1
|
Iqbal J, Courville E, Kazim SF, Kogan M, Schmidt MH, Bowers CA. Role of nanotechnology in neurosurgery: A review of recent advances and their applications. World Neurosurg X 2024; 22:100298. [PMID: 38455250 PMCID: PMC10918265 DOI: 10.1016/j.wnsx.2024.100298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Affiliation(s)
- Javed Iqbal
- School of Medicine, King Edward Medical University, Lahore, Pakistan
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Evan Courville
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Syed Faraz Kazim
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Michael Kogan
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Meic H. Schmidt
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Christian A. Bowers
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| |
Collapse
|
2
|
Gao X, Li S, Yang Y, Yang S, Yu B, Zhu Z, Ma T, Zheng Y, Wei B, Hao Y, Wu H, Zhang Y, Guo L, Gao X, Wei Y, Xue B, Li J, Feng X, Lu L, Xia B, Huang J. A Novel Magnetic Responsive miR-26a@SPIONs-OECs for Spinal Cord Injury: Triggering Neural Regeneration Program and Orienting Axon Guidance in Inhibitory Astrocytic Environment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304487. [PMID: 37789583 PMCID: PMC10646239 DOI: 10.1002/advs.202304487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/12/2023] [Indexed: 10/05/2023]
Abstract
Addressing the challenge of promoting directional axonal regeneration in a hostile astrocytic scar, which often impedes recovery following spinal cord injury (SCI), remains a daunting task. Cell transplantation is a promising strategy to facilitate nerve restoration in SCI. In this research, a pro-regeneration system is developed, namely miR-26a@SPIONs-OECs, for olfactory ensheathing cells (OECs), a preferred choice for promoting nerve regeneration in SCI patients. These entities show high responsiveness to external magnetic fields (MF), leading to synergistic multimodal cues to enhance nerve regeneration. First, an MF stimulates miR-26a@SPIONs-OECs to release extracellular vesicles (EVs) rich in miR-26a. This encourages axon growth by inhibiting PTEN and GSK-3β signaling pathways in neurons. Second, miR-26a@SPIONs-OECs exhibit a tendency to migrate and orientate along the direction of the MF, thereby potentially facilitating neuronal reconnection through directional neurite elongation. Third, miR-26a-enriched EVs from miR-26a@SPIONs-OECs can interact with host astrocytes, thereby diminishing inhibitory cues for neurite growth. In a rat model of SCI, the miR-26a@SPIONs-OECs system led to significantly improved morphological and motor function recovery. In summary, the miR-26a@SPIONS-OECs pro-regeneration system offers innovative insights into engineering exogenous cells with multiple additional cues, augmenting their efficacy for stimulating and guiding nerve regeneration within a hostile astrocytic scar in SCI.
Collapse
Affiliation(s)
- Xue Gao
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Shengyou Li
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Yujie Yang
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Shijie Yang
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Beibei Yu
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Zhijie Zhu
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Teng Ma
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Yi Zheng
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Bin Wei
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Yiming Hao
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Haining Wu
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Yongfeng Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Lingli Guo
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Xueli Gao
- School of Ecology and EnvironmentNorthwestern Polytechnical UniversityXi'an710072P. R. China
| | - Yitao Wei
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Borui Xue
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Jianzhong Li
- Department of Thoracic SurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Xue Feng
- Department of Cell BiologySchool of MedicineNorthwest UniversityXi'an710032P. R. China
| | - Lei Lu
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi International Joint Research Center for Oral DiseasesDepartment of Oral Anatomy and Physiology and TMDSchool of Stomatologythe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Bing Xia
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Jinghui Huang
- Department of OrthopaedicsXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| |
Collapse
|
3
|
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
|
4
|
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: 18.3] [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
|
5
|
Chen C, Ge J, Gao Y, Chen L, Cui J, Zeng J, Gao M. Ultrasmall superparamagnetic iron oxide nanoparticles: A next generation contrast agent for magnetic resonance imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1740. [PMID: 34296533 DOI: 10.1002/wnan.1740] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
