracking of TSCs without cytotoxicity at relatively lower Fe concentrations (50, 100 and 200 μg/mL). The optimal Fe concentration of PLGA/IO MPs was determined to be 100 μg/mL, thus laying a foundation for the further study of dual-modal PA/MRI tracking of TSCs in vivo and promoting the repair of injured tendon.
Collapse
Affiliation(s)
- Man Lu
- Chongqing Key laboratory of Ultrasound Molecular Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Ultrasound Medical Center, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xueqing Cheng
- Ultrasound Medical Center, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingzhen Jiang
- Ultrasound Medical Center, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- North Sichuan Medical College, Nanchong, China
| | - TingTing Li
- Ultrasound Medical Center, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenqi Zhang
- Ultrasound Medical Center, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chialing Tsauo
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China
| | - Yin Liu
- Department of Pharmacology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China
- Department of Anesthesiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Zhigang Wang
- Chongqing Key laboratory of Ultrasound Molecular Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
39
|
Chen X, Zhou H, Li X, Duan N, Hu S, Liu Y, Yue Y, Song L, Zhang Y, Li D, Wang Z. Plectin-1 Targeted Dual-modality Nanoparticles for Pancreatic Cancer Imaging. EBioMedicine 2018; 30:129-137. [PMID: 29574092 PMCID: PMC5952251 DOI: 10.1016/j.ebiom.2018.03.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 01/17/2023] Open
Abstract
Background Biomarker-targeted molecular imaging holds promise for early detection of pancreatic cancer. The aim of this study was to design and evaluate a plectin-1 targeted multi-functional nanoparticle probe for pancreatic cancer imaging. Methods 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-amino(polyethylene glycol) (DSPE-PEG-NH2)-modified superparamagnetic iron oxide (Fe3O4) nanoparticles (SPION) were conjugated with plectin-1 antibody and/or Cy7 to create the multi-functional targeted nanoparticle targeted probe (Plectin-SPION-Cy7) or non-targeted probe (SPION-Cy7). Pancreatic carcinoma cell lines expressing plectin-1 were cultured with the targeted or control probes and then were imaged using confocal laser scanning microscopy and magnetic resonance imaging (MRI). Accumulations of the nanoparticles in pancreatic tumor xenografted mice were determined by MRI and fluorescence imaging. Results In vitro optical imaging and MRI showed that the targeted nanoparticles were highly accumulated in MIAPaCa2 and XPA-1 carcinoma cells but not in non-carcinoma MIN6 cells, which was further confirmed by Prussian blue staining. In vivo MRI showed a significant T2 signal reduction. Prussian blue staining further confirmed that the plectin-1 targeted nanoparticles were highly accumulated in the tumor mass but not in normal pancreatic tissues, or in the liver and kidney, and few nanoparticles were observed in the tumors of mice injected with SPION-Cy7. Conclusions Our data demonstrate that plectin-1 targeted fluorescence and MR dual-functional nanoparticle can visualize pancreatic cancer, and it has great potential to be used with various imaging devices for pancreatic cancer detection. We designed a plectin-1 targeted dual-modality nanoparticle (Plectin-SPION-Cy7). The targeted nanoparticles were highly accumulated in carcinoma cells but not in non-carcinoma cells. Plectin-1 targeted dual-functional nanoparticle has great potential in pancreatic cancer detection.
