1
|
Shan Y, Zhang M, Tao E, Wang J, Wei N, Lu Y, Liu Q, Hao K, Zhou F, Wang G. Pharmacokinetic characteristics of mesenchymal stem cells in translational challenges. Signal Transduct Target Ther 2024; 9:242. [PMID: 39271680 PMCID: PMC11399464 DOI: 10.1038/s41392-024-01936-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/15/2024] Open
Abstract
Over the past two decades, mesenchymal stem/stromal cell (MSC) therapy has made substantial strides, transitioning from experimental clinical applications to commercial products. MSC therapies hold considerable promise for treating refractory and critical conditions such as acute graft-versus-host disease, amyotrophic lateral sclerosis, and acute respiratory distress syndrome. Despite recent successes in clinical and commercial applications, MSC therapy still faces challenges when used as a commercial product. Current detection methods have limitations, leaving the dynamic biodistribution, persistence in injured tissues, and ultimate fate of MSCs in patients unclear. Clarifying the relationship between the pharmacokinetic characteristics of MSCs and their therapeutic effects is crucial for patient stratification and the formulation of precise therapeutic regimens. Moreover, the development of advanced imaging and tracking technologies is essential to address these clinical challenges. This review provides a comprehensive analysis of the kinetic properties, key regulatory molecules, different fates, and detection methods relevant to MSCs and discusses concerns in evaluating MSC druggability from the perspective of integrating pharmacokinetics and efficacy. A better understanding of these challenges could improve MSC clinical efficacy and speed up the introduction of MSC therapy products to the market.
Collapse
Affiliation(s)
- Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Mengying Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Enxiang Tao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jing Wang
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Ning Wei
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Yi Lu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qing Liu
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Kun Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
2
|
Yadav P, Singh SK, Rajput S, Allawadhi P, Khurana A, Weiskirchen R, Navik U. Therapeutic potential of stem cells in regeneration of liver in chronic liver diseases: Current perspectives and future challenges. Pharmacol Ther 2024; 253:108563. [PMID: 38013053 DOI: 10.1016/j.pharmthera.2023.108563] [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: 09/15/2023] [Revised: 11/04/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
The deposition of extracellular matrix and hyperplasia of connective tissue characterizes chronic liver disease called hepatic fibrosis. Progression of hepatic fibrosis may lead to hepatocellular carcinoma. At this stage, only liver transplantation is a viable option. However, the number of possible liver donors is less than the number of patients needing transplantation. Consequently, alternative cell therapies based on non-stem cells (e.g., fibroblasts, chondrocytes, keratinocytes, and hepatocytes) therapy may be able to postpone hepatic disease, but they are often ineffective. Thus, novel stem cell-based therapeutics might be potentially important cutting-edge approaches for treating liver diseases and reducing patient' suffering. Several signaling pathways provide targets for stem cell interventions. These include pathways such as TGF-β, STAT3/BCL-2, NADPH oxidase, Raf/MEK/ERK, Notch, and Wnt/β-catenin. Moreover, mesenchymal stem cells (MSCs) stimulate interleukin (IL)-10, which inhibits T-cells and converts M1 macrophages into M2 macrophages, producing an anti-inflammatory environment. Furthermore, it inhibits the action of CD4+ and CD8+ T cells and reduces the activity of TNF-α and interferon cytokines by enhancing IL-4 synthesis. Consequently, the immunomodulatory and anti-inflammatory capabilities of MSCs make them an attractive therapeutic approach. Importantly, MSCs can inhibit the activation of hepatic stellate cells, causing their apoptosis and subsequent promotion of hepatocyte proliferation, thereby replacing dead hepatocytes and reducing liver fibrosis. This review discusses the multidimensional therapeutic role of stem cells as cell-based therapeutics in liver fibrosis.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sumeet Kumar Singh
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sonu Rajput
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Prince Allawadhi
- Department of Pharmacy, Vaish Institute of Pharmaceutical Education and Research (VIPER), Pandit Bhagwat Dayal Sharma University of Health Sciences (Pt. B. D. S. UHS), Rohtak, Haryana 124001, India
| | - Amit Khurana
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| |
Collapse
|
3
|
Armstrong M, Wilken E, Freppon F, Masthoff M, Faber C, Xiao D. Dynamic cell tracking using time-lapse MRI with variable temporal resolution Cartesian sampling. Magn Reson Med 2023; 90:2443-2453. [PMID: 37466029 DOI: 10.1002/mrm.29796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 06/03/2023] [Accepted: 06/25/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE Temporal resolution of time-lapse MRI to track individual iron-labeled cells is limited by the required data-acquisition time to fill k-space and to reach sufficient SNR. Although motion of slowly patrolling monocytes can be resolved, detection of fast-moving immune cells requires improved acquisition and reconstruction strategies. THEORY AND METHODS For accelerated MRI cell tracking, a Cartesian sampling scheme was designed, in which the fully sampled and undersampled k-space data for different acceleration factors were acquired simultaneously, and multiple undersampling ratios could be chosen retrospectively. Compressed-sensing reconstruction was applied using dictionary learning and low-rank constraints. Detection of iron-labeled monocytes was evaluated with simulations, rotating phantom experiments and in vivo mouse brain measurements at 9.4 T. RESULTS Fully sampled and 2.4-times and 4.8-times accelerated images were reconstructed and had sufficient contrast-to-noise ratio (CNR) for single cells to be resolved and followed dynamically. The phantom experiments showed an improvement in CNR of 6.1% per μm/s in the 4.8-times undersampled images. Geometric distortion of cells caused by motion was visibly reduced in the accelerated images, which enabled detection of moving cells with velocities of up to 7.0 μm/s. In vivo, additional cells were resolved in the accelerated images due to the improved temporal resolution. CONCLUSION The easy-to-implement flexible Cartesian sampling scheme with compressed-sensing reconstruction permits simultaneous acquisition of both fully sampled and high temporal resolution images. The CNR of moving cells is effectively improved, enabling the recovery of high velocity cells with sufficient contrast at virtually no cost.
Collapse
Affiliation(s)
- Mark Armstrong
- Physics Department, University of Windsor, Windsor, Canada
| | - Enrica Wilken
- Clinic for Radiology, University of Münster, Münster, Germany
| | - Felix Freppon
- Clinic for Radiology, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic for Radiology, University of Münster, Münster, Germany
| | - Cornelius Faber
- Clinic for Radiology, University of Münster, Münster, Germany
| | - Dan Xiao
- Physics Department, University of Windsor, Windsor, Canada
| |
Collapse
|
4
|
Alizadeh R, Asghari A, Taghizadeh-Hesary F, Moradi S, Farhadi M, Mehdizadeh M, Simorgh S, Nourazarian A, Shademan B, Susanabadi A, Kamrava K. Intranasal delivery of stem cells labeled by nanoparticles in neurodegenerative disorders: Challenges and opportunities. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1915. [PMID: 37414546 DOI: 10.1002/wnan.1915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 05/05/2023] [Accepted: 06/11/2023] [Indexed: 07/08/2023]
Abstract
Neurodegenerative disorders occur through progressive loss of function or structure of neurons, with loss of sensation and cognition values. The lack of successful therapeutic approaches to solve neurologic disorders causes physical disability and paralysis and has a significant socioeconomic impact on patients. In recent years, nanocarriers and stem cells have attracted tremendous attention as a reliable approach to treating neurodegenerative disorders. In this regard, nanoparticle-based labeling combined with imaging technologies has enabled researchers to survey transplanted stem cells and fully understand their fate by monitoring their survival, migration, and differentiation. For the practical implementation of stem cell therapies in the clinical setting, it is necessary to accurately label and follow stem cells after administration. Several approaches to labeling and tracking stem cells using nanotechnology have been proposed as potential treatment strategies for neurological diseases. Considering the limitations of intravenous or direct stem cell administration, intranasal delivery of nanoparticle-labeled stem cells in neurological disorders is a new method of delivering stem cells to the central nervous system (CNS). This review describes the challenges and limitations of stem cell-based nanotechnology methods for labeling/tracking, intranasal delivery of cells, and cell fate regulation as theragnostic labeling. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Rafieh Alizadeh
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alimohamad Asghari
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Salah Moradi
- Department of Life Science Engineering, Faculty of New Science and Technology, University of Tehran, Tehran, Iran
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Susanabadi
- Department of Anesthesia and Pain Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Kamran Kamrava
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Chen Y, Hou S. Recent progress in the effect of magnetic iron oxide nanoparticles on cells and extracellular vesicles. Cell Death Discov 2023; 9:195. [PMID: 37380637 DOI: 10.1038/s41420-023-01490-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/30/2023] Open
Abstract
At present, iron oxide nanoparticles (IONPs) are widely used in the biomedical field. They have unique advantages in targeted drug delivery, imaging and disease treatment. However, there are many things to pay attention to. In this paper, we reviewed the fate of IONPs in different cells and the influence on the production, separation, delivery and treatment of extracellular vesicles. It aims to provide cutting-edge knowledge related to iron oxide nanoparticles. Only by ensuring the safety and effectiveness of IONPs can their application in biomedical research and clinic be further improved.
Collapse
Affiliation(s)
- Yuling Chen
- Institute of Disaster and Emergency Medicine, Tianjin University, 300072, Tianjin, China.
- Key Laboratory for Disaster Medicine Technology, 300072, Tianjin, China.
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, 300072, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, 300072, Tianjin, China
| |
Collapse
|
6
|
Nazeer SS, Saraswathy A, Nimi N, Sarathkumar E, Resmi AN, Shenoy SJ, Jayasree RS. Fluorescent carbon dots tailored iron oxide nano hybrid system for in vivooptical imaging of liver fibrosis. Methods Appl Fluoresc 2023; 11. [PMID: 36854197 DOI: 10.1088/2050-6120/acc009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 02/28/2023] [Indexed: 03/02/2023]
Abstract
Hybrid nanoparticles are innovative invention of last decade designed to overcome limitations of single-component nanoparticles by introducing multiple functionalities through combining two or more different nanoparticles. In this study, we are reporting development of magneto-fluorescent hybrid nanoparticles by combining iron oxide and carbon nanoparticles to enablein vivofluorescence imaging which also has all the required characteristic properties to use as Magnetic Resonance Imaging (MRI) contrast agent. In order to achieve dual-functional imaging, alginate and pullulan coated super paramagnetic iron oxide nanoparticles (ASPION and PSPION) and Carbon dots (Cdts) were synthesised separately. ASPIONs and PSPIONs were further chemically conjugated with Cdts and developed dual-functional nanohybrid particles ASPION-Cdts and PSPION-Cdts. Subsequently, evaluation of the materials for its size, functionalisation efficiency, fluorescence and magnetic properties, biocompatibility and cellular uptake efficiency has been carried out. Fluorescence imaging of liver fibrosis was performedin vivoin rodent model of liver fibrosis using the two nanohybrids, which is further confirmed by high fluorescence signal from the harvested liver.