As a research hotspot, the development of magnetic resonance imaging (MRI) contrast agents has attracted great attention over the past decades for improving the accuracy of diagnosis. Ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles with core diameter smaller than 5.0 nm are expected to become a next generation of contrast agents owing to their excellent MRI performance, long blood circulation time upon proper surface modification, renal clearance capacity, and remarkable biosafety profile. On top of these merits, USPIO nanoparticles are used for developing not only T1 contrast agents, but also T2 /T1 switchable contrast agents via assembly/disassembly approaches. In recent years, as a new type of contrast agents, USPIO nanoparticles have shown considerable applications in the diagnosis of various diseases such as vascular pathological changes and inflammations apart from malignant tumors. In this review, we are focusing on the state-of-the-art developments and the latest applications of USPIO nanoparticles as MRI contrast agents to discuss their advantages and future prospects. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Can Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Jianxian Ge
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Yun Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Lei Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Jiabin Cui
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China.,Shanghai University of Medicine and Health Sciences (SUMHS), Shanghai, China
| |
Collapse
|
6
|
Zhuang L, Kong Y, Yang S, Lu F, Gong Z, Zhan S, Liu M. Dynamic changes of inflammation and apoptosis in cerebral ischemia‑reperfusion injury in mice investigated by ferumoxytol‑enhanced magnetic resonance imaging. Mol Med Rep 2021; 23:282. [PMID: 33604682 PMCID: PMC7905325 DOI: 10.3892/mmr.2021.11921] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 03/13/2020] [Indexed: 01/21/2023] Open
Abstract
The inflammatory response and apoptosis are key factors in cerebral ischemia-reperfusion injury. The severity of the inflammatory reaction and apoptosis has an important impact on the prognosis of stroke. The ultrasmall superparamagnetic iron oxide particle has provided an effective magnetic resonance molecular imaging method for dynamic observation of the cell infiltration process in vivo. The aims of the present study were to investigate the inflammatory response of cerebral ischemia-reperfusion injury in mice using ferumoxytol-enhanced magnetic resonance imaging, and to observe the dynamic changes of inflammatory response and apoptosis. In the present study a C57BL/6n mouse cerebral ischemia-reperfusion model was established by blocking the right middle cerebral artery with an occluding suture. Subsequently, the mice were injected with ferumoxytol via the tail vein, and magnetic resonance scanning was performed at corresponding time points to observe the signal changes. Furthermore, blood samples were used to measure the level of serum inflammatory factors, and histological staining was performed to assess the number of iron-swallowing microglial cells and apoptotic cells. The present results suggested that there was no significant difference in the serum inflammatory factors tumor necrosis factor-α and interleukin 1β between the middle cerebral artery occlusion (MCAO) and MCAO + ferumoxytol groups injected with ferumoxytol and physiological saline. The lowest signal ratio in the negative enhancement region was decreased 24 h after reperfusion in mice injected with ferumoxytol. The proportion of iron-swallowing microglial cells and TUNEL-positive cells were the highest at 24 h after reperfusion, and decreased gradually at 48 and 72 h after reperfusion. Therefore, the present results indicated that ferumoxytol injection of 18 mg Fe/kg does not affect the inflammatory response in the acute phase of cerebral ischemia and reperfusion. Ferumoxytol-enhanced magnetic resonance imaging can be used as an effective means to monitor the inflammatory response in the acute phase of cerebral ischemia-reperfusion injury. Furthermore, it was found that activation of the inflammatory response and apoptosis in the acute stage of cerebral ischemia-reperfusion injury is consistent.
Collapse
Affiliation(s)
- Lihua Zhuang
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yingnan Kong
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Shuohui Yang
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Fang Lu
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Zhigang Gong
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Songhua Zhan
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Mengxiao Liu
- MR Scientific Marketing, Siemens Healthcare, Shanghai 201318, P.R. China
| |
Collapse
|
7
|
Raviraj V, Pham BTT, Kim BJ, Pham NTH, Kok LF, Painter N, Delic NC, Jones SK, Hawkett BS, Lyons JG. Non-invasive transdermal delivery of chemotherapeutic molecules in vivo using superparamagnetic iron oxide nanoparticles. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00079-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
The skin is both a target and a potential conduit for the delivery of drugs, but its cornified cell layer resists penetration by most molecules. This study investigated the potential of superparamagnetic iron oxide nanoparticles to facilitate the transdermal delivery of anticancer agents.