Molecular-based radiographic tests hold the promise to help precisely identifying pancreatic malignant lesions and their precursors at early stages. Previous studies showed that plectin-1 highly expressed in pancreatic ductal adenocarcinoma but not in non-carcinoma tissues. In the current study, we designed a plectin-1 targeted dual-modality nanoparticle (Plectin-SPION-Cy7). In vitro and in vivo data both indicated that plectin-1 targeted nanoparticles were highly accumulated in carcinoma cells/tissues but not in non-carcinoma cells/tissues. These results show that plectin-1 targeted fluorescence and MR dual-functional nanoparticle is useful for pancreatic cancer detection.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Division of Nephrology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Hao Zhou
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Xiaoshuang Li
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Na Duan
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Shouyou Hu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Yongkang Liu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Yali Yue
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Lina Song
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Yifen Zhang
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
40
|
Chauhan DS, Prasad R, Devrukhkar J, Selvaraj K, Srivastava R. Disintegrable NIR Light Triggered Gold Nanorods Supported Liposomal Nanohybrids for Cancer Theranostics. Bioconjug Chem 2018; 29:1510-1518. [PMID: 29281790 DOI: 10.1021/acs.bioconjchem.7b00801] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this work, facile synthesis and application of targeted, dual therapeutic gold nanorods-liposome (GNR-Lipos) nanohybrid for imaging guided photothermal therapy and chemotherapy is investigated. The dual therapeutic GNR-Lipos nanohybrid consists of GNR supported, and doxorubicin (DOX) loaded liposome. GNRs not only serve as a photothermal agent and increase the drug release in intracellular environment of cancer cells, but also provide mechanical strength to liposomes by being decorated both inside and outside of bilayer surfaces. The designed nanohybrid shows a remarkable response for synergistic chemophotothermal therapy compared to only chemotherapy or photothermal therapy. The NIR response, efficient uptake by the cells, disintegration of GNR-Lipos nanohybrid, and synergistic therapeutic effect of photothermal and chemotherapy over breast cancer cells MDA-MB-231 are studied for the better development of a biocompatible nanomaterial based multifunctional cancer theranostic agent.
Collapse
Affiliation(s)
- Deepak S Chauhan
- Department of Biosciences and Bioengineering , IIT Bombay , Powai, Mumbai - 400076 , India
| | | | - Janhavi Devrukhkar
- Department of Biosciences and Bioengineering , IIT Bombay , Powai, Mumbai - 400076 , India
| | | | - Rohit Srivastava
- Department of Biosciences and Bioengineering , IIT Bombay , Powai, Mumbai - 400076 , India
| |
Collapse
|
41
|
Barrow M, Taylor A, Fuentes-Caparrós AM, Sharkey J, Daniels LM, Mandal P, Park BK, Murray P, Rosseinsky MJ, Adams DJ. SPIONs for cell labelling and tracking using MRI: magnetite or maghemite? Biomater Sci 2017; 6:101-106. [PMID: 29188240 PMCID: PMC5793703 DOI: 10.1039/c7bm00515f] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/23/2017] [Indexed: 12/15/2022]
Abstract
Although there is extensive literature covering the biomedical applications of superparamagnetic iron oxide nanoparticles (SPIONs), the phase of the iron oxide core used is not often taken into account when cell labelling and tracking studies for regenerative medicine are considered. Here, we use a co-precipitation reaction to synthesise particles of both magnetite- (Fe3O4) and maghemite- (γ-Fe2O3) based cores and consider whether the extra synthesis step to make maghemite based particles is advantageous for cell tracking.
Collapse
Affiliation(s)
- Michael Barrow
- Department of Chemistry , University of Liverpool , Liverpool , UK . ;
| | - Arthur Taylor
- Centre for Preclinical Imaging , Institute of Translational Medicine , University of Liverpool , Liverpool , UK
| | | | - Jack Sharkey
- Centre for Preclinical Imaging , Institute of Translational Medicine , University of Liverpool , Liverpool , UK
| | - Luke M. Daniels
- Department of Chemistry , University of Liverpool , Liverpool , UK . ;
| | - Pranab Mandal
- Department of Chemistry , University of Liverpool , Liverpool , UK . ;
| | - B. Kevin Park
- MRC Centre for Drug Safety Science , Department of Clinical and Molecular Pharmacology , University of Liverpool , Liverpool , UK
| | - Patricia Murray