Collapse
Affiliation(s)
- Shaiju S Nazeer
- Department of Chemistry, Indian Institute of Space Sciences and Technology, Thiruvananthapuram-695547, Kerala, India
- Division of Biophotonics and Imaging, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences &Technology, Poojappura, Thiruvananthapuram-695012, Kerala, India
| | - Ariya Saraswathy
- Department of Physics, HHMSPBNSS College, Thiruvananthapuram-695040, Kerala, India
- Division of Biophotonics and Imaging, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences &Technology, Poojappura, Thiruvananthapuram-695012, Kerala, India
| | - Nirmala Nimi
- Division of Biophotonics and Imaging, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences &Technology, Poojappura, Thiruvananthapuram-695012, Kerala, India
| | - Elangovan Sarathkumar
- Division of Biophotonics and Imaging, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences &Technology, Poojappura, Thiruvananthapuram-695012, Kerala, India
| | - A N Resmi
- Division of Biophotonics and Imaging, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences &Technology, Poojappura, Thiruvananthapuram-695012, Kerala, India
| | - Sachin J Shenoy
- Division of In Vivo Models and Testing, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences &Technology, Poojappura, Thiruvananthapuram-695012, Kerala, India
| | - Ramapurath S Jayasree
- Division of Biophotonics and Imaging, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences &Technology, Poojappura, Thiruvananthapuram-695012, Kerala, India
| |
Collapse
|
7
|
Baldea I, Petran A, Florea A, Sevastre-Berghian A, Nenu I, Filip GA, Cenariu M, Radu MT, Iacovita C. Magnetic Nanoclusters Stabilized with Poly[3,4-Dihydroxybenzhydrazide] as Efficient Therapeutic Agents for Cancer Cells Destruction. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:933. [PMID: 36903811 PMCID: PMC10005337 DOI: 10.3390/nano13050933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 06/18/2023]
Abstract
Magnetic structures exhibiting large magnetic moments are sought after in theranostic approaches that combine magnetic hyperthermia treatment (MH) and diagnostic magnetic resonance imaging in oncology, since they offer an enhanced magnetic response to an external magnetic field. We report on the synthesized production of a core-shell magnetic structure using two types of magnetite nanoclusters (MNC) based on a magnetite core and polymer shell. This was achieved through an in situ solvothermal process, using, for the first time, 3,4-dihydroxybenzhydrazide (DHBH) and poly[3,4-dihydroxybenzhydrazide] (PDHBH) as stabilizers. Transmission electron microscopy (TEM) analysis showed the formation of spherical MNC, X-ray photoelectronic spectroscopy (XPS) and Fourier transformed infrared (FT-IR) analysis proved the existence of the polymer shell. Magnetization measurement showed saturation magnetization values of 50 emu/g for PDHBH@MNC and 60 emu/g for DHBH@MNC with very low coercive field and remanence, indicating that the MNC are in a superparamagnetic state at room temperature and are thus suitable for biomedical applications. MNCs were investigated in vitro, on human normal (dermal fibroblasts-BJ) and tumor (colon adenocarcinoma-CACO2, and melanoma-A375) cell lines, in view of toxicity, antitumor effectiveness and selectivity upon magnetic hyperthermia. MNCs exhibited good biocompatibility and were internalized by all cell lines (TEM), with minimal ultrastructural changes. By means of flowcytometry apoptosis detection, fluorimetry, spectrophotometry for mitochondrial membrane potential, oxidative stress, ELISA-caspases, and Western blot-p53 pathway, we show that MH efficiently induced apoptosis mostly via the membrane pathway and to a lower extent by the mitochondrial pathway, the latter mainly observed in melanoma. Contrarily, the apoptosis rate was above the toxicity limit in fibroblasts. Due to its coating, PDHBH@MNC showed selective antitumor efficacy and can be further used in theranostics since the PDHBH polymer provides multiple reaction sites for the attachment of therapeutic molecules.
Collapse
Affiliation(s)
- Ioana Baldea
- Department of Physiology, Iuliu Hatieganu University of Medicine and Pharmacy, Clinicilor 1–3 Str., 400012 Cluj-Napoca, Romania
| | - Anca Petran
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67–103 Donat Str., 400293 Cluj-Napoca, Romania
| | - Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Pasteur 6 Str., 400349 Cluj-Napoca, Romania
| | - Alexandra Sevastre-Berghian
- Department of Physiology, Iuliu Hatieganu University of Medicine and Pharmacy, Clinicilor 1–3 Str., 400012 Cluj-Napoca, Romania
| | - Iuliana Nenu
- Department of Physiology, Iuliu Hatieganu University of Medicine and Pharmacy, Clinicilor 1–3 Str., 400012 Cluj-Napoca, Romania
| | - Gabriela Adriana Filip
- Department of Physiology, Iuliu Hatieganu University of Medicine and Pharmacy, Clinicilor 1–3 Str., 400012 Cluj-Napoca, Romania
| | - Mihai Cenariu
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, Calea Manastur 3–5 Str., 400658 Cluj-Napoca, Romania
| | - Maria Teodora Radu
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67–103 Donat Str., 400293 Cluj-Napoca, Romania
| | - Cristian Iacovita
- Department of Pharmaceutical Physics-Biophysics, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Str., 400349 Cluj-Napoca, Romania
| |
Collapse
|
8
|
A Review of Biomimetic and Biodegradable Magnetic Scaffolds for Bone Tissue Engineering and Oncology. Int J Mol Sci 2023; 24:ijms24054312. [PMID: 36901743 PMCID: PMC10001544 DOI: 10.3390/ijms24054312] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Bone defects characterized by limited regenerative properties are considered a priority in surgical practice, as they are associated with reduced quality of life and high costs. In bone tissue engineering, different types of scaffolds are used. These implants represent structures with well-established properties that play an important role as delivery vectors or cellular systems for cells, growth factors, bioactive molecules, chemical compounds, and drugs. The scaffold must provide a microenvironment with increased regenerative potential at the damage site. Magnetic nanoparticles are linked to an intrinsic magnetic field, and when they are incorporated into biomimetic scaffold structures, they can sustain osteoconduction, osteoinduction, and angiogenesis. Some studies have shown that combining ferromagnetic or superparamagnetic nanoparticles and external stimuli such as an electromagnetic field or laser light can enhance osteogenesis and angiogenesis and even lead to cancer cell death. These therapies are based on in vitro and in vivo studies and could be included in clinical trials for large bone defect regeneration and cancer treatments in the near future. We highlight the scaffolds' main attributes and focus on natural and synthetic polymeric biomaterials combined with magnetic nanoparticles and their production methods. Then, we underline the structural and morphological aspects of the magnetic scaffolds and their mechanical, thermal, and magnetic properties. Great attention is devoted to the magnetic field effects on bone cells, biocompatibility, and osteogenic impact of the polymeric scaffolds reinforced with magnetic nanoparticles. We explain the biological processes activated due to magnetic particles' presence and underline their possible toxic effects. We present some studies regarding animal tests and potential clinical applications of magnetic polymeric scaffolds.
Collapse
|
9
|
Extracellular magnetic labeling of biomimetic hydrogel-induced human mesenchymal stem cell spheroids with ferumoxytol for MRI tracking. Bioact Mater 2023; 19:418-428. [PMID: 35574059 PMCID: PMC9079175 DOI: 10.1016/j.bioactmat.2022.04.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 02/07/2023] Open
Abstract
Labeling of mesenchymal stem cells (MSCs) with superparamagnetic iron oxide nanoparticles (SPIONs) has emerged as a potential method for magnetic resonance imaging (MRI) tracking of transplanted cells in tissue repair studies and clinical trials. Labeling of MSCs using clinically approved SPIONs (ferumoxytol) requires the use of transfection reagents or magnetic field, which largely limits their clinical application. To overcome this obstacle, we established a novel and highly effective method for magnetic labeling of MSC spheroids using ferumoxytol. Unlike conventional methods, ferumoxytol labeling was done in the formation of a mechanically tunable biomimetic hydrogel-induced MSC spheroids. Moreover, the labeled MSC spheroids exhibited strong MRI T2 signals and good biosafety. Strikingly, the encapsulated ferumoxytol was localized in the extracellular matrix (ECM) of the spheroids instead of the cytoplasm, minimizing the cytotoxicity of ferumoxytol and maintaining the viability and stemness properties of biomimetic hydrogel-induced MSC spheroids. This demonstrates the potential of this method for post-transplantation MRI tracking in the clinic. An extracellular magnetic labeling method was developed for MSC spheroids using ferumoxytol. Ferumoxytol encapsulated into abundant ECM proteins network of MSC spheroids ensured this method is stable and durable. Uniformly sized magnetic spheroids induced by mechanically tunable biomimetic hydrogels promoted MSCs stemness properties. Magnetically labeled MSC spheroids exhibited superior MRI imaging both in vitro and in vivo.
Collapse
|
10
|
Abu-El-Rub E, Khasawneh RR, Almahasneh F. Prodigious therapeutic effects of combining mesenchymal stem cells with magnetic nanoparticles. World J Stem Cells 2022; 14:513-526. [PMID: 36157526 PMCID: PMC9350622 DOI: 10.4252/wjsc.v14.i7.513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/18/2022] [Accepted: 06/22/2022] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have gained wide-ranging reputation in the medical research community due to their promising regenerative abilities. MSCs can be isolated from various resources mostly bone marrow, Adipose tissues and Umbilical cord. Huge advances have been achieved in comprehending the possible mechanisms underlying the therapeutic functions of MSCs. Despite the proven role of MSCs in repairing and healing of many disease modalities, many hurdles hinder the transferring of these cells in the clinical settings. Among the most reported problems encountering MSCs therapy in vivo are loss of tracking signal post-transplantation, insufficient migration, homing and engraftment post-infusion, and undesirable differentiation at the site of injury. Magnetic nano particles (MNPs) have been used widely for various biomedical applications. MNPs have a metallic core stabilized by an outer coating material and their ma gnetic properties can be modulated by an external magnetic field. These magnetic properties of MNPs were found to enhance the quality of diagnostic imaging procedures and can be used to create a carrying system for targeted delivery of therapeutic substances mainly drug, genes and stem cells. Several studies highlighted the advantageous outcomes of combining MSCs with MNPs in potentiating their tracking, monitoring, homing, engraftment and differentiation. In this review, we will discuss the role of MNPs in promoting the therapeutic profile of MSCs which may improve the success rate of MSCs transplantation and solve many challenges that delay their clinical applicability.
Collapse
Affiliation(s)
- Ejlal Abu-El-Rub
- Department of Physiology and Pathophysiology, Yarmouk University, Irbid 21163, Jordan
| | - Ramada R Khasawneh
- Department of Anatomy and Histology, Yarmouk University, Irbid 21163, Jordan.
| | - Fatimah Almahasneh
- Department of Physiology and Pathophysiology, Yarmouk University, Irbid 21163, Jordan
| |
Collapse
|
11
|
Cell-based drug delivery systems and their in vivo fate. Adv Drug Deliv Rev 2022; 187:114394. [PMID: 35718252 DOI: 10.1016/j.addr.2022.114394] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022]
Abstract
Cell-based drug delivery systems (DDSs) have received attention recently because of their unique biological properties and self-powered functions, such as excellent biocompatibility, low immunogenicity, long circulation time, tissue-homingcharacteristics, and ability to cross biological barriers. A variety of cells, including erythrocytes, stem cells, and lymphocytes, have been explored as functional vectors for the loading and delivery of various therapeutic payloads (e.g., small-molecule and nucleic acid drugs) for subsequent disease treatment. These cell-based DDSs have their own unique in vivo fates, which are attributed to various factors, including their biological properties and functions, the loaded drugs and loading process, physiological and pathological circumstances, and the body's response to these carrier cells, which result in differences in drug delivery efficiency and therapeutic effect. In this review, we summarize the main cell-based DDSs and their biological properties and functions, applications in drug delivery and disease treatment, and in vivo fate and influencing factors. We envision that the unique biological properties, combined with continuing research, will enable development of cell-based DDSs as friendly drug vectors for the safe, effective, and even personalized treatment of diseases.
Collapse
|
12
|
Chen Y, Hou S. Application of magnetic nanoparticles in cell therapy. Stem Cell Res Ther 2022; 13:135. [PMID: 35365206 PMCID: PMC8972776 DOI: 10.1186/s13287-022-02808-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
Fe3O4 magnetic nanoparticles (MNPs) are biomedical materials that have been approved by the FDA. To date, MNPs have been developed rapidly in nanomedicine and are of great significance. Stem cells and secretory vesicles can be used for tissue regeneration and repair. In cell therapy, MNPs which interact with external magnetic field are introduced to achieve the purpose of cell directional enrichment, while MRI to monitor cell distribution and drug delivery. This paper reviews the size optimization, response in external magnetic field and biomedical application of MNPs in cell therapy and provides a comprehensive view.
Collapse
Affiliation(s)
- Yuling Chen
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China. .,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China.
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China.,Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| |
Collapse
|
13
|
Dasari A, Xue J, Deb S. Magnetic Nanoparticles in Bone Tissue Engineering. NANOMATERIALS 2022; 12:nano12050757. [PMID: 35269245 PMCID: PMC8911835 DOI: 10.3390/nano12050757] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 12/25/2022]
Abstract
Large bone defects with limited intrinsic regenerative potential represent a major surgical challenge and are associated with a high socio-economic burden and severe reduction in the quality of life. Tissue engineering approaches offer the possibility to induce new functional bone regeneration, with the biomimetic scaffold serving as a bridge to create a microenvironment that enables a regenerative niche at the site of damage. Magnetic nanoparticles have emerged as a potential tool in bone tissue engineering that leverages the inherent magnetism of magnetic nano particles in cellular microenvironments providing direction in enhancing the osteoinductive, osteoconductive and angiogenic properties in the design of scaffolds. There are conflicting opinions and reports on the role of MNPs on these scaffolds, such as the true role of magnetism, the application of external magnetic fields in combination with MNPs, remote delivery of biomechanical stimuli in-vivo and magnetically controlled cell retention or bioactive agent delivery in promoting osteogenesis and angiogenesis. In this review, we focus on the role of magnetic nanoparticles for bone-tissue-engineering applications in both disease modelling and treatment of injuries and disease. We highlight the materials-design pathway from implementation strategy through the selection of materials and fabrication methods to evaluation. We discuss the advances in this field and unmet needs, current challenges in the development of ideal materials for bone-tissue regeneration and emerging strategies in the field.