Results
Chemotherapeutic cancer drugs were applied with or without nanoparticles to the skin of hairless mice, and their ability to penetrate the skin was assessed using fluorescence microscopy and tumor growth. Nanoparticles enhanced the penetration of the skin by doxorubicin and 5-fluorouracil as determined by fluorescence microscopy and growth retardation of experimental melanoma in immunocompetent, syngeneic mice. This drug enhancement did not require conjugation or encapsulation of the drugs by the nanoparticles—simple co-administration sufficed. Nanoparticles applied topically to melanomas increased the cytotoxicity and immune cell infiltration induced by co-administered 5-fluorouracil, and also reduced vascularization of the tumors independently of 5-fluorouracil.
Conclusion
Correctly formulated superparamagnetic iron oxide nanoparticles can facilitate the chemotherapeutic effectiveness of cytotoxic drugs on skin tumors by both increasing their transdermal penetration and ameliorating host–tumor interactions. This enhancement of skin penetration occurs without the need for conjugation or encapsulation of the co-administered drugs, and so will likely be applicable to other drugs, also.
Collapse
|
8
|
Kayani Z, Dehdari Vais R, Soratijahromi E, Mohammadi S, Sattarahmady N. Curcumin-gold-polyethylene glycol nanoparticles as a nanosensitizer for photothermal and sonodynamic therapies: In vitro and animal model studies. Photodiagnosis Photodyn Ther 2020; 33:102139. [PMID: 33310015 DOI: 10.1016/j.pdpdt.2020.102139] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/21/2020] [Accepted: 12/04/2020] [Indexed: 12/18/2022]
Abstract
Photothermal and ultrasound therapies are novel non-invasive strategies for tumor treatment which are equipped with a photosensitizer and sonosensitizer subsequent activation by laser irradiation and ultrasound exposure. In this study, curcumin-gold-polyethylene glycol nanoparticles (Cur-Au NPs-PEG) were synthesized, and the dual role in photothermal (PTT) and sonodynamic (SDT) therapies of melanoma cancer was evaluated. The toxicity effect of Cur-Au NPs-PEG against a mouse malignant melanoma cell line C540 (B16/F10) was firstly inspected in vitro. Cur-Au NPs-PEG provided a hyperthermal microenvironment and generated reactive oxygen species upon PTT and STD, respectively, with representing synergism effects. Studies in vivo in a tumor-bearing animal also demonstrate the superiority of PTT and SDT in destroying melanoma tumor.
Collapse
Affiliation(s)
- Z Kayani
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - R Dehdari Vais
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - E Soratijahromi
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - S Mohammadi
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Radiology Technology, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - N Sattarahmady
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Zeng Y, Li Z, Zhu H, Gu Z, Zhang H, Luo K. Recent Advances in Nanomedicines for Multiple Sclerosis Therapy. ACS APPLIED BIO MATERIALS 2020; 3:6571-6597. [PMID: 35019387 DOI: 10.1021/acsabm.0c00953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yujun Zeng
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqian Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, California 91711, United States
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
McGinley LM, Willsey MS, Kashlan ON, Chen KS, Hayes JM, Bergin IL, Mason SN, Stebbins AW, Kwentus JF, Pacut C, Kollmer J, Sakowski SA, Bell CB, Chestek CA, Murphy GG, Patil PG, Feldman EL. Magnetic resonance imaging of human neural stem cells in rodent and primate brain. Stem Cells Transl Med 2020; 10:83-97. [PMID: 32841522 PMCID: PMC7780819 DOI: 10.1002/sctm.20-0126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Stem cell transplantation therapies are currently under investigation for central nervous system disorders. Although preclinical models show benefit, clinical translation is somewhat limited by the absence of reliable noninvasive methods to confirm targeting and monitor transplanted cells in vivo. Here, we assess a novel magnetic resonance imaging (MRI) contrast agent derived from magnetotactic bacteria, magneto‐endosymbionts (MEs), as a translatable methodology for in vivo tracking of stem cells after intracranial transplantation. We show that ME labeling provides robust MRI contrast without impairment of cell viability or other important therapeutic features. Labeled cells were visualized immediately post‐transplantation and over time by serial MRI in nonhuman primate and mouse brain. Postmortem tissue analysis confirmed on‐target grft location, and linear correlations were observed between MRI signal, cell engraftment, and tissue ME levels, suggesting that MEs may be useful for determining graft survival or rejection. Overall, these findings indicate that MEs are an effective tool for in vivo tracking and monitoring of cell transplantation therapies with potential relevance to many cellular therapy applications.
Collapse
Affiliation(s)
- Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew S Willsey
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Osama N Kashlan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin S Chen
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - John M Hayes
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ingrid L Bergin
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shayna N Mason
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Aaron W Stebbins
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Crystal Pacut
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer Kollmer
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Caleb B Bell
- Bell Biosystems, San Francisco, California, USA.,G4S Capital & Ikigai Accelerator, Santa Clara, California, USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Electrical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Neuroscience and Robotics Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Geoffrey G Murphy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Parag G Patil
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Wang F, Wang Z, Wang F, Dong K, Zhang J, Sun YJ, Liu CF, Xing MJ, Cheng X, Wei S, Zheng JW, Zhao XF, Wang XM, Fu J, Song HF. Comparative strategies for stem cell biodistribution in a preclinical study. Acta Pharmacol Sin 2020; 41:572-580. [PMID: 31705124 PMCID: PMC7470780 DOI: 10.1038/s41401-019-0313-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy represents the potential alternative effective strategy for some diseases that lack effective treatment currently. Correspondingly, it is crucial to establish high-sensitive and reliable quantification assay for tracing exogenous cell migration. In the present study, we first used both bioluminescence imaging (BLI) indirect labeling (human norepinephrine transporter-luciferase reporter system) and 89zirconium (89Zr)-hNSCs direct labeling combined with positron emission tomography/computer tomography (PET/CT) system for tracking human neural stem cells (hNSCs) migration into the brain via nasal administration in preclinical study. But the above two methods failed to give the biodistribution profile due to their low sensitivity. Considering its superior sensitivity and absolute quantitation capability, we developed and validated the droplet digital PCR (ddPCR) targeting species-specific gene in frozen and paraffin sections, slices, and whole blood with the sensitivity of 100–200 hNSCs. Accurate and high throughput quantification could be performed using ddPCR with the coefficient of variation (CVs) of lower quality control (LQC) below 30%. In combination with immunohistochemistry and ddPCR, we confirmed the migration of hNSCs into the brain via nasal administration, which supported the efficacy of hNSCs in MPTP-treated mice, an animal model of Parkinson’s disease. In conclusion, the present study is the first to report the application of ddPCR in the pharmacokinetics profile description of tracking of hNSCs in preclinical studies.
Collapse
|
12
|
Zare S, Mehrabani D, Jalli R, Saeedi Moghadam M, Manafi N, Mehrabani G, Jamhiri I, Ahadian S. MRI-Tracking of Dental Pulp Stem Cells In Vitro and In Vivo Using Dextran-Coated Superparamagnetic Iron Oxide Nanoparticles. J Clin Med 2019; 8:E1418. [PMID: 31505807 PMCID: PMC6780915 DOI: 10.3390/jcm8091418] [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: 08/12/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was to track dental pulp stem cells (DPSCs) labeled with dextran-coated superparamagnetic iron oxide nanoparticles (SPIONs) using magnetic resonance imaging (MRI). Dental pulp was isolated from male Sprague Dawley rats and cultured in Dulbecco's modified Eagle's medium F12 (DMEM-F12) and 10% fetal bovine serum. Effects of SPIONs on morphology, viability, apoptosis, stemness, and osteogenic and adipogenic differentiation of DPSCs were assessed. Prussian blue staining and MRI were conducted to determine in vitro efficiency of SPIONs uptake by the cells. Both non-labeled and labeled DPSCs were adherent to culture plates and showed spindle-shape morphologies, respectively. They were positive for osteogenic and adipogenic induction and expression of cluster of differentiation (CD) 73 and CD90 biomarkers, but negative for expression of CD34 and CD45 biomarkers. The SPIONs were non-toxic and did not induce apoptosis in doses less than 25 mg/mL. Internalization of the SPIONs within the DPSCs was confirmed by Prussian blue staining and MRI. Our findings revealed that the MRI-based method could successfully monitor DPSCs labeled with dextran-coated SPIONs without any significant effect on osteogenic and adipogenic differentiation, viability, and stemness of DPSCs. We provided the in vitro evidence supporting the feasibility of an MRI-based method to monitor DPSCs labeled with SPIONs without any significant reduction in viability, proliferation, and differentiation properties of labeled cells, showing that internalization of SPIONs within DPSCs were not toxic at doses less than 25 mg/mL. In general, the SPION labeling does not seem to impair cell survival or differentiation. SPIONs are biocompatible, easily available, and cost effective, opening a new avenue in stem cell labeling in regenerative medicine.
Collapse
Affiliation(s)
- Shahrokh Zare
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
- Department of Biochemistry, School of Biotechnology and Agriculture, Shiraz Branch, Islamic Azad University, Shiraz, Fars 71987-74731, Iran.
| | - Davood Mehrabani
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71987-74731, Iran.
- Comparative and Experimental Medicine Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| | - Reza Jalli
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
| | - Mahdi Saeedi Moghadam
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
| | - Navid Manafi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Tehran 14348-75451, Iran.
| | - Golshid Mehrabani
- Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA 02118, USA.
| | - Iman Jamhiri
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
| | - Samad Ahadian
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90049, USA.
| |
Collapse
|
13
|
Huang B, Jiang J, Kang M, Liu P, Sun H, Li BG, Wang WJ. Synthesis of block cationic polyacrylamide precursors using an aqueous RAFT dispersion polymerization. RSC Adv 2019; 9:12370-12383. [PMID: 35515873 PMCID: PMC9063656 DOI: 10.1039/c9ra02716e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 11/21/2022] Open
Abstract
Synthesis of cationic polyacrylamides (CPAMs) by introducing cationic polymer precursors followed by chain extension of acrylamide (AM) homopolymer blocks via RAFT polymerization is a promising approach for engineering high-performance CPAMs. However, the aqueous solution polymerization of AM usually leads to high viscosity, thus limiting the solid content in the polymerization system. Herein a novel approach is introduced that uses a random copolymer of AM and methacryloxyethyltrimethyl ammonium chloride (DMC) as a macro RAFT chain transfer agent (mCTA) and stabilizer for aqueous RAFT dispersion polymerization of AM. The AM/DMC random copolymers synthesized by RAFT solution polymerization, having narrow dispersities (Đ s) at different molecular weights and cationic degrees (C s), could serve as the mCTA, which was confirmed by mCTA chain extension in aqueous solution polymerization of AM under different C s, solid contents, AM addition contents, extended PAM block lengths, and mCTA chain lengths. The block CPAMs had a Đ value of less than 1.2. A model was developed using the method of moments with consideration of the diffusion control effect, for further understanding the chain extension kinetics. Predicted polymerization kinetics provided an accurate fit of the experimental data. The AM/DMC random copolymers were further used for aqueous RAFT dispersion polymerization of AM under different polymerization temperatures, C s, and mCTA chain lengths. The resulting products had a milky appearance, and the block copolymers had Đ s of less than 1.3. Higher C s and longer chain lengths on mCTAs were beneficial for stabilizing the polymerization systems and produced smaller particle sizes and less particle aggregation. The products remained stable at room temperature storage for more than a month. The results indicate that aqueous RAFT dispersion polymerization using random copolymers of AM and DMC at moderate cationic degrees as a stabilizer and mCTA is a suitable approach for synthesizing CPAM block precursors at an elevated solid content.
Collapse
Affiliation(s)
- Bo Huang
- State Key Lab of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University 38 Zheda Road Hangzhou 310027 China
| | - Jie Jiang
- State Key Lab of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University 38 Zheda Road Hangzhou 310027 China
| | - Mutian Kang
- State Key Lab of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University 38 Zheda Road Hangzhou 310027 China
| | - Pingwei Liu
- State Key Lab of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University 38 Zheda Road Hangzhou 310027 China .,Institute of Zhejiang University - Quzhou 78 Jiuhua Boulevard North Quzhou China 324000
| | - Hailong Sun
- State Key Laboratory of Hydraulics and Mountain River Engineering, Sichuan University 24 South Section 1, Yihuan Road Chengdu China 610064
| | - Bo-Geng Li
- State Key Lab of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University 38 Zheda Road Hangzhou 310027 China
| | - Wen-Jun Wang
- State Key Lab of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University 38 Zheda Road Hangzhou 310027 China .,Institute of Zhejiang University - Quzhou 78 Jiuhua Boulevard North Quzhou China 324000
| |
Collapse
|
14
|
Alipour M, Nabavi SM, Arab L, Vosough M, Pakdaman H, Ehsani E, Shahpasand K. Stem cell therapy in Alzheimer's disease: possible benefits and limiting drawbacks. Mol Biol Rep 2018; 46:1425-1446. [PMID: 30565076 DOI: 10.1007/s11033-018-4499-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the sixth leading cause of death globally and the main reason for dementia in elderly people. AD is a long-term and progressive neurodegenerative disorder that steadily worsens memory and communicating skills eventually leads to a disabled person of performing simple daily tasks. Unfortunately, numerous clinical trials exploring new therapeutic drugs have encountered disappointing outcomes in terms of improved cognitive performance since they are not capable of halting or stimulating the regeneration of already-damaged neural cells, and merely provide symptomatic relief. Therefore, a deeper understanding of the mechanism of action of stem cell may contribute to the development of novel and effective therapies. The revolutionary discovery of stem cells has cast a new hope for the development of disease-modifying treatments for AD, in terms of their potency in the replenishment of lost cells via differentiating towards specific lineages, stimulating in situ neurogenesis, and delivering the therapeutic agents to the brain. Herein, firstly, we explore the pathophysiology of AD. Next, we summarize the most recent preclinical stem cell reports designed for AD treatment, their benefits and outcomes according to cell type. We briefly review relevant clinical trials and their potential clinical applications in order to find a unique solution to effectively relieve the patients' pain.
Collapse
Affiliation(s)
- Masoume Alipour
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat highway, P.O. Box 19395-4644, Tehran, Iran
| | - Seyed Massood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat highway, P.O. Box 19395-4644, Tehran, Iran
| | - Leila Arab
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat highway, P.O. Box 19395-4644, Tehran, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Pakdaman
- Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Ehsani
- Department of Biology, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat highway, P.O. Box 19395-4644, Tehran, Iran.
| |
Collapse
|
15
|
Azevedo-Pereira RL, Rangel B, Tovar-Moll F, Gasparetto EL, Attias M, Zaverucha-do-Valle C, Jasmin, Mendez-Otero R. Superparamagnetic iron oxide nanoparticles as a tool to track mouse neural stem cells in vivo. Mol Biol Rep 2018; 46:191-198. [PMID: 30421128 DOI: 10.1007/s11033-018-4460-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022]
Abstract
Cell transplantation offers a promising approach in many neurological disorders. Neural stem (NS) cells are potential candidates for cell therapy. The ability to track the grafted cells in the host tissue will refine this therapy. Superparamagnetic iron oxide nanoparticles (SPION) have been suggested as a feasible method, but there is no consensus about its safety. Here we investigated the feasibility of label NS cells with SPION and track by MRI after transplantation into mouse striatum with SPION cells and its therapeutic effects by grafting the cells into mouse striatum. We demonstrated that SPION-labeled NS cells display normal patterns of cellular processes including proliferation, migration, differentiation and neurosphere formation. Transmission electron microscopy reveals SPION in the cytoplasm of the cells, which was confirmed by microanalysis. Neurons and astrocytes generated from SPION-labeled NS cells were able to carry nanoparticles after 7 days under differentiation. SPION-labeled NS cells transplanted into striatum of mice were detected by magnetic resonance imaging (MRI) and microscopy 51 days later. In agreement with others reports, we demonstrated that NS cells are able to incorporate SPION in vitro without altering the stemness, and can survive and be tracked by MRI after they have been grafted into mice striatum.
Collapse
Affiliation(s)
- Ricardo Luiz Azevedo-Pereira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Bárbara Rangel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Marcia Attias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila Zaverucha-do-Valle
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Jasmin
- Núcleo Multidisciplinar de Pesquisa em Biologia-Duque de Caxias, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Abstract
Objective: Gliomas are the most common neoplasm of the central nervous system (CNS); however, traditional imaging techniques do not show the boundaries of tumors well. Some researchers have found a new therapeutic mode to combine nanoparticles, which are nanosized particles with various properties for specific therapeutic purposes, and stem cells for tracing gliomas. This review provides an introduction of the basic understanding and clinical applications of the combination of stem cells and nanoparticles as a contrast agent for glioma imaging. Data Sources: Studies published in English up to and including 2017 were extracted from the PubMed database with the selected key words of “stem cell,” “glioma,” “nanoparticles,” “MRI,” “nuclear imaging,” and “Fluorescence imaging.” Study Selection: The selection of studies focused on both preclinical studies and basic studies of tracking glioma with nanoparticle-labeled stem cells. Results: Studies have demonstrated successful labeling of stem cells with multiple types of nanoparticles. These labeled stem cells efficiently migrated to gliomas of varies models and produced signals sensitively captured by different imaging modalities. Conclusion: The use of nanoparticle-labeled stem cells is a promising imaging platform for the tracking and treatment of gliomas.
Collapse
Affiliation(s)
- Shuang-Lin Deng
- Department of Neurosurgical Oncology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yun-Qian Li
- Department of Neurosurgical Oncology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Gang Zhao
- Department of Neurosurgical Oncology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
17
|
Liu XY, Zhou CB, Fang C. Nanomaterial-involved neural stem cell research: Disease treatment, cell labeling, and growth regulation. Biomed Pharmacother 2018; 107:583-597. [PMID: 30114642 DOI: 10.1016/j.biopha.2018.08.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) have been widely investigated for their potential in the treatment of various diseases and transplantation therapy. However, NSC growth regulation, labeling, and its application to disease diagnosis and treatment are outstanding challenges. Recently, nanomaterials have shown promise for various applications including genetic modification, imaging, and controlled drug release. Here we summarize the recent progress in the use of nanomaterials in combination with NSCs for disease treatment and diagnosis, cell labeling, and NSC growth regulation. The toxicity of nanomaterials to NSCs is also discussed.
Collapse
Affiliation(s)
- Xiang-Yu Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Cheng-Bin Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240 China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
18
|
Liu Y, Li J, Xu K, Gu J, Huang L, Zhang L, Liu N, Kong J, Xing M, Zhang L, Zhang L. Characterization of superparamagnetic iron oxide nanoparticle-induced apoptosis in PC12 cells and mouse hippocampus and striatum. Toxicol Lett 2018; 292:151-161. [PMID: 29715513 DOI: 10.1016/j.toxlet.2018.04.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/30/2018] [Accepted: 04/26/2018] [Indexed: 12/15/2022]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been widely used as theranostic drug-carrier and MRI contrast agent. Their potential effects are still in blank while SPIONs are used for brain. The present study aims to investigate SPIONs' neurotoxicity in vitro and in vivo using stereotaxic technique. By co-incubating SPIONs with dopaminergic neuronal PC12 cells, we found that SPIONs had a dose-dependent cytotoxic in PC12 cells at 60-200 ug/mL but not at 10-50 ug/mL, it reduced cell viability, decreased the capacity of PC12 cells to extend neurites in response to nerve growth factor (NGF), induced a reduction of the tyrosine hydroxylase protein, while increasing PC12 cell apoptosis. Accordingly, the no-observed-adverse-effect level (NOAEL) of current SPIONs was 50 ug/mL in vitro, which would be useful for human health risk assessment. While directly injecting the SPIONs into the dorsal striatum or hippocampus, 7 and 14 days after surgery, nanoparticles decreased the TH+ fiber density in both the dorsal striatum and the hippocampus. A behavioral evaluation demonstrated that SPIONs attenuated the animals' motor coordination and spatial memory, as evaluated by the rotarod test and the Morris water maze. We further examined mitogen-activated protein kinase (MAPK) activation and found that c-Jun N-terminal kinase (JNK) was activated after SPIONs treatment. It suggests that the SPIONs-induced neurotoxicity might be mediated through the JNK signaling pathway. SPIONs could possibly induce neurotoxic effects on the dorsal striatum and hippocampus.
Collapse
Affiliation(s)
- Yutong Liu
- Guangdong Provincial Key Laboratory of Proteomics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Juan Li
- Guangdong Provincial Key Laboratory of Proteomics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Kaige Xu
- Department of Mechanical Engineering, Biochemistry and Medical Genetics, University of Manitoba, Manitoba Institute of Child Health, Winnipeg, MB R3T 2N2, Canada
| | - Jingjing Gu
- Guangdong Provincial Key Laboratory of Proteomics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lu Huang
- Guangdong Provincial Key Laboratory of Proteomics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lei Zhang
- Guangdong Provincial Key Laboratory of Proteomics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - N Liu
- Elderly Health Services Research Center, Southern Medical University, Guangzhou, 510515, China
| | - Jiming Kong
- Southern Medical University-University of Manitoba Geriatric Medicine Joint Laboratory, Canada
| | - Malcolm Xing
- Department of Mechanical Engineering, Biochemistry and Medical Genetics, University of Manitoba, Manitoba Institute of Child Health, Winnipeg, MB R3T 2N2, Canada.
| | - Lin Zhang
- Guangdong Provincial Key Laboratory of Proteomics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Proteomics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China; Elderly Health Services Research Center, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
19
|
Pham BTT, Colvin EK, Pham NTH, Kim BJ, Fuller ES, Moon EA, Barbey R, Yuen S, Rickman BH, Bryce NS, Bickley S, Tanudji M, Jones SK, Howell VM, Hawkett BS. Biodistribution and Clearance of Stable Superparamagnetic Maghemite Iron Oxide Nanoparticles in Mice Following Intraperitoneal Administration. Int J Mol Sci 2018; 19:E205. [PMID: 29320407 PMCID: PMC5796154 DOI: 10.3390/ijms19010205] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/17/2017] [Accepted: 12/27/2017] [Indexed: 12/21/2022] Open
Abstract
Nanomedicine is an emerging field with great potential in disease theranostics. We generated sterically stabilized superparamagnetic iron oxide nanoparticles (s-SPIONs) with average core diameters of 10 and 25 nm and determined the in vivo biodistribution and clearance profiles. Healthy nude mice underwent an intraperitoneal injection of these s-SPIONs at a dose of 90 mg Fe/kg body weight. Tissue iron biodistribution was monitored by atomic absorption spectroscopy and Prussian blue staining. Histopathological examination was performed to assess tissue toxicity. The 10 nm s-SPIONs resulted in higher tissue-iron levels, whereas the 25 nm s-SPIONs peaked earlier and cleared faster. Increased iron levels were detected in all organs and body fluids tested except for the brain, with notable increases in the liver, spleen, and the omentum. The tissue-iron returned to control or near control levels within 7 days post-injection, except in the omentum, which had the largest and most variable accumulation of s-SPIONs. No obvious tissue changes were noted although an influx of macrophages was observed in several tissues suggesting their involvement in s-SPION sequestration and clearance. These results demonstrate that the s-SPIONs do not degrade or aggregate in vivo and intraperitoneal administration is well tolerated, with a broad and transient biodistribution. In an ovarian tumor model, s-SPIONs were shown to accumulate in the tumors, highlighting their potential use as a chemotherapy delivery agent.
Collapse
Affiliation(s)
- Binh T T Pham
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia.
| | - Emily K Colvin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW 2065, Australia.
- Sydney Medical School-Northern, University of Sydney, Sydney, NSW 2006, Australia.
| | - Nguyen T H Pham
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia.
| | - Byung J Kim
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia.
| | - Emily S Fuller
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW 2065, Australia.
- Sydney Medical School-Northern, University of Sydney, Sydney, NSW 2006, Australia.
| | - Elizabeth A Moon
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW 2065, Australia.
| | - Raphael Barbey
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