- Centre for Preclinical Imaging , Institute of Translational Medicine , University of Liverpool , Liverpool , UK
| | | | - Dave J. Adams
- Department of Chemistry , University of Liverpool , Liverpool , UK . ;
- School of Chemistry , College of Science and Engineering , University of Glasgow , Glasgow , G12 8QQ , UK
| |
Collapse
|
42
|
Andreiuk B, Reisch A, Lindecker M, Follain G, Peyriéras N, Goetz JG, Klymchenko AS. Fluorescent Polymer Nanoparticles for Cell Barcoding In Vitro and In Vivo. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1701582. [PMID: 28791769 DOI: 10.1002/smll.201701582] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 06/26/2017] [Indexed: 06/07/2023]
Abstract
Fluorescent polymer nanoparticles for long-term labeling and tracking of living cells with any desired color code are developed. They are built from biodegradable poly(lactic-co-glycolic acid) polymer loaded with cyanine dyes (DiO, DiI, and DiD) with the help of bulky fluorinated counterions, which minimize aggregation-caused quenching. At the single particle level, these particles are ≈20-fold brighter than quantum dots of similar color. Due to their identical 40 nm size and surface properties, these nanoparticles are endocytosed equally well by living cells. Mixing nanoparticles of three colors in different proportions generates a homogeneous RGB (red, green, and blue) barcode in cells, which is transmitted through many cell generations. Cell barcoding is validated on 7 cell lines (HeLa, KB, embryonic kidney (293T), Chinese hamster ovary, rat basophilic leucemia, U97, and D2A1), 13 color codes, and it enables simultaneous tracking of co-cultured barcoded cell populations for >2 weeks. It is also applied to studying competition among drug-treated cell populations. This technology enabled six-color imaging in vivo for (1) tracking xenografted cancer cells and (2) monitoring morphogenesis after microinjection in zebrafish embryos. In addition to a robust method of multicolor cell labeling and tracking, this work suggests that multiple functions can be co-localized inside cells by combining structurally close nanoparticles carrying different functions.
Collapse
Affiliation(s)
- Bohdan Andreiuk
- Laboratoire de Biophotonique et Pharmacologie, UMR 7213 CNRS, Faculté de Pharmacie, Université de Strasbourg, 74, Route du Rhin, BP 60024, 67401, Illkirch, France
| | - Andreas Reisch
- Laboratoire de Biophotonique et Pharmacologie, UMR 7213 CNRS, Faculté de Pharmacie, Université de Strasbourg, 74, Route du Rhin, BP 60024, 67401, Illkirch, France
| | - Marion Lindecker
- Laboratoire de Biophotonique et Pharmacologie, UMR 7213 CNRS, Faculté de Pharmacie, Université de Strasbourg, 74, Route du Rhin, BP 60024, 67401, Illkirch, France
| | - Gautier Follain
- MN3T, Inserm U1109, LabEx Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, 67000, France
| | - Nadine Peyriéras
- CNRS USR3695 BioEmergences, Avenue de la Terrasse, 91190, Gif-sur-Yvette, France
| | - Jacky G Goetz
- MN3T, Inserm U1109, LabEx Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, 67000, France
| | - Andrey S Klymchenko
- Laboratoire de Biophotonique et Pharmacologie, UMR 7213 CNRS, Faculté de Pharmacie, Université de Strasbourg, 74, Route du Rhin, BP 60024, 67401, Illkirch, France
| |
Collapse
|
43
|
Quantum dots mediated embryotoxicity via placental damage. Reprod Toxicol 2017; 73:222-231. [DOI: 10.1016/j.reprotox.2017.08.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 08/05/2017] [Accepted: 08/18/2017] [Indexed: 11/17/2022]
|
44
|
Joris F, Valdepérez D, Pelaz B, Wang T, Doak SH, Manshian BB, Soenen SJ, Parak WJ, De Smedt SC, Raemdonck K. Choose your cell model wisely: The in vitro nanoneurotoxicity of differentially coated iron oxide nanoparticles for neural cell labeling. Acta Biomater 2017; 55:204-213. [PMID: 28373085 DOI: 10.1016/j.actbio.2017.03.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 02/06/2023]
Abstract
Currently, there is a large interest in the labeling of neural stem cells (NSCs) with iron oxide nanoparticles (IONPs) to allow MRI-guided detection after transplantation in regenerative medicine. For such biomedical applications, excluding nanotoxicity is key. Nanosafety is primarily evaluated in vitro where an immortalized or cancer cell line of murine origin is often applied, which is not necessarily an ideal cell model. Previous work revealed clear neurotoxic effects of PMA-coated IONPs in distinct cell types that could potentially be applied for nanosafety studies regarding neural cell labeling. Here, we aimed to assess if DMSA-coated IONPs could be regarded as a safer alternative for this purpose and how the cell model impacted our nanosafety optimization study. Hereto, we evaluated cytotoxicity, ROS production, calcium levels, mitochondrial homeostasis and cell morphology in six related neural cell types, namely neural stem cells, an immortalized cell line and a cancer cell line from human and murine origin. The cell lines mostly showed similar responses to both IONPs, which were frequently more pronounced for the PMA-IONPs. Of note, ROS and calcium levels showed opposite trends in the human and murine NSCs, indicating the importance of the species. Indeed, the human cell models were overall more sensitive than their murine counterpart. Despite the clear cell type-specific nanotoxicity profiles, our multiparametric approach revealed that the DMSA-IONPs outperformed the PMA-IONPs in terms of biocompatibility in each cell type. However, major cell type-dependent variations in the observed effects additionally warrant the use of relevant human cell models. STATEMENT OF SIGNIFICANCE Inorganic nanoparticle (NP) optimization is chiefly performed in vitro. For the optimization of iron oxide (IO)NPs for neural stem cell labeling in the context of regenerative medicine human or rodent neural stem cells, immortalized or cancer cell lines are applied. However, the use of certain cell models can be questioned as they phenotypically differ from the target cell. The impact of the neural cell model on nanosafety remains relatively unexplored. Here we evaluated cell homeostasis upon exposure to PMA- and DMSA-coated IONPs. Of note, the DMSA-IONPs outperformed the PMA-IONPs in each cell type. However, distinct cell type-specific effects were witnessed, indicating that nanosafety should be evaluated in a human cell model that represents the target cell as closely as possible.
Collapse
Affiliation(s)
- Freya Joris
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Daniel Valdepérez
- Philipps University of Marburg, Department of Physics, Renthof 7, D-35037 Marburg, Germany
| | - Beatriz Pelaz
- Philipps University of Marburg, Department of Physics, Renthof 7, D-35037 Marburg, Germany
| | - Tianqiang Wang
- Philipps University of Marburg, Department of Physics, Renthof 7, D-35037 Marburg, Germany
| | - Shareen H Doak
- Institute of Life Sciences, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK
| | - Bella B Manshian
- Biomedical MRI Unit/MoSAIC, Department of Medicine, KULeuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Stefaan J Soenen
- Biomedical MRI Unit/MoSAIC, Department of Medicine, KULeuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Wolfgang J Parak
- Philipps University of Marburg, Department of Physics, Renthof 7, D-35037 Marburg, Germany
| | - Stefaan C De Smedt
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium.
| | - Koen Raemdonck
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| |
Collapse
|
45
|
Cheng Y, Sun C, Ou X, Liu B, Lou X, Xia F. Dual-targeted peptide-conjugated multifunctional fluorescent probe with AIEgen for efficient nucleus-specific imaging and long-term tracing of cancer cells. Chem Sci 2017; 8:4571-4578. [PMID: 28626568 PMCID: PMC5471453 DOI: 10.1039/c7sc00402h] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/19/2017] [Indexed: 12/20/2022] Open
Abstract
Precisely targeted transportation of a long-term tracing regent to a nucleus with low toxicity is one of the most challenging concerns in revealing cancer cell behaviors. Here, we report a dual-targeted peptide-conjugated multifunctional fluorescent probe (cNGR-CPP-NLS-RGD-PyTPE, TCNTP) with aggregation-induced emission (AIE) characteristic, for efficient nucleus-specific imaging and long-term and low-toxicity tracing of cancer cells. TCNTP mainly consists of two components: one is a functionalized combinatorial peptide (TCNT) containing two targeted peptides (cNGR and RGD), a cell-penetrating peptide (CPP) and a nuclear localization signal (NLS), which can specifically bind to a cell surface and effectively enter into the nucleus; the other one is an AIE-active tetraphenylethene derivative (PyTPE, a typical AIEgen) as fluorescence imaging reagent. In the presence of aminopeptidase N (CD13) and integrin αvβ3, TCNTP can specifically bind to both of them using cNGR and RGD, respectively, lighting up its yellow fluorescence. Because it contains CPP, TCNTP can be effectively integrated into the cytoplasm, and then be delivered into the nucleus with the help of NLS. TCNTP exhibited strong fluorescence in the nucleus of CD13 and integrin αvβ3 overexpression cells due to the specific targeting ability, efficient transport capacity and AIE characteristic in a more crowded space. Furthermore, TCNTP can be applied for long-term tracing in living cells, scarcely affecting normal cells with negligible toxicity in more than ten passages.
Collapse
Affiliation(s)
- Yong Cheng
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
- National Engineering Research Center for Nanomedicine , Department of Biomedical Engineering , College of Life Science and Technology , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China
| | - Chunli Sun
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
| | - Xiaowen Ou
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
| | - Bifeng Liu
- National Engineering Research Center for Nanomedicine , Department of Biomedical Engineering , College of Life Science and Technology , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China
| | - Xiaoding Lou
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
| | - Fan Xia
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
- National Engineering Research Center for Nanomedicine , Department of Biomedical Engineering , College of Life Science and Technology , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China
| |
Collapse
|
46
|
Elgqvist J. Nanoparticles as Theranostic Vehicles in Experimental and Clinical Applications-Focus on Prostate and Breast Cancer. Int J Mol Sci 2017; 18:E1102. [PMID: 28531102 PMCID: PMC5455010 DOI: 10.3390/ijms18051102] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/13/2017] [Accepted: 05/15/2017] [Indexed: 12/27/2022] Open
Abstract
Prostate and breast cancer are the second most and most commonly diagnosed cancer in men and women worldwide, respectively. The American Cancer Society estimates that during 2016 in the USA around 430,000 individuals were diagnosed with one of these two types of cancers, and approximately 15% of them will die from the disease. In Europe, the rate of incidences and deaths are similar to those in the USA. Several different more or less successful diagnostic and therapeutic approaches have been developed and evaluated in order to tackle this issue and thereby decrease the death rates. By using nanoparticles as vehicles carrying both diagnostic and therapeutic molecular entities, individualized targeted theranostic nanomedicine has emerged as a promising option to increase the sensitivity and the specificity during diagnosis, as well as the likelihood of survival or prolonged survival after therapy. This article presents and discusses important and promising different kinds of nanoparticles, as well as imaging and therapy options, suitable for theranostic applications. The presentation of different nanoparticles and theranostic applications is quite general, but there is a special focus on prostate cancer. Some references and aspects regarding breast cancer are however also presented and discussed. Finally, the prostate cancer case is presented in more detail regarding diagnosis, staging, recurrence, metastases, and treatment options available today, followed by possible ways to move forward applying theranostics for both prostate and breast cancer based on promising experiments performed until today.
Collapse
Affiliation(s)
- Jörgen Elgqvist
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden.
- Department of Physics, University of Gothenburg, 412 96 Gothenburg, Sweden.
| |
Collapse
|
47
|
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
|
48
|
Martins IM, Reis RL, Azevedo HS. Phage Display Technology in Biomaterials Engineering: Progress and Opportunities for Applications in Regenerative Medicine. ACS Chem Biol 2016; 11:2962-2980. [PMID: 27661443 DOI: 10.1021/acschembio.5b00717] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The field of regenerative medicine has been gaining momentum steadily over the past few years. The emphasis in regenerative medicine is to use various in vitro and in vivo approaches that leverage the intrinsic healing mechanisms of the body to treat patients with disabling injuries and chronic diseases such as diabetes, osteoarthritis, and degenerative disorders of the cardiovascular and central nervous system. Phage display has been successfully employed to identify peptide ligands for a wide variety of targets, ranging from relatively small molecules (enzymes, cell receptors) to inorganic, organic, and biological (tissues) materials. Over the past two decades, phage display technology has advanced tremendously and has become a powerful tool in the most varied fields of research, including biotechnology, materials science, cell biology, pharmacology, and diagnostics. The growing interest in and success of phage display libraries is largely due to its incredible versatility and practical use. This review discusses the potential of phage display technology in biomaterials engineering for applications in regenerative medicine.
Collapse
Affiliation(s)
- Ivone M. Martins
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- CEB − Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
| | - Rui L. Reis
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Helena S. Azevedo
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
- Institute
of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom
| |
Collapse
|
49
|
Liu W, Naydenov B, Chakrabortty S, Wuensch B, Hübner K, Ritz S, Cölfen H, Barth H, Koynov K, Qi H, Leiter R, Reuter R, Wrachtrup J, Boldt F, Scheuer J, Kaiser U, Sison M, Lasser T, Tinnefeld P, Jelezko F, Walther P, Wu Y, Weil T. Fluorescent Nanodiamond-Gold Hybrid Particles for Multimodal Optical and Electron Microscopy Cellular Imaging. NANO LETTERS 2016; 16:6236-6244. [PMID: 27629492 DOI: 10.1021/acs.nanolett.6b02456] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
There is a continuous demand for imaging probes offering excellent performance in various microscopy techniques for comprehensive investigations of cellular processes by more than one technique. Fluorescent nanodiamond-gold nanoparticles (FND-Au) constitute a new class of "all-in-one" hybrid particles providing unique features for multimodal cellular imaging including optical imaging, electron microscopy, and, and potentially even quantum sensing. Confocal and optical coherence microscopy of the FND-Au allow fast investigations inside living cells via emission, scattering, and photothermal imaging techniques because the FND emission is not quenched by AuNPs. In electron microscopy, transmission electron microscopy (TEM) and scanning transmission electron microscopy (STEM) analysis of FND-Au reveals greatly enhanced contrast due to the gold particles as well as an extraordinary flickering behavior in three-dimensional cellular environments originating from the nanodiamonds. The unique multimodal imaging characteristics of FND-Au enable detailed studies inside cells ranging from statistical distributions at the entire cellular level (micrometers) down to the tracking of individual particles in subcellular organelles (nanometers). Herein, the processes of endosomal membrane uptake and release of FNDs were elucidated for the first time by the imaging of individual FND-Au hybrid nanoparticles with single-particle resolution. Their convenient preparation, the availability of various surface groups, their flexible detection modalities, and their single-particle contrast in combination with the capability for endosomal penetration and low cytotoxicity make FND-Au unique candidates for multimodal optical-electronic imaging applications with great potential for emerging techniques, such as quantum sensing inside living cells.
Collapse
Affiliation(s)
- Weina Liu
- Department of Organic Chemistry III/Macromolecular Chemistry, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| | - Boris Naydenov
- Institute for Quantum Optics, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Sabyasachi Chakrabortty
- Department of Organic Chemistry III/Macromolecular Chemistry, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| | - Bettina Wuensch
- NanoBioSciences Group, Institute for Physical & Theoretical Chemistry, and Braunschweig Integrated Centre of Systems Biology (BRICS), and Laboratory for Emerging Nanometrology (LENA), Braunschweig University of Technology , Pockelsstrasse 14, 38106 Braunschweig, Germany
| | - Kristina Hübner
- NanoBioSciences Group, Institute for Physical & Theoretical Chemistry, and Braunschweig Integrated Centre of Systems Biology (BRICS), and Laboratory for Emerging Nanometrology (LENA), Braunschweig University of Technology , Pockelsstrasse 14, 38106 Braunschweig, Germany
| | - Sandra Ritz
- Institute of Molecular Biology (IMB) GmbH , Ackermannweg 4, 55128 Mainz, Germany
| | - Helmut Cölfen
- Physical Chemistry, Department of Chemistry, University of Konstanz , Universitätsstraße 10, 78457 Konstanz, Germany
| | - Holger Barth
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center , Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Kaloian Koynov
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| | - Haoyuan Qi
- Central Facility for Electron Microscopy, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Robert Leiter
- Central Facility for Electron Microscopy, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Rolf Reuter
- Institute of Physics, University of Stuttgart , Pfaffenwaldring 57, 70569 Stuttgart, Germany
| | - Jörg Wrachtrup
- Institute of Physics, University of Stuttgart , Pfaffenwaldring 57, 70569 Stuttgart, Germany
| | - Felix Boldt
- Department of Organic Chemistry III/Macromolecular Chemistry, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| | - Jonas Scheuer
- Institute for Quantum Optics, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Ute Kaiser
- Central Facility for Electron Microscopy, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Miguel Sison
- Laboratoire d'Optique Biomédicale, École Polytechnique Fédérale de Lausanne , BM 5143, Station 17, CH-1015 Lausanne, Switzerland
| | - Theo Lasser
- Laboratoire d'Optique Biomédicale, École Polytechnique Fédérale de Lausanne , BM 5143, Station 17, CH-1015 Lausanne, Switzerland
| | - Philip Tinnefeld
- NanoBioSciences Group, Institute for Physical & Theoretical Chemistry, and Braunschweig Integrated Centre of Systems Biology (BRICS), and Laboratory for Emerging Nanometrology (LENA), Braunschweig University of Technology , Pockelsstrasse 14, 38106 Braunschweig, Germany
| | - Fedor Jelezko
- Institute for Quantum Optics, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Yuzhou Wu
- Department of Organic Chemistry III/Macromolecular Chemistry, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology , 430074 Wuhan, P. R. China
| | - Tanja Weil
- Department of Organic Chemistry III/Macromolecular Chemistry, Ulm University , Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
50
|
Xiong R, Joris F, Liang S, De Rycke R, Lippens S, Demeester J, Skirtach A, Raemdonck K, Himmelreich U, De Smedt SC, Braeckmans K. Cytosolic Delivery of Nanolabels Prevents Their Asymmetric Inheritance and Enables Extended Quantitative in Vivo Cell Imaging. NANO LETTERS 2016; 16:5975-5986. [PMID: 27684962 DOI: 10.1021/acs.nanolett.6b01411] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Long-term in vivo imaging of cells is crucial for the understanding of cellular fate in biological processes in cancer research, immunology, or in cell-based therapies such as beta cell transplantation in type I diabetes or stem cell therapy. Traditionally, cell labeling with the desired contrast agent occurs ex vivo via spontaneous endocytosis, which is a variable and slow process that requires optimization for each particular label-cell type combination. Following endocytic uptake, the contrast agents mostly remain entrapped in the endolysosomal compartment, which leads to signal instability, cytotoxicity, and asymmetric inheritance of the labels upon cell division. Here, we demonstrate that these disadvantages can be circumvented by delivering contrast agents directly into the cytoplasm via vapor nanobubble photoporation. Compared to classic endocytic uptake, photoporation resulted in 50 and 3 times higher loading of fluorescent dextrans and quantum dots, respectively, with improved signal stability and reduced cytotoxicity. Most interestingly, cytosolic delivery by photoporation prevented asymmetric inheritance of labels by daughter cells over subsequent cell generations. Instead, unequal inheritance of endocytosed labels resulted in a dramatic increase in polydispersity of the amount of labels per cell with each cell division, hindering accurate quantification of cell numbers in vivo over time. The combined benefits of cell labeling by photoporation resulted in a marked improvement in long-term cell visibility in vivo where an insulin producing cell line (INS-1E cell line) labeled with fluorescent dextrans could be tracked for up to two months in Swiss nude mice compared to 2 weeks for cells labeled by endocytosis.
Collapse
Affiliation(s)
- Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
- Centre for Nano- and Biophotonics, Ghent University , 9000 Ghent, Belgium
| | - Freya Joris
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
| | - Sayuan Liang
- Biomedical NMR Unit, Faculty of Medicine, Katholieke Universiteit Leuven , 3000 Leuven, Belgium
| | - Riet De Rycke
- Inflammation Research Center, Image Core Facility, VIB , 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University , 9052 Ghent, Belgium
| | - Saskia Lippens
- Inflammation Research Center, Image Core Facility, VIB , 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University , 9052 Ghent, Belgium
| | - Jo Demeester
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
| | - Andre Skirtach
- Department of Molecular Biotechnology, Ghent University , 9000 Ghent, Belgium
- Max-Planck Institute of Colloids and Interfaces , 14424 Potsdam, Germany
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
| | - Uwe Himmelreich
- Biomedical NMR Unit, Faculty of Medicine, Katholieke Universiteit Leuven , 3000 Leuven, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
- Centre for Nano- and Biophotonics, Ghent University , 9000 Ghent, Belgium
- Univ Lille 1, Univ Lille Nord France, IEMN, UMR 8520, 59652 Villeneuve Dascq, France
- Univ Lille 1, Univ Lille Nord France, Lab Phys Lasers Atomes & Mol, UMR 8523, 59655 Villeneuve Dascq, France
| |
Collapse
|