Collapse
Affiliation(s)
- Akshith Dasari
- Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Floor 17 Tower Wing, Guy’s Hospital, London Bridge, London SE19RT, UK; (A.D.); (J.X.)
- Faculty of Life Sciences & Medicine, King’s College London, Guy’s Campus, London SE11UL, UK
| | - Jingyi Xue
- Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Floor 17 Tower Wing, Guy’s Hospital, London Bridge, London SE19RT, UK; (A.D.); (J.X.)
| | - Sanjukta Deb
- Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Floor 17 Tower Wing, Guy’s Hospital, London Bridge, London SE19RT, UK; (A.D.); (J.X.)
- Correspondence:
| |
Collapse
|
14
|
Smart Magnetic Nanocarriers for Multi-Stimuli On-Demand Drug Delivery. NANOMATERIALS 2022; 12:nano12030303. [PMID: 35159647 PMCID: PMC8840331 DOI: 10.3390/nano12030303] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/10/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
In this study, we report the realization of drug-loaded smart magnetic nanocarriers constituted by superparamagnetic iron oxide nanoparticles encapsulated in a dual pH- and temperature-responsive poly (N-vinylcaprolactam-co-acrylic acid) copolymer to achieve highly controlled drug release and localized magnetic hyperthermia. The magnetic core was constituted by flower-like magnetite nanoparticles with a size of 16.4 nm prepared by the polyol approach, with good saturation magnetization and a high specific absorption rate. The core was encapsulated in poly (N-vinylcaprolactam-co-acrylic acid) obtaining magnetic nanocarriers that revealed reversible hydration/dehydration transition at the acidic condition and/or at temperatures above physiological body temperature, which can be triggered by magnetic hyperthermia. The efficacy of the system was proved by loading doxorubicin with very high encapsulation efficiency (>96.0%) at neutral pH. The double pH- and temperature-responsive nature of the magnetic nanocarriers facilitated a burst, almost complete release of the drug at acidic pH under hyperthermia conditions, while a negligible amount of doxorubicin was released at physiological body temperature at neutral pH, confirming that in addition to pH variation, drug release can be improved by hyperthermia treatment. These results suggest this multi-stimuli-sensitive nanoplatform is a promising candidate for remote-controlled drug release in combination with magnetic hyperthermia for cancer treatment.
Collapse
|
15
|
Zhang W, Gaikwad H, Groman EV, Purev E, Simberg D, Wang G. Highly aminated iron oxide nanoworms for simultaneous manufacturing and labeling of chimeric antigen receptor T cells. JOURNAL OF MAGNETISM AND MAGNETIC MATERIALS 2022; 541:168480. [PMID: 34720339 PMCID: PMC8553019 DOI: 10.1016/j.jmmm.2021.168480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Cell based therapies including chimeric antigen receptor (CAR) T cells are promising for treating leukemias and solid cancers. At the same time, there is interest in enhancing the functionality of these cells via surface decoration with nanoparticles (backpacking). Magnetic nanoparticle cell labeling is of particular interest due to opportunities for magnetic separation, in vivo manipulation, drug delivery and magnetic resonance imaging (MRI). While modification of T cells with magnetic nanoparticles (MNPs) was explored before, we questioned whether MNPs are compatible with CAR-T cells when introduced during the manufacturing process. We chose highly aminated 120 nm crosslinked iron oxide nanoworms (CLIO NWs, ~36,000 amines per NW) that could efficiently label different adherent cell lines and we used CD123 CAR-T cells as the labeling model. The CD123 CAR-T cells were produced in the presence of CLIO NWs, CLIO NWs plus protamine sulfate (PS), or PS only. The transduction efficiency of lentiviral CD123 CAR with only NWs was ~23% lower than NW+PS and PS groups (~33% and 35%, respectively). The cell viability from these three transduction conditions was not reduced within CAR-T cell groups, though lower compared to non-transduced T cells (mock T). Use of CLIO NWs instead of, or together with cationic protamine sulfate for enhancement of lentiviral transduction resulted in comparable levels of CAR expression and viability but decreased the proportion of CD8+ cells and increased the proportion of CD4+ cells. CD123 CAR-T transduced in the presence of CLIO NWs, CLIO NWs plus PS, or PS only, showed similar level of cytotoxicity against leukemic cell lines. Furthermore, fluorescence microscopy imaging demonstrated that CD123 CAR-T cells labeled with CLIO NW formed rosettes with CD123+ leukemic cells as the non-labeled CAR-T cells, indicating that the CAR-T targeting to tumor cells has maintained after CLIO NW labeling. The in vivo trafficking of the NW labeled CAR-T cells showed the accumulation of CAR-T labeled with NWs primarily in the bone marrow and spleen. CAR-T cells can be magnetically labeled during their production while maintaining functionality using the positively charged iron oxide NWs, which enable the in vivo biodistribution and tracking of CAR-T cells.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Hanmant Gaikwad
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ernest V. Groman
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Enkhtsetseg Purev
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Corresponding Authors: (Dmitri Simberg), (Guankui Wang)
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Corresponding Authors: (Dmitri Simberg), (Guankui Wang)
| |
Collapse
|
16
|
Nano-engineered immune cells as "guided missiles" for cancer therapy. J Control Release 2021; 341:60-79. [PMID: 34785315 DOI: 10.1016/j.jconrel.2021.11.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
Immune cells can actively regulate tumors or inflammatory sites and have good biocompatibility and safety. Currently, they are one of the most promising candidates for drug delivery systems. Moreover, immune cells can significantly extend the circulation time of nanoparticles and have broad-spectrum tumor-targeting properties. This article first introduces the immune cell types most commonly used in recent years, analyzes their advantages and disadvantages, and elucidates their application in anti-tumor therapy. Next, the various ways of loading nanoparticles on immune cells that have been used in recent years are summarized and simply divided into two categories: backpacks and Trojan horses. Finally, the two "mountains" that stand in front of us when using immune cells as cell carriers, off-target problems and effective release strategies, are discussed.
Collapse
|
17
|
Wei H, Hu Y, Wang J, Gao X, Qian X, Tang M. Superparamagnetic Iron Oxide Nanoparticles: Cytotoxicity, Metabolism, and Cellular Behavior in Biomedicine Applications. Int J Nanomedicine 2021; 16:6097-6113. [PMID: 34511908 PMCID: PMC8418330 DOI: 10.2147/ijn.s321984] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been widely investigated and applied in the field of biomedicine due to their excellent superparamagnetic properties and reliable traceability. However, with the optimization of core composition, shell types and transfection agents, the cytotoxicity and metabolism of different SPIONs have great differences, and the labeled cells also show different cellular behaviors. Therefore, a holistic review of the construction and application of SPIONs is desired. This review focuses the advances of SPIONs in the field of biomedicine in recent years. After summarizing the toxicity of different SPIONs, the uptake, distribution and metabolism of SPIONs in vitro were discussed. Then, the regulation of labeled-cells behavior is outlined. Furthermore, the major challenges in the optimization process of SPIONs and insights on its future developments are proposed.
Collapse
Affiliation(s)
- Hao Wei
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline, Nanjing, 210008, People's Republic of China
| | - Yangnan Hu
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, People's Republic of China
| | - Junguo Wang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline, Nanjing, 210008, People's Republic of China
| | - Xia Gao
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline, Nanjing, 210008, People's Republic of China
| | - Xiaoyun Qian
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline, Nanjing, 210008, People's Republic of China
| | - Mingliang Tang
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, People's Republic of China.,Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, 215000, People's Republic of China
| |
Collapse
|
18
|
Huang H, Du X, He Z, Yan Z, Han W. Nanoparticles for Stem Cell Tracking and the Potential Treatment of Cardiovascular Diseases. Front Cell Dev Biol 2021; 9:662406. [PMID: 34277609 PMCID: PMC8283769 DOI: 10.3389/fcell.2021.662406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/12/2021] [Indexed: 01/15/2023] Open
Abstract
Stem cell-based therapies have been shown potential in regenerative medicine. In these cells, mesenchymal stem cells (MSCs) have the ability of self-renewal and being differentiated into different types of cells, such as cardiovascular cells. Moreover, MSCs have low immunogenicity and immunomodulatory properties, and can protect the myocardium, which are ideal qualities for cardiovascular repair. Transplanting mesenchymal stem cells has demonstrated improved outcomes for treating cardiovascular diseases in preclinical trials. However, there still are some challenges, such as their low rate of migration to the ischemic myocardium, low tissue retention, and low survival rate after the transplantation. To solve these problems, an ideal method should be developed to precisely and quantitatively monitor the viability of the transplanted cells in vivo for providing the guidance of clinical translation. Cell imaging is an ideal method, but requires a suitable contrast agent to label and track the cells. This article reviews the uses of nanoparticles as contrast agents for tracking MSCs and the challenges of clinical use of MSCs in the potential treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Huihua Huang
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Health Science Center, Shenzhen, China
| | - Xuejun Du
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| | - Zhiguo He
- Advanced Materials Institute, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Zifeng Yan
- Advanced Materials Institute, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Wei Han
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
19
|
Cui M, Zhang HR, Ouyang F, Guo YQ, Li RF, Duan SF, Xiong TD, Wang YL, Wang XQ. Dual Enzyme-Like Performances of PLGA Grafted Maghemite Nanocrystals and Their Synergistic Chemo/Chemodynamic Treatment for Human Lung Adenocarcinoma A549 Cells. J Biomed Nanotechnol 2021; 17:1007-1019. [PMID: 34167616 DOI: 10.1166/jbn.2021.3062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In recent years, the emergence of non-toxic but catalytically active inorganic nanoparticles has attracted great attention for cancer treatment, but the therapeutic effect has been affected by the limited reactive oxygen species in tumors. Therefore, the combination of chemotherapy and chemodynamic therapy is regarded as a promising therapeutic strategy. In this paper, we reported the preparation and bioactivity evaluation of poly(lactic acid-co-glycolic acid) (PLGA) grafted-γ-Fe₂O₃ nanoparticles with dual response of endogenous peroxidase and catalase like activities. Our hypothesis is that PLGAgrafted γ-Fe₂O₃ nanoparticles could be used as a drug delivery system for the anti-tumor drug doxorubicin to inhibit the growth of lung adenocarcinoma A549 cells; meanwhile, based on its mimic enzyme properties, this kind of nanoparticles could be combined with doxorubicin in the treatment of A549 cells. Our experimental results showed that the PLGAgrafted γ-Fe₂O₃ nanoparticles could simulate the activity of catalase and decompose hydrogen peroxide into H₂O and oxygen in neutral tumor microenvironment, thus reducing the oxidative damage caused by hydrogenperoxide to lung adenocarcinoma A549 cells. In acidic microenvironment, PLGA grafted γ-Fe₂O₃ nanoparticles could simulate the activity of peroxidase and effectively catalyze the decomposition of hydrogen peroxide to generate highly toxic hydroxyl radicals, which could cause the death of A549 cells. Furthermore, the synergistic effect of peroxidase-like activity of PLGA-grafted γ-Fe₂O₃ nanoparticles and doxorubicin could accelerate the apoptosisand destruction of A549 cells, thus enhancing the antitumor effect of doxorubicin-loaded PLGA-grafted γ-Fe₂O₃ nanoparticles. Therefore, this study provides an effective nanoplatform based on dual inorganic biomimetic nanozymes for the treatment of lung cancer.
Collapse
Affiliation(s)
- Miao Cui
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, P. R. China
| | - Hui-Ru Zhang
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, P. R. China
| | - Fan Ouyang
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, P. R. China
| | - Yu-Qi Guo
- Henan Provincial People's Hospital, Zhengzhou 450003, P. R. China
| | - Rui-Fang Li
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, P. R. China
| | - Shao-Feng Duan
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Tian-Di Xiong
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, P. R. China
| | - Yun-Long Wang
- Henan Bioengineering Research Center, Zhengzhou 450046, P. R. China
| | - Xue-Qin Wang
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, P. R. China
| |
Collapse
|
20
|
Radeloff K, Ramos Tirado M, Haddad D, Breuer K, Müller J, Hochmuth S, Hackenberg S, Scherzad A, Kleinsasser N, Radeloff A. Superparamagnetic Iron Oxide Particles (VSOPs) Show Genotoxic Effects but No Functional Impact on Human Adipose Tissue-Derived Stromal Cells (ASCs). MATERIALS 2021; 14:ma14020263. [PMID: 33430323 PMCID: PMC7825809 DOI: 10.3390/ma14020263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
Abstract
Adipose tissue-derived stromal cells (ASCs) represent a capable source for cell-based therapeutic approaches. For monitoring a cell-based application in vivo, magnetic resonance imaging (MRI) of cells labeled with iron oxide particles is a common method. It is the aim of the present study to analyze potential DNA damage, cytotoxicity and impairment of functional properties of human (h)ASCs after labeling with citrate-coated very small superparamagnetic iron oxide particles (VSOPs). Cytotoxic as well as genotoxic effects of the labeling procedure were measured in labeled and unlabeled hASCs using the MTT assay, comet assay and chromosomal aberration test. Trilineage differentiation was performed to evaluate an impairment of the differentiation potential due to the particles. Proliferation as well as migration capability were analyzed after the labeling procedure. Furthermore, the labeling of the hASCs was confirmed by Prussian blue staining, transmission electron microscopy (TEM) and high-resolution MRI. Below the concentration of 0.6 mM, which was used for the procedure, no evidence of genotoxic effects was found. At 0.6 mM, 1 mM as well as 1.5 mM, an increase in the number of chromosomal aberrations was determined. Cytotoxic effects were not observed at any concentration. Proliferation, migration capability and differentiation potential were also not affected by the procedure. Labeling with VSOPs is a useful labeling method for hASCs that does not affect their proliferation, migration and differentiation potential. Despite the absence of cytotoxicity, however, indications of genotoxic effects have been demonstrated.
Collapse
Affiliation(s)
- Katrin Radeloff
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Oldenburg, 26122 Oldenburg, Germany; (J.M.); (S.H.); (A.R.)
- Correspondence:
| | - Mario Ramos Tirado
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (M.R.T.); (S.H.); (A.S.); (N.K.)
| | - Daniel Haddad
- Fraunhofer Development Center X-ray Technology EZRT, Department Magnetic Resonance and X-ray Imaging, A Division of Fraunhofer Institute for Integrated Circuits IIS, 97074 Wuerzburg, Germany;
| | - Kathrin Breuer
- Department of Radiation Oncology, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Jana Müller
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Oldenburg, 26122 Oldenburg, Germany; (J.M.); (S.H.); (A.R.)
| | - Sabine Hochmuth
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Oldenburg, 26122 Oldenburg, Germany; (J.M.); (S.H.); (A.R.)
| | - Stephan Hackenberg
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (M.R.T.); (S.H.); (A.S.); (N.K.)
| | - Agmal Scherzad
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (M.R.T.); (S.H.); (A.S.); (N.K.)
| | - Norbert Kleinsasser
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, 97080 Wuerzburg, Germany; (M.R.T.); (S.H.); (A.S.); (N.K.)
| | - Andreas Radeloff
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Oldenburg, 26122 Oldenburg, Germany; (J.M.); (S.H.); (A.R.)
| |
Collapse
|
21
|
Xu H, Qiu Y, Xiong Z, Shao W, Zhang Q, Tang G. Tracking mesenchymal stem cells with Ir(III) complex-encapsulated nanospheres in cranium defect with postmenopausal osteoporosis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111842. [PMID: 33641885 DOI: 10.1016/j.msec.2020.111842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 01/15/2023]
Abstract
Osteoporosis (OP) is a significant public health problem with associated fragility fractures, thereby causing large bone defects and difficulty in self-repair. The introduction of human mesenchymal stem cells (hMSCs) is the most promising platform in bone tissue engineering for OP therapy, which induces less side effects than conventional medication. However, the safety and efficiency of the cell-based OP therapy requires the ability to monitor the cell's outcome and biodistribution after cell transplantation. Therefore, we designed an in vivo system to track hMSCs in real time and simultaneously attempted to obtain a significant therapeutic effect during the bone repair process. In this study, we synthesized Ir(III) complex, followed by encapsulation with biodegradable methoxy-poly(ethylene glycol) poly(lactic-co-glycolic acid) nanospheres through double emulsions strategy. The Ir(III) complex nanospheres did not affect hMSC proliferation, stemness, and differentiation and realized highly efficient and long-term cellular labeling for at least 25 days in vivo. The optimal transplantation conditions were also determined first by injecting a gradient number of labeled hMSCs percutaneously into the cranial defect of the nude mouse model. Next, we applied this method to ovariectomy-induced OP mice. Results showed long-term optical imaging with high fluorescence intensity and computed tomography (CT) scanning with significantly increased bone formation between the osteoporotic and sham-operated bones. During the tracking process, two mice from each group were sacrificed at two representative time points to examine the bony defect bridging via micro-CT morphometric analyses. Our data showed remarkable promise for efficient hMSC tracking and encouraging treatment in bioimaging-guided OP stem cell therapy.
Collapse
Affiliation(s)
- Hong Xu
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yanchang Road, Shanghai 200072, P. R. China; Department of Radiology, Northern Jiangsu People's Hospital, Clinical Medical School of Yangzhou University, No. 98 Nantong West Road, Yangzhou, Jiangsu 225001, P. R. China
| | - Yuyou Qiu
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yanchang Road, Shanghai 200072, P. R. China
| | - Zuogang Xiong
- Department of Radiology, Ping An Healthcare Diagnostics Center, No. 199 Kaibin Road, Shanghai 200030, P. R. China
| | - Wenjun Shao
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Qi Zhang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China.
| | - Guangyu Tang
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yanchang Road, Shanghai 200072, P. R. China.
| |
Collapse
|
22
|
Papan P, Kantapan J, Sangthong P, Meepowpan P, Dechsupa N. Iron (III)-Quercetin Complex: Synthesis, Physicochemical Characterization, and MRI Cell Tracking toward Potential Applications in Regenerative Medicine. CONTRAST MEDIA & MOLECULAR IMAGING 2020; 2020:8877862. [PMID: 33456403 PMCID: PMC7785384 DOI: 10.1155/2020/8877862] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 12/20/2022]
Abstract
In cell therapy, contrast agents T1 and T2 are both needed for the labeling and tracking of transplanted stem cells over extended periods of time through magnetic resonance imaging (MRI). Importantly, the metal-quercetin complex via coordination chemistry has been studied extensively for biomedical applications, such as anticancer therapies and imaging probes. Herein, we report on the synthesis, characterization, and labeling of the iron (III)-quercetin complex, "IronQ," in circulating proangiogenic cells (CACs) and also explore tracking via the use of a clinical 1.5 Tesla (T) MRI scanner. Moreover, IronQ had a paramagnetic T1 positive contrast agent property with a saturation magnetization of 0.155 emu/g at 1.0 T and longitudinal relaxivity (r1) values of 2.29 and 3.70 mM-1s-1 at 1.5 T for water and human plasma, respectively. Surprisingly, IronQ was able to promote CAC growth in conventional cell culture systems without the addition of specific growth factors. Increasing dosages of IronQ from 0 to 200 μg/mL led to higher CAC uptake, and maximum labeling time was achieved in 10 days. The accumulated IronQ in CACs was measured by two methodologies, an inductively coupled plasma optical emission spectrometry (ICP-EOS) and T1-weighted MRI. In our research, we confirmed that IronQ has excellent dual functions with the use of an imaging probe for MRI. IronQ can also act as a stimulating agent by favoring circulating proangiogenic cell differentiation. Optimistically, IronQ is considered beneficial for alternative labeling and in the tracking of circulation proangiogenic cells and/or other stem cells in applications of cell therapy through noninvasive magnetic resonance imaging in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Phakorn Papan
- Research Unit of Molecular Imaging Probes and Radiobiology, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jiraporn Kantapan
- Research Unit of Molecular Imaging Probes and Radiobiology, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Padchanee Sangthong
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Puttinan Meepowpan
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nathupakorn Dechsupa
- Research Unit of Molecular Imaging Probes and Radiobiology, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
23
|
Campos F, Bonhome-Espinosa AB, Carmona R, Durán JDG, Kuzhir P, Alaminos M, López-López MT, Rodriguez IA, Carriel V. In vivo time-course biocompatibility assessment of biomagnetic nanoparticles-based biomaterials for tissue engineering applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111476. [PMID: 33255055 DOI: 10.1016/j.msec.2020.111476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 01/17/2023]
Abstract
Novel artificial tissues with potential usefulness in local-based therapies have been generated by tissue engineering using magnetic-responsive nanoparticles (MNPs). In this study, we performed a comprehensive in vivo characterization of bioengineered magnetic fibrin-agarose tissue-like biomaterials. First, in vitro analyses were performed and the cytocompatibility of MNPs was demonstrated. Then, bioartificial tissues were generated and subcutaneously implanted in Wistar rats and their biodistribution, biocompatibility and functionality were analysed at the morphological, histological, haematological and biochemical levels as compared to injected MNPs. Magnetic Resonance Image (MRI), histology and magnetometry confirmed the presence of MNPs restricted to the grafting area after 12 weeks. Histologically, we found a local initial inflammatory response that decreased with time. Structural, ultrastructural, haematological and biochemical analyses of vital organs showed absence of damage or failure. This study demonstrated that the novel magnetic tissue-like biomaterials with improved biomechanical properties fulfil the biosafety and biocompatibility requirements for future clinical use and support the use of these biomaterials as an alternative delivery route for magnetic nanoparticles.
Collapse
Affiliation(s)
- Fernando Campos
- Department of Histology, Tissue Engineering Group, Faculty of Medicine, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Ana B Bonhome-Espinosa
- Department of Applied Physics, University of Granada, Avenida de la Fuente Nueva, 18071 Granada, Spain
| | - Ramón Carmona
- Department of Cell Biology, Faculty of Sciences, University of Granada, Campus Fuentenueva s/n, Granada, Spain
| | - Juan D G Durán
- Department of Applied Physics, University of Granada, Avenida de la Fuente Nueva, 18071 Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Pavel Kuzhir
- Université Côte d'Azur, CNRS UMR 7010, Institute of Physics of Nice, Parc Valrose, 06108 Nice, France
| | - Miguel Alaminos
- Department of Histology, Tissue Engineering Group, Faculty of Medicine, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Modesto T López-López
- Department of Applied Physics, University of Granada, Avenida de la Fuente Nueva, 18071 Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.
| | - Ismael A Rodriguez
- Department of Histology, Tissue Engineering Group, Faculty of Medicine, University of Granada, Granada, Spain; Department of Histology, Faculty of Dentistry, Nacional University of Cordoba, Cordoba, Argentina.
| | - Víctor Carriel
- Department of Histology, Tissue Engineering Group, Faculty of Medicine, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| |
Collapse
|
24
|
Vakili-Ghartavol R, Momtazi-Borojeni AA, Vakili-Ghartavol Z, Aiyelabegan HT, Jaafari MR, Rezayat SM, Arbabi Bidgoli S. Toxicity assessment of superparamagnetic iron oxide nanoparticles in different tissues. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:443-451. [PMID: 32024389 DOI: 10.1080/21691401.2019.1709855] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been employed in several biomedical applications where they facilitate both diagnostic and therapeutic aims. Although the potential benefits of SPIONs with different surface chemistry and conjugated targeting ligands/proteins are considerable, complicated interactions between these nanoparticles (NPs) and cells leading to toxic impacts could limit their clinical applications. Hence, elevation of our knowledge regarding the SPION-related toxicity is necessary. Here, the present review article will consider current studies and compare the potential toxic effect of SPIONs with or without identical surface chemistries on different cell lines. It centers on cellular and molecular mechanisms underlying toxicity of SPIONs. Likewise, emphasis is being dedicated for toxicity of SPIONs in various cell lines, in vitro and animal models, in vivo.
Collapse
Affiliation(s)
- Roghayyeh Vakili-Ghartavol
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Abbas Momtazi-Borojeni
- Halal Research Center of IRI, FDA, Tehran, Iran.,Nanotechnology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeynab Vakili-Ghartavol
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hammed Tanimowo Aiyelabegan
- Department of Medical Biochemistry and Pharmacology, College of Pure and Applied Sciences, Kwara State University Malete, Nigeria
| | - Mahmoud Reza Jaafari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology-Pharmacology, Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Sepideh Arbabi Bidgoli
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University (IAUPS), Tehran, Iran
| |
Collapse
|
25
|
Silva LHA, Silva MC, Vieira JB, Lima ECD, Silva RC, Weiss DJ, Morales MM, Cruz FF, Rocco PRM. Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis. Stem Cells Transl Med 2020; 9:1244-1256. [PMID: 32538526 PMCID: PMC7519769 DOI: 10.1002/sctm.20-0004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/02/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Silicosis is a pneumoconiosis caused by inhaled crystalline silica microparticles, which trigger inflammatory responses and granuloma formation in pulmonary parenchyma, thus affecting lung function. Although systemic administration of mesenchymal stromal cells (MSCs) ameliorates lung inflammation and attenuates fibrosis in experimental silicosis, it does not reverse collagen deposition and granuloma formation. In an attempt to improve the beneficial effects of MSCs, magnetic targeting (MT) has arisen as a potential means of prolonging MSC retention in the lungs. In this study, MSCs were incubated with magnetic nanoparticles and magnets were used for in vitro guidance of these magnetized MSCs and to enhance their retention in the lungs in vivo. In vitro assays indicated that MT improved MSC transmigration and expression of chemokine receptors. In vivo, animals implanted with magnets for 48 hours had significantly more magnetized MSCs in the lungs, suggesting improved MSC retention. Seven days after magnet removal, silicotic animals treated with magnetized MSCs and magnets showed significant reductions in static lung elastance, resistive pressure, and granuloma area. In conclusion, MT is a viable technique to prolong MSC retention in the lungs, enhancing their beneficial effects on experimentally induced silicosis. MT may be a promising strategy for enhancing MSC therapies for chronic lung diseases.
Collapse
Affiliation(s)
- Luisa H A Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Rio de Janeiro, Brazil.,Rio de Janeiro Innovation Network in Nanosystems for Health - NanoSAÚDE/FAPERJ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana C Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana B Vieira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emilia C D Lima
- Institute of Chemistry, Federal University of Goias, Goiânia, Goiás, Brazil
| | - Renata C Silva
- National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, Rio de Janeiro, Brazil
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, College of Medicine, Burlington, Vermont, USA
| | - Marcelo M Morales
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Rio de Janeiro, Brazil.,Rio de Janeiro Innovation Network in Nanosystems for Health - NanoSAÚDE/FAPERJ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Rio de Janeiro, Brazil.,Rio de Janeiro Innovation Network in Nanosystems for Health - NanoSAÚDE/FAPERJ, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Assessment of proliferation, migration and differentiation potentials of bone marrow mesenchymal stem cells labeling with silica-coated and amine-modified superparamagnetic iron oxide nanoparticles. Cytotechnology 2020; 72:513-525. [PMID: 32394163 DOI: 10.1007/s10616-020-00397-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/04/2020] [Indexed: 10/24/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles have been widely used for cell labeling in preclinical and clinical studies, to improve labeling efficiency, particle conjugation and surface modifications are developed, but some modified SPIONs exert side-effect on physiological activity of cells, which cannot be served as ideal cell tracker. In this study, amine-modified silica-coated SPIO (SPIO@SiO2-NH2, SPIO@S-N) nanoparticles were used to label bone marrow derived mesenchymal stem cells (BM-MSCs), then the stem cell potentials were evaluated. It was found BM-MSCs could be efficiently labeled by SPIO@S-N nanoparticles. After labeling, the BM-MSCs viability kept well and the migration ability increased, but the osteogenesis and adipogenesis potentials were not impaired. In steroid associated osteonecrosis (SAON) bone defect model, stem cell implantation was performed by injection of SPIO@S-N labeled BM-MSCs into marrow cavity locally, it was found the SPIO positive cells homed to the periphery of defect region in control group, but were recruited to the defect region in poly lactic-coglycolic acid/tricalcium phosphate (PLGA/TCP) scaffold implantation group. In conclusion, SPIO@S-N nanoparticles promoted migration while retained proliferation and differentiation ability of BM-MSCs, implying this kind of nanoparticles could be served not only an ideal tracking marker but also an accelerator for stem cell homing during tissue repair.
Collapse
|
27
|
Fahmy HM, Abd El-Daim TM, Mohamed HAAENE, Mahmoud EAAEQ, Abdallah EAS, Mahmoud Hassan FEZ, Maihop DI, Amin AEAE, Mustafa ABE, Hassan FMA, Mohamed DME, Shams-Eldin EMM. Multifunctional nanoparticles in stem cell therapy for cellular treating of kidney and liver diseases. Tissue Cell 2020; 65:101371. [PMID: 32746989 DOI: 10.1016/j.tice.2020.101371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
The review gives an overview of the mechanisms of internalization and distribution of nanoparticles in stem cells this is achieved via providing analysis of the methods used in exploring the migration routes of stem cells, and their reciprocity. In addition, exploring microenvironment target in the body, and tracking the fate of exogenously transplanted stem cells by using innovative and non-invasive techniques will also be discussed. Such techniques like magnetic resonance imaging (MRI), multimodality tracking, optical imaging, and nuclear medicine imaging, which were designed to follow up stem cell migration. This review will explain the various distinctive strategies to enhance homing of labeled stem cells with nanoparticles into damaged hepatic and renal tissues, this purpose was obtained by inducing a specific gene into stem cells, various chemokines, and applying an external magnetic field. Also, this work illustrates how to improve nanoparticles uptake by using transfection agents or covalently binding an exogenous protein (i.e., Human immunodeficiency virus-Tat protein) or conjugating a receptor-specific monoclonal antibody or make modifications to iron coat. It contains stem cell labeling methods such as extracellular labeling and internalization approaches. Ultimately, our review indicates trails of researchers in nanoparticles utilization in stem cell therapy in both kidney and liver diseases.
Collapse
|
28
|
Yun WS, Aryal S, Ahn YJ, Seo YJ, Key J. Engineered iron oxide nanoparticles to improve regenerative effects of mesenchymal stem cells. Biomed Eng Lett 2020; 10:259-273. [PMID: 32477611 DOI: 10.1007/s13534-020-00153-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/14/2020] [Accepted: 02/26/2020] [Indexed: 12/16/2022] Open
Abstract
Abstract Mesenchymal stem cells (MSCs) based therapies are a major field of regenerative medicine. However, the success of MSC therapy relies on the efficiency of its delivery and retention, differentiation, and secreting paracrine factors at the target sites. Recent studies show that superparamagnetic iron oxide nanoparticles (SPIONs) modulate the regenerative effects of MSCs. After interacting with the cell membrane of MSCs, SPIONs can enter the cells via the endocytic pathway. The physicochemical properties of nanoparticles, including size, surface charge (zeta-potential), and surface ligand, influence their interactions with MSC, such as cellular uptake, cytotoxicity, homing factors, and regenerative related factors (VEGF, TGF-β1). Therefore, in-depth knowledge of the physicochemical properties of SPIONs might be a promising lead in regenerative and anti-inflammation research using SPIONs mediated MSCs. In this review, recent research on SPIONs with MSCs and the various designs of SPIONs are examined and summarized. Graphic abstract A graphical abstract describes important parameters in the design of superparamagnetic iron oxide nanoparticles, affecting mesenchymal stem cells. These physicochemical properties are closely related to the mesenchymal stem cells to achieve improved cellular responses such as homing factors and cell uptake.
Collapse
Affiliation(s)
- Wan Su Yun
- 1Department of Biomedical Engineering, Yonsei University, 1 Yonseidae-gil, Wonju, Gangwon-do South Korea
| | - Susmita Aryal
- 1Department of Biomedical Engineering, Yonsei University, 1 Yonseidae-gil, Wonju, Gangwon-do South Korea
| | - Ye Ji Ahn
- 2Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, South Korea.,3Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Young Joon Seo
- 2Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, South Korea.,3Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Jaehong Key
- 1Department of Biomedical Engineering, Yonsei University, 1 Yonseidae-gil, Wonju, Gangwon-do South Korea
| |
Collapse
|
29
|
Moayeri A, Darvishi M, Amraei M. Homing of Super Paramagnetic Iron Oxide Nanoparticles (SPIONs) Labeled Adipose-Derived Stem Cells by Magnetic Attraction in a Rat Model of Parkinson's Disease. Int J Nanomedicine 2020; 15:1297-1308. [PMID: 32161459 PMCID: PMC7049746 DOI: 10.2147/ijn.s238266] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/07/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Stem cell therapies for neurodegenerative diseases such as Parkinson’s disease (PD) are intended to replace lost dopaminergic neurons. The basis of this treatment is to guide the migration of transplanted cells into the target tissue or injury site. The aim of this study is an evaluation of the homing of superparamagnetic iron oxide nanoparticles (SPIONs) labeled adipose-derived stem cells (ADSC) by an external magnetic field in a rat model of PD. Methods ADSCs were obtained from perinephric regions of male adult rats and cultured in a DMEM medium. ADSC markers were assessed by immunostaining with CD90, CD105, CD49d, and CD45. The SPION was coated using poly-L-lysine hydrobromide and transfection was determined in rat ADSC using the GFP reporter gene. For this in vivo study, rats with PD were divided into five groups: a positive control group, a control group with PD (lesion with 6-HD injection), and three treatment groups: the PD/ADSC group (PD transplant with ADSCs transfected by BrdU), PD/ADSC/SPION group (PD transplant with ADSCs labeled with SPION and transfected by GFP), and the PD/ADSC/SPION/EM group (PD transplant with ADSCs labeled with SPION and transfected by GFP induced with external magnet). Results ADSCs were immunoreactive to fat markers CD90 (90.73±1.7), CD105 (87.4±2.9) and CD49d (79.6±2.6), with negative immunostaining at the hematopoietic stem cell marker (CD45: 1.4±0.4). The efficiency of cells with SPION/PLL was about 96% of ADSC. The highest number of GFP-positive cells was in the ADSC/SPION/EM group (54.5±1.3), which was significantly different from that in ADSC/SPION group (30.83±3 and P<0.01). Conclusion Transfection of ADSC by SPION/PLL is an appropriate protocol for cell therapy. External magnets can be used for the delivery and homing of transplanted stem cells in the target tissue.
Collapse
Affiliation(s)
- Ardeshir Moayeri
- Department of Anatomy, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Marzieh Darvishi
- Department of Anatomy, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Mansour Amraei
- Department of Physiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
30
|
Demin AM, Mekhaev AV, Kandarakov OF, Popenko VI, Leonova OG, Murzakaev AM, Kuznetsov DK, Uimin MA, Minin AS, Shur VY, Belyavsky AV, Krasnov VP. L-Lysine-modified Fe 3O 4 nanoparticles for magnetic cell labeling. Colloids Surf B Biointerfaces 2020; 190:110879. [PMID: 32135495 DOI: 10.1016/j.colsurfb.2020.110879] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 12/31/2022]
Abstract
The efficiency of magnetic labeling with L-Lys-modified Fe3O4 magnetic nanoparticles (MNPs) and the stability of magnetization of rat adipose-derived mesenchymal stem cells, lineage-negative (Lin(-)) hematopoietic progenitor cells from mouse bone marrow and human leukemia K562 cells were studied. For this purpose, covalent modification of MNPs with 3-aminopropylsilane and N-di-Fmoc-L-lysine followed by removal of N-protecting groups was carried out. Since the degree of hydroxylation of the surface of the starting nanoparticles plays a crucial role in the silanization reaction and the possibility of obtaining stable colloidal solutions. In present work we for the first time performed a comparative qualitative and quantitative evaluation of the number of adsorbed water molecules and hydroxyl groups on the surface of chemically and physically obtained Fe3O4 MNPs using comprehensive FTIR spectroscopy and thermogravimetric analysis. The results obtained can be further used for magnetic labeling of cells in experiments in vitro and in vivo.
Collapse
Affiliation(s)
- Alexander M Demin
- Postovsky Institute of Organic Synthesis, Russian Academy of Sciences (Ural Branch), 22 S. Kovalevskoy St., Yekaterinburg, 620990, Russia.
| | - Alexander V Mekhaev
- Postovsky Institute of Organic Synthesis, Russian Academy of Sciences (Ural Branch), 22 S. Kovalevskoy St., Yekaterinburg, 620990, Russia
| | - Oleg F Kandarakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., Moscow 119991, Russia
| | - Vladimir I Popenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., Moscow 119991, Russia
| | - Olga G Leonova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., Moscow 119991, Russia
| | - Aidar M Murzakaev
- Institute of Electrophysics, Russian Academy of Sciences (Ural Branch), 106 Amudsen St., Yekaterinburg, 620016, Russia; Institute of Natural Sciences and Mathematics, Ural Federal University, 51 Lenin Ave., Yekaterinburg 620000, Russia
| | - Dmitry K Kuznetsov
- Institute of Natural Sciences and Mathematics, Ural Federal University, 51 Lenin Ave., Yekaterinburg 620000, Russia
| | - Mikhail A Uimin
- Mikheev Institute of Metal Physics, Russian Academy of Sciences (Ural Branch), 18 S. Kovalevskoy St., Yekaterinburg, 620990, Russia
| | - Artem S Minin
- Mikheev Institute of Metal Physics, Russian Academy of Sciences (Ural Branch), 18 S. Kovalevskoy St., Yekaterinburg, 620990, Russia
| | - Vladimir Ya Shur
- Institute of Natural Sciences and Mathematics, Ural Federal University, 51 Lenin Ave., Yekaterinburg 620000, Russia
| | - Alexander V Belyavsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., Moscow 119991, Russia
| | - Victor P Krasnov
- Postovsky Institute of Organic Synthesis, Russian Academy of Sciences (Ural Branch), 22 S. Kovalevskoy St., Yekaterinburg, 620990, Russia; Institute of Chemical Engineering, Ural Federal University, 19 Mira St., Yekaterinburg, 620002, Russia
| |
Collapse
|
31
|
Shrestha B, DeLuna F, Anastasio MA, Yong Ye J, Brey EM. Photoacoustic Imaging in Tissue Engineering and Regenerative Medicine. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:79-102. [PMID: 31854242 PMCID: PMC7041335 DOI: 10.1089/ten.teb.2019.0296] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/13/2019] [Indexed: 12/16/2022]
Abstract
Several imaging modalities are available for investigation of the morphological, functional, and molecular features of engineered tissues in small animal models. While research in tissue engineering and regenerative medicine (TERM) would benefit from a comprehensive longitudinal analysis of new strategies, researchers have not always applied the most advanced methods. Photoacoustic imaging (PAI) is a rapidly emerging modality that has received significant attention due to its ability to exploit the strong endogenous contrast of optical methods with the high spatial resolution of ultrasound methods. Exogenous contrast agents can also be used in PAI for targeted imaging. Applications of PAI relevant to TERM include stem cell tracking, longitudinal monitoring of scaffolds in vivo, and evaluation of vascularization. In addition, the emerging capabilities of PAI applied to the detection and monitoring of cancer and other inflammatory diseases could be exploited by tissue engineers. This article provides an overview of the operating principles of PAI and its broad potential for application in TERM. Impact statement Photoacoustic imaging, a new hybrid imaging technique, has demonstrated high potential in the clinical diagnostic applications. The optical and acoustic aspect of the photoacoustic imaging system works in harmony to provide better resolution at greater tissue depth. Label-free imaging of vasculature with this imaging can be used to track and monitor disease, as well as the therapeutic progression of treatment. Photoacoustic imaging has been utilized in tissue engineering to some extent; however, the full benefit of this technique is yet to be explored. The increasing availability of commercial photoacoustic systems will make application as an imaging tool for tissue engineering application more feasible. This review first provides a brief description of photoacoustic imaging and summarizes its current and potential application in tissue engineering.
Collapse
Affiliation(s)
- Binita Shrestha
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas
| | - Frank DeLuna
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas
| | - Mark A. Anastasio
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Jing Yong Ye
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas
| | - Eric M. Brey
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas
| |
Collapse
|
32
|
Ferraz FS, López JL, Lacerda SMSN, Procópio MS, Figueiredo AFA, Martins EMN, Guimarães PPG, Ladeira LO, Kitten GT, Dias FF, Domingues RZ, Costa GMJ. Biotechnological approach to induce human fibroblast apoptosis using superparamagnetic iron oxide nanoparticles. J Inorg Biochem 2020; 206:111017. [PMID: 32120160 DOI: 10.1016/j.jinorgbio.2020.111017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 01/06/2023]
Abstract
Cancer-Associated Fibroblasts (CAFs) contribute to tumour progression and have received significant attention as a therapeutic target. These cells produce growth factors, cytokines and chemokines, stimulating cancer cell proliferation and inhibiting their apoptosis. Recent advances in drug delivery have demonstrated a significant promise of iron oxide nanoparticles in clinics as theranostic agents, mainly due to their magnetic properties. Here, we designed superparamagnetic iron oxide nanoparticles (SPIONs) to induce apoptosis of human fibroblasts. SPIONs were synthesized via co-precipitation method and coated with sodium citrate (SPION_Cit). We assessed the intracellular uptake of SPIONs by human fibroblast cells, as well as their cytotoxicity and ability to induce thermal effects under the magnetic field. The efficiency and time of nanoparticle internalization were assessed by Prussian Blue staining, flow cytometry and transmission electron microscopy. SPIONs_Cit were detected in the cytoplasm of human fibroblasts 15 min after in vitro exposure, entering into cells mainly via endocytosis. Analyses through Cell Titer Blue assay, AnnexinV-fluorescein isothiocyanate (FITC) and propidium iodide (PI) cellular staining demonstrated that concentrations below 8 × 10-2 mg/mL of SPIONs_Cit did not alter cell viability of human fibroblast. Furthermore, it was also demonstrated that SPIONs_Cit associated with alternating current magnetic field were able to induce hyperthermia and human fibroblast cell death in vitro, mainly through apoptosis (83.5%), activating caspase 8 (extrinsic apoptotic via) after a short exposure period. Collectively these findings suggest that our nanoplatform is biocompatible and can be used for therapeutic purposes in human biological systems, such as inducing apoptosis of CAFs.
Collapse
Affiliation(s)
- Fausto S Ferraz
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jorge L López
- Center for Biological and Natural Sciences, Federal University of Acre, Rio Branco, AC, Brazil
| | - Samyra M S N Lacerda
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcela S Procópio
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - André F A Figueiredo
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Estefânia M N Martins
- Laboratory of Chemistry of Nanostructures, Nuclear Technology Development Center, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro P G Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz O Ladeira
- Laboratory of Nanomaterials, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gregory T Kitten
- Microscopy Center, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe F Dias
- Microscopy Center, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rosana Z Domingues
- Laboratory of Chemistry, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Guilherme M J Costa
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
33
|
Kotzabasaki MI, Sotiropoulos I, Sarimveis H. QSAR modeling of the toxicity classification of superparamagnetic iron oxide nanoparticles (SPIONs) in stem-cell monitoring applications: an integrated study from data curation to model development. RSC Adv 2020; 10:5385-5391. [PMID: 35498319 PMCID: PMC9049038 DOI: 10.1039/c9ra09475j] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/21/2020] [Indexed: 11/23/2022] Open
Abstract
The use of in silico approaches for the prediction of biomedical properties of nano-biomaterials (NBMs) can play a significant role in guiding and reducing wetlab experiments. Computational methods, such as data mining and machine learning techniques, can increase the efficiency and reduce the time and cost required for hazard and risk assesment and for designing new safer NBMs. A major obstacle in developing accurate and well-validated in silico models such as Nano Quantitative Structure-Activity Relationships (Nano-QSARs) is that although the volume of data published in the literature is increasing, the data are fragmented in many different publications and are not sufficiently curated for modelling purposes. Moreover, NBMs exhibit high complexity and heterogeneity in their structures, making data collection and curation and QSAR model development more challenging compared to traditional small molecules. The aim of this study was to construct and fully validate a Nano-QSAR model for the prediction of toxicological properties of superparamagnetic iron oxide nanoparticles (SPIONs), focusing on their application as Magnetic Resonance Imaging (MRI) contrast agents for non-invasive stem cell labelling and tracking. To achieve this goal, we first performed an extensive search through the literature for collecting and curating relevant data and we developed a dataset containing both physicochemical and toxicological properties of SPIONs. The data were analysed next, using Automated machine learning (Auto-ML) approaches for optimising the development and validation of nanotoxicity classification QSAR models of SPIONs. Further analysis of relative attribute importances revealed that physicochemical properties such as the size and the magnetic core are the dominant attributes correlated to the toxicity of SPIONs. Our results suggest that as more systematic information from NBM experimental tests becomes available, computational tools could play an important role in supporting the safety-by-design (SbD) concept in regenerative medicine and disease therapeutics.
Collapse
Affiliation(s)
- Marianna I Kotzabasaki
- School of Chemical Engineering, National Technical University of Athens 9 Heroon Polytechneiou Street, Zografou Campus 15780 Athens Greece +302107723138 +302107723236 +306936396688 +302107723237
| | - Iason Sotiropoulos
- School of Chemical Engineering, National Technical University of Athens 9 Heroon Polytechneiou Street, Zografou Campus 15780 Athens Greece +302107723138 +302107723236 +306936396688 +302107723237
| | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens 9 Heroon Polytechneiou Street, Zografou Campus 15780 Athens Greece +302107723138 +302107723236 +306936396688 +302107723237
| |
Collapse
|
34
|
Adjei IM, Yang H, Plumton G, Maldonado-Camargo L, Dobson J, Rinaldi C, Jiang H, Sharma B. Multifunctional nanoparticles for intracellular drug delivery and photoacoustic imaging of mesenchymal stem cells. Drug Deliv Transl Res 2020; 9:652-666. [PMID: 30784022 DOI: 10.1007/s13346-019-00621-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Strategies that control the differentiation of mesenchymal stem cells (MSC) and enable image-guided cell implantation and longitudinal monitoring could advance MSC-based therapies for bone defects and injuries. Here we demonstrate a multifunctional nanoparticle system that delivers resveratrol (RESV) intracellularly to improve osteogenesis and enables photoacoustic imaging of MSCs. RESV-loaded nanoparticles (RESV-NPs), formulated from poly (lactic-co-glycolic) acid and iron oxide, enhanced the stability of RESV by 18-fold and served as photoacoustic tomography (PAT) contrast for MSCs. Pre-loading MSCs with RESV-NP upregulated RUNX2 expression with a resultant increase in mineralization by 27% and 45% compared to supplementation with RESV-NP and free RESV, respectively, in 2-dimensional cultures. When grown in polyethylene glycol-based hydrogels, MSCs pre-loaded with RESV-NPs increased the overall level and homogeneity of mineralization compared to those supplemented with free RESV or RESV-NP. The PAT detected RESV-NP-loaded MSCs with a resolution of 1500 cells/μL, which ensured imaging of MSCs upon encapsulation in a PEG-based hydrogel and implantation within the rodent cranium. Significantly, RESV-NP-loaded MSCs in hydrogels did not show PAT signal dilution over time or a reduction in signal upon osteogenic differentiation. This multifunctional NP platform has the potential to advance translation of stem cell-based therapies, by improving stem cell function and consistency via intracellular drug delivery, and enabling the use of a promising emerging technology to monitor cells in a clinically relevant manner.
Collapse
Affiliation(s)
- Isaac M Adjei
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, JG56, Gainesville, FL, 32611, USA
| | - Hao Yang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, JG56, Gainesville, FL, 32611, USA
| | - Glendon Plumton
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, JG56, Gainesville, FL, 32611, USA
| | | | - Jon Dobson
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, JG56, Gainesville, FL, 32611, USA.,Department of Materials Science and Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Carlos Rinaldi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, JG56, Gainesville, FL, 32611, USA.,Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Huabei Jiang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, JG56, Gainesville, FL, 32611, USA
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, JG56, Gainesville, FL, 32611, USA.
| |
Collapse
|
35
|
Jasim KA, Gesquiere AJ. Ultrastable and Biofunctionalizable Conjugated Polymer Nanoparticles with Encapsulated Iron for Ferroptosis Assisted Chemodynamic Therapy. Mol Pharm 2019; 16:4852-4866. [PMID: 31613630 DOI: 10.1021/acs.molpharmaceut.9b00737] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We report the development of novel tumor-targeted conjugated polymer nanoparticles (CPNPs) carrying iron for chemodynamic therapy (CDT). Tumor cell killing proceeds through ferroptosis, a reactive oxygen species (ROS) mechanism that is not dependent on external activation by, for example, light, as is the case in photodynamic therapy (PDT). The ferroptosis mechanism is also not heavily reliant on oxygen availability and is, therefore, promising for the treatment of hypoxic tumors. In this work, we apply this development to the case study of melanoma, a difficult to treat cancer in advanced stages due to resistance to chemotherapy. The iron-carrying CPNPs reported here are targeted to endothelin-B receptors (EDNRB) through endothelin-3 surface moieties (EDN3-CPNPs). Our results show excellent targeting to tumor cells that overexpress EDNRB, specifically for melanoma and bladder tumor cells. In these cases, efficient cell killing, over 80% at higher doses, was found. Conversely, tumor cells not targeted by the EDN3-CPNPs show little effects of CDT, with tumor cell death under 20% in most cases. The outcomes of our work demonstrate that EDN3-CPNPs enable ferroptosis-assisted CDT and present a new therapeutic avenue for tumor treatment.
Collapse
Affiliation(s)
- Khalaf A Jasim
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, United States.,NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, United States.,Department of Chemistry, College of Science, Tikrit University, Tikrit 34001, Iraq
| | - Andre J Gesquiere
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, United States.,NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, United States.,Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida 32816, United States.,The College of Optics and Photonics (CREOL), University of Central Florida, Orlando, Florida 32816, United States
| |
Collapse
|
36
|
Anna IM, Sathy BN, Ashokan A, Gowd GS, Ramachandran R, Kochugovindan Unni AK, Manohar M, Chulliyath D, Nair S, Bhakoo K, Koyakutty M. nCP:Fe—A Biomineral Magnetic Nanocontrast Agent for Tracking Implanted Stem Cells in Brain Using MRI. ACS APPLIED BIO MATERIALS 2019; 2:5390-5403. [DOI: 10.1021/acsabm.9b00709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ida M. Anna
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Binulal N. Sathy
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Anusha Ashokan
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Genekehal Siddaramana Gowd
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Ranjith Ramachandran
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | | | - Maneesh Manohar
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - DeepthiMol Chulliyath
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Shantikumar Nair
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Kishore Bhakoo
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), Helios, Singapore 138667, Singapore
| | - Manzoor Koyakutty
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| |
Collapse
|
37
|
Plan Sangnier A, Van de Walle AB, Curcio A, Le Borgne R, Motte L, Lalatonne Y, Wilhelm C. Impact of magnetic nanoparticle surface coating on their long-term intracellular biodegradation in stem cells. NANOSCALE 2019; 11:16488-16498. [PMID: 31453605 DOI: 10.1039/c9nr05624f] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Magnetic nanoparticles (MNPs) internalized within stem cells have paved the way for remote magnetic cell manipulation and imaging in regenerative medicine. A full understanding of their interactions with stem cells and of their fate in the intracellular environment is then required, in particular with respect to their surface coatings. Here, we investigated the biological interactions of MNPs composed of an identical magnetic core but coated with different molecules: phosphonoacetic acid, polyethylene glycol phosphonic carboxylic acid, caffeic acid, citric acid, and polyacrylic acid. These coatings vary in the nature of the chelating function, the number of binding sites, and the presence or absence of a polymer. The nanoparticle magnetism was systematically used as an indicator of their internalization within human stem cells and of their structural long-term biodegradation in a 3D stem cell spheroid model. Overall, we evidence that the coating impacts the aggregation status of the nanoparticles and subsequently their uptake within stem cells, but it has little effect on their intracellular degradation. Only a high number of chelating functions (polyacrylic acid) had a significant protective effect. Interestingly, when the nanoparticles aggregated prior to cellular internalization, less degradation was also observed. Finally, for all coatings, a robust dose-dependent intracellular degradation rate was demonstrated, with higher doses of internalized nanoparticles leading to a lower degradation extent.
Collapse
Affiliation(s)
- Anouchka Plan Sangnier
- Laboratoire Matière et Systèmes, Complexes MSC, UMR 7057, CNRS & University Paris Diderot, 75205, Paris Cedex 13, France. and Inserm, U1148, Laboratory for Vascular Translational Science, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France.
| | - Aurore B Van de Walle
- Laboratoire Matière et Systèmes, Complexes MSC, UMR 7057, CNRS & University Paris Diderot, 75205, Paris Cedex 13, France.
| | - Alberto Curcio
- Laboratoire Matière et Systèmes, Complexes MSC, UMR 7057, CNRS & University Paris Diderot, 75205, Paris Cedex 13, France.
| | - Rémi Le Borgne
- Institut Jacques Monod, CNRS UMR 7592, Sorbonne Paris Cité, Université Paris Diderot, Paris, France
| | - Laurence Motte
- Inserm, U1148, Laboratory for Vascular Translational Science, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France.
| | - Yoann Lalatonne
- Inserm, U1148, Laboratory for Vascular Translational Science, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France. and Services de Biochimie et de Médecine Nucléaire, Hôpital Avicenne Assistance Publique-Hôpitaux de Paris, F-93009 Bobigny, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes, Complexes MSC, UMR 7057, CNRS & University Paris Diderot, 75205, Paris Cedex 13, France.
| |
Collapse
|
38
|
Duan J, Du J, Jin R, Zhu W, Liu L, Yang L, Li M, Gong Q, Song B, Anderson JM, Ai H. Iron oxide nanoparticles promote vascular endothelial cells survival from oxidative stress by enhancement of autophagy. Regen Biomater 2019; 6:221-229. [PMID: 31404327 PMCID: PMC6683953 DOI: 10.1093/rb/rbz024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/15/2019] [Accepted: 05/28/2019] [Indexed: 02/05/2023] Open
Abstract
Dextran-coated superparamagnetic iron oxide nanoparticles (Dex-SPIONs) are excellent magnetic resonance imaging contrast agents for disease diagnosis and therapy. They can be delivered to target tissues mainly though vascular endothelium cells, which are major targets of oxidative stress. In cardiovascular cells, autophagy serves primarily on a pro-survival approach that protects the cells from oxidative stress even some autophagy inducers have been developed for adjuvant therapy of cardiovascular disorders. Our study demonstrated that the nanoparticles could be taken up by human umbilical vein endothelial cells (HUVECs) without causing obvious cytotoxicity but triggering autophagy. Furthermore, our results revealed that Dex-SPIONs could enhance HUVECs survival and reverse the reduction of nitric oxide secretion under the condition of H2O2 damage. However, these effects could be diminished by the autophagy inhibitor. In particular, we discovered that Dex-SPIONs evoked autophagy in HUVECs by reducing the phosphorylation of PRAS40, an upstream regulator of autophagy initiation. These results suggested that Dex-SPIONs functions as an autophagic-related antioxidant in HUVECs which may be utilized as an adjuvant therapy to cardiovascular disease associated with oxidative stress.
Collapse
Affiliation(s)
- Jimei Duan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P.R. China
| | - Jiuju Du
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P.R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P.R. China
- Correspondence address. National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, P.R. China. Tel: +86-28-8541-3991; Fax: +86-28-8541-3991; E-mail: (R.J.); (H.A.)
| | - Wencheng Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P.R. China
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Li Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P.R. China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P.R. China
| | - Mengye Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P.R. China
| | - Qiyong Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - James M Anderson
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Macromolecular Science, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, P.R. China
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, P.R. China
- Correspondence address. National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, P.R. China. Tel: +86-28-8541-3991; Fax: +86-28-8541-3991; E-mail: (R.J.); (H.A.)
| |
Collapse
|
39
|
Evaluation of Chemical and Green Synthesized Iron Oxide Nanoparticles’ Associated Renal Toxicity in Different Experimental Models: A Comparative Study. J CLUST SCI 2019. [DOI: 10.1007/s10876-018-01492-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
40
|
Dulińska-Litewka J, Łazarczyk A, Hałubiec P, Szafrański O, Karnas K, Karewicz A. Superparamagnetic Iron Oxide Nanoparticles-Current and Prospective Medical Applications. MATERIALS 2019; 12:ma12040617. [PMID: 30791358 PMCID: PMC6416629 DOI: 10.3390/ma12040617] [Citation(s) in RCA: 263] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 02/02/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
Abstract
The recent, fast development of nanotechnology is reflected in the medical sciences. Superparamagnetic Iron Oxide Nanoparticles (SPIONs) are an excellent example. Thanks to their superparamagnetic properties, SPIONs have found application in Magnetic Resonance Imaging (MRI) and magnetic hyperthermia. Unlike bulk iron, SPIONs do not have remnant magnetization in the absence of the external magnetic field; therefore, a precise remote control over their action is possible. This makes them also useful as a component of the advanced drug delivery systems. Due to their easy synthesis, biocompatibility, multifunctionality, and possibility of further surface modification with various chemical agents, SPIONs could support many fields of medicine. SPIONs have also some disadvantages, such as their high uptake by macrophages. Nevertheless, based on the ongoing studies, they seem to be very promising in oncological therapy (especially in the brain, breast, prostate, and pancreatic tumors). The main goal of our paper is, therefore, to present the basic properties of SPIONs, to discuss their current role in medicine, and to review their applications in order to inspire future developments of new, improved SPION systems.
Collapse
Affiliation(s)
- Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland.
| | - Agnieszka Łazarczyk
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland.
| | - Przemysław Hałubiec
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland.
| | - Oskar Szafrański
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland.
| | - Karolina Karnas
- Department of Chemistry, Jagiellonian University, 2 Gronostajowa St., 30-387 Kraków, Poland.
| | - Anna Karewicz
- Department of Chemistry, Jagiellonian University, 2 Gronostajowa St., 30-387 Kraków, Poland.
| |
Collapse
|
41
|
Yoo J, Yun C, Bui N, Oh J, Nam S. Photoacoustic Monitoring of the Viability of Mesenchymal Stem Cells Labeled with Indocyanine Green. Ing Rech Biomed 2019. [DOI: 10.1016/j.irbm.2018.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Hiremath CG, Kariduraganavar MY, Hiremath MB. Synergistic delivery of 5-fluorouracil and curcumin using human serum albumin-coated iron oxide nanoparticles by folic acid targeting. Prog Biomater 2018; 7:297-306. [PMID: 30565175 PMCID: PMC6304179 DOI: 10.1007/s40204-018-0104-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/11/2018] [Indexed: 01/24/2023] Open
Abstract
Human serum albumin is the most abundant protein in plasma with the ability to bind to a variety of drug molecules. Magnetic nanoparticles are being extensively used in drug delivery due to its intrinsic magnetic properties. In this work, we have synthesized human serum albumin-coated citrate-functionalized iron oxide nanoparticles by CDI coupling. Furthermore, folic acid was decorated on human serum albumin by EDC and NHS coupling to confer targetability. Two cytotoxic drugs 5-fluorouracil (5FU) and curcumin were co-delivered. Wherein, the former is an anticancer agent and latter is a drug resistance depressor of former. The nanoparticles showed good aqueous dispersibility with a zeta potential of - 49.1 mV and magnetic core size in the range of 10-15 nm, thus exhibiting good magnetic property with magnetic saturation of 33.59 emu/g. Controlled drug release behavior was noticed in both drugs with faster release profile of 5FU. Nanoparticles also showed good cytotoxicity with lower IC50 values in the presence of magnetic field. The contrasting difference was noticed in folic acid-decorated and non-decorated composites, similarly in the presence of magnetic field where cell uptake was enhanced.
Collapse
Affiliation(s)
- Chinmay G. Hiremath
- Department of Biotechnology and Microbiology, Karnatak University, Dharwad, Karnataka 580003 India
| | | | - Murigendra B. Hiremath
- Department of Biotechnology and Microbiology, Karnatak University, Dharwad, Karnataka 580003 India
| |
Collapse
|
43
|
Van de Walle A, Faissal W, Wilhelm C, Luciani N. Role of growth factors and oxygen to limit hypertrophy and impact of high magnetic nanoparticles dose during stem cell chondrogenesis. Comput Struct Biotechnol J 2018; 16:532-542. [PMID: 30524668 PMCID: PMC6260287 DOI: 10.1016/j.csbj.2018.10.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/26/2022] Open
Abstract
Due to an unmet clinical need of curative treatments for osteoarthritic patients, tissue engineering strategies that propose the development of cartilage tissue replacements from stem cells have emerged. Some of these strategies are based on the internalization of magnetic nanoparticles into stem cells to then initiate the chondrogenesis via magnetic compaction. A major difficulty is to drive the chondrogenic differentiation of the cells such as they produce an extracellular matrix free of hypertrophic collagen. An additional difficulty has to be overcome when nanoparticles are used, knowing that a high dose of nanoparticles can limit the chondrogenesis. We here propose a gene-based analysis of the effects of chemical factors (growth factors, hypoxia) on the chondrogenic differentiation of human mesenchymal stem cells both with and without nanoparticles. We focus on the synthesis of two of the most important constituents present in the cartilaginous extracellular matrix (Collagen II and Aggrecan) and on the expression of collagen X, the signature of hypertrophic cartilage, in order to provide a quantitative index of the type of cartilage produced (i.e. hyaline, hypertrophic). We demonstrate that by applying specific environmental conditions, gene expression can be directed toward the production of hyaline cartilage, with limited hypertrophy. Besides, a combination of the growth factors IGF-1, TGF-β3, with a hypoxic conditioning remarkably reduced the impact of high nanoparticles concentration.
Collapse
Affiliation(s)
| | | | - Claire Wilhelm
- Corresponding authors at: Laboratoire MSC, UMR 7057 CNRS, University Paris Diderot, France.
| | - Nathalie Luciani
- Corresponding authors at: Laboratoire MSC, UMR 7057 CNRS, University Paris Diderot, France.
| |
Collapse
|
44
|
Kerans FFA, Lungaro L, Azfer A, Salter DM. The Potential of Intrinsically Magnetic Mesenchymal Stem Cells for Tissue Engineering. Int J Mol Sci 2018; 19:E3159. [PMID: 30322202 PMCID: PMC6214112 DOI: 10.3390/ijms19103159] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/04/2018] [Accepted: 10/09/2018] [Indexed: 12/16/2022] Open
Abstract
The magnetization of mesenchymal stem cells (MSC) has the potential to aid tissue engineering approaches by allowing tracking, targeting, and local retention of cells at the site of tissue damage. Commonly used methods for magnetizing cells include optimizing uptake and retention of superparamagnetic iron oxide nanoparticles (SPIONs). These appear to have minimal detrimental effects on the use of MSC function as assessed by in vitro assays. The cellular content of magnetic nanoparticles (MNPs) will, however, decrease with cell proliferation and the longer-term effects on MSC function are not entirely clear. An alternative approach to magnetizing MSCs involves genetic modification by transfection with one or more genes derived from Magnetospirillum magneticum AMB-1, a magnetotactic bacterium that synthesizes single-magnetic domain crystals which are incorporated into magnetosomes. MSCs with either or mms6 and mmsF genes are followed by bio-assimilated synthesis of intracytoplasmic magnetic nanoparticles which can be imaged by magnetic resonance (MR) and which have no deleterious effects on MSC proliferation, migration, or differentiation. The stable transfection of magnetosome-associated genes in MSCs promotes assimilation of magnetic nanoparticle synthesis into mammalian cells with the potential to allow MR-based cell tracking and, through external or internal magnetic targeting approaches, enhanced site-specific retention of cells for tissue engineering.
Collapse
Affiliation(s)
- Fransiscus F A Kerans
- Centre for Genomics and Experimental Medicine, MRC IGMM, University of Edinburgh, Edinburgh EH4 2XU, UK.
| | - Lisa Lungaro
- Centre for Genomics and Experimental Medicine, MRC IGMM, University of Edinburgh, Edinburgh EH4 2XU, UK.
| | - Asim Azfer
- Centre for Genomics and Experimental Medicine, MRC IGMM, University of Edinburgh, Edinburgh EH4 2XU, UK.
| | - Donald M Salter
- Centre for Genomics and Experimental Medicine, MRC IGMM, University of Edinburgh, Edinburgh EH4 2XU, UK.
| |
Collapse
|
45
|
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
|
46
|
Rajkovic O, Potjewyd G, Pinteaux E. Regenerative Medicine Therapies for Targeting Neuroinflammation After Stroke. Front Neurol 2018; 9:734. [PMID: 30233484 PMCID: PMC6129611 DOI: 10.3389/fneur.2018.00734] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/13/2018] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a major pathological event following ischemic stroke that contributes to secondary brain tissue damage leading to poor functional recovery. Following the initial ischemic insult, post-stroke inflammatory damage is driven by initiation of a central and peripheral innate immune response and disruption of the blood-brain barrier (BBB), both of which are triggered by the release of pro-inflammatory cytokines and infiltration of circulating immune cells. Stroke therapies are limited to early cerebral blood flow reperfusion, and whilst current strategies aim at targeting neurodegeneration and/or neuroinflammation, innovative research in the field of regenerative medicine aims at developing effective treatments that target both the acute and chronic phase of inflammation. Anti-inflammatory regenerative strategies include the use of nanoparticles and hydrogels, proposed as therapeutic agents and as a delivery vehicle for encapsulated therapeutic biological factors, anti-inflammatory drugs, stem cells, and gene therapies. Biomaterial strategies-through nanoparticles and hydrogels-enable the administration of treatments that can more effectively cross the BBB when injected systemically, can be injected directly into the brain, and can be 3D-bioprinted to create bespoke implants within the site of ischemic injury. In this review, these emerging regenerative and anti-inflammatory approaches will be discussed in relation to ischemic stroke, with a perspective on the future of stroke therapies.
Collapse
Affiliation(s)
- Olivera Rajkovic
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Geoffrey Potjewyd
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
47
|
Zeng Y, Zhu J, Wang J, Parasuraman P, Busi S, Nauli SM, Wáng YXJ, Pala R, Liu G. Functional probes for cardiovascular molecular imaging. Quant Imaging Med Surg 2018; 8:838-852. [PMID: 30306063 PMCID: PMC6177368 DOI: 10.21037/qims.2018.09.19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/17/2018] [Indexed: 12/26/2022]
Abstract
Cardiovascular diseases (CVDs) are a severely threatening disorder and frequently cause death in industrialized countries, posing critical challenges to modern research and medicine. Molecular imaging has been heralded as the solution to many problems encountered in individuals living with CVD. The use of probes in cardiovascular molecular imaging is causing a paradigmatic shift from regular imaging techniques, to future advanced imaging technologies, which will facilitate the acquisition of vital information at the cellular and molecular level. Advanced imaging for CVDs will help early detection of disease development, allow early therapeutic intervention, and facilitate better understanding of fundamental biological processes. To promote a better understanding of cardiovascular molecular imaging, this article summarizes the current developments in the use of molecular probes, highlighting some of the recent advances in probe design, preparation, and functional modification.
Collapse
Affiliation(s)
- Yun Zeng
- Department of Pharmacology, Xiamen Medical College, Xiamen 361008, China
| | - Jing Zhu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Junqing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- Department of Imaging and Interventional Radiology, Prince of Wales Hospital, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Paramanantham Parasuraman
- Departments of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Siddhardha Busi
- Departments of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Surya M. Nauli
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, California, USA
| | - Yì Xiáng J. Wáng
- Department of Imaging and Interventional Radiology, Prince of Wales Hospital, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Rajasekharreddy Pala
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, California, USA
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
48
|
Xia Y, Sun J, Zhao L, Zhang F, Liang XJ, Guo Y, Weir MD, Reynolds MA, Gu N, Xu HHK. Magnetic field and nano-scaffolds with stem cells to enhance bone regeneration. Biomaterials 2018; 183:151-170. [PMID: 30170257 DOI: 10.1016/j.biomaterials.2018.08.040] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/10/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
Novel strategies utilizing magnetic nanoparticles (MNPs) and magnetic fields are being developed to enhance bone tissue engineering efficacy. This article first reviewed cutting-edge research on the osteogenic enhancements via magnetic fields and MNPs. Then the current developments in magnetic strategies to improve the cells, scaffolds and growth factor deliveries were described. The magnetic-cell strategies included cell labeling, targeting, patterning, and gene modifications. MNPs were incorporated to fabricate magnetic composite scaffolds, as well as to construct delivery systems for growth factors, drugs and gene transfections. The novel methods using magnetic nanoparticles and scaffolds with magnetic fields and stem cells increased the osteogenic differentiation, angiogenesis and bone regeneration by 2-3 folds over those of the controls. The mechanisms of magnetic nanoparticles and scaffolds with magnetic fields and stem cells to enhance bone regeneration were identified as involving the activation of signaling pathways including MAPK, integrin, BMP and NF-κB. Potential clinical applications of magnetic nanoparticles and scaffolds with magnetic fields and stem cells include dental, craniofacial and orthopedic treatments with substantially increased bone repair and regeneration efficacy.
Collapse
Affiliation(s)
- Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Jianfei Sun
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Liang Zhao
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yu Guo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Michael D Weir
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
49
|
Adamiano A, Iafisco M, Sandri M, Basini M, Arosio P, Canu T, Sitia G, Esposito A, Iannotti V, Ausanio G, Fragogeorgi E, Rouchota M, Loudos G, Lascialfari A, Tampieri A. On the use of superparamagnetic hydroxyapatite nanoparticles as an agent for magnetic and nuclear in vivo imaging. Acta Biomater 2018; 73:458-469. [PMID: 29689381 DOI: 10.1016/j.actbio.2018.04.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 12/14/2022]
Abstract
The identification of alternative biocompatible magnetic NPs for advanced clinical application is becoming an important need due to raising concerns about iron accumulation in soft issues associated to the administration of superparamagnetic iron oxide nanoparticles (NPs). Here, we report on the performance of previously synthetized iron-doped hydroxyapatite (FeHA) NPs as contrast agent for magnetic resonance imaging (MRI). The MRI contrast abilities of FeHA and Endorem® (dextran coated iron oxide NPs) were assessed by 1H nuclear magnetic resonance relaxometry and their performance in healthy mice was monitored by a 7 Tesla scanner. FeHA applied a higher contrast enhancement, and had a longer endurance in the liver with respect to Endorem® at iron equality. Additionally, a proof of concept of FeHA use as scintigraphy imaging agent for positron emission tomography (PET) and single photon emission computed tomography (SPECT) was given labeling FeHA with 99mTc-MDP by a straightforward surface functionalization process. Scintigraphy/x-ray fused imaging and ex vivo studies confirmed its dominant accumulation in the liver, and secondarily in other organs of the mononuclear phagocyte system. FeHA efficiency as MRI-T2 and PET-SPECT imaging agent combined to its already reported intrinsic biocompatibility qualifies it as a promising material for innovative nanomedical applications. STATEMENT OF SIGNIFICANCE The ability of iron-doped hydroxyapatite nanoaprticles (FeHA) to work in vivo as imaging agents for magnetic resonance (MR) and nuclear imaging is demonstrated. FeHA applied an higher MR contrast in the liver, spleen and kidneys of mice with respect to Endorem®. The successful radiolabeling of FeHA allowed for scintigraphy/X-ray and ex vivo biodistribution studies, confirming MR results and envisioning FeHA application for dual-imaging.
Collapse
|
50
|
Ding Z, Liu P, Hu D, Sheng Z, Yi H, Gao G, Wu Y, Zhang P, Ling S, Cai L. Redox-responsive dextran based theranostic nanoparticles for near-infrared/magnetic resonance imaging and magnetically targeted photodynamic therapy. Biomater Sci 2018; 5:762-771. [PMID: 28256661 DOI: 10.1039/c6bm00846a] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Photodynamic therapy (PDT) is a site-specific treatment of cancer using much lower optical power densities with minimal nonspecific damage to normal tissues. To improve the therapeutic efficiency of PDT, we fabricated a multifunctional theranostic nanoparticle system (DSSCe6@Fe3O4 NPs) by loading Fe3O4 nanoparticles in redox-responsive chlorin e6 (Ce6)-conjugated dextran nanoparticles for near-infrared (NIR)/magnetic resonance (MR) dual-modality imaging and magnetic targeting. The obtained DSSCe6@Fe3O4 NPs demonstrated a uniform nanospherical morphology consisting of Fe3O4 clusters. The fluorescence signal of Ce6 of this theranostic system could turn "ON" from a self-quenching state in a reductive intracellular environment. T2-Weighted MR imaging revealed a high transverse relaxivity (r2) measured to be 194.4 S-1 mM-1, confirming that it was also a distinctive contrast agent in T2-weighted MR imaging. Confocal images and flow cytometry results showed that the cellular uptake of DSSCe6@Fe3O4 NPs was enhanced effectively under an extra magnetic field, which resulted in promoted PDT therapeutic efficiency. In vivo MR imaging showed that DSSCe6@Fe3O4 NPs effectively accumulated in tumors under an extra magnetic field. These results illustrated that the DSSCe6@Fe3O4 NPs could be a promising theranostic system for both NIR/MR imaging-guided PDT precision therapy.
Collapse
Affiliation(s)
- Zexuan Ding
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China. and Nano Science and Technology Institute, University of Science & Technology of China, Suzhou, 215123, P. R. China
| | - Peng Liu
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China. and Department of Chemical and Biomolecular Engineering, National University of Singapore, Kent Ridge 117576, Singapore
| | - Dehong Hu
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Zonghai Sheng
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Huqiang Yi
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Guanhui Gao
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Yayun Wu
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Shaozhi Ling
- General Hospital of Chinese Armed Police Forces, Beijing 100039, P.R. China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| |
Collapse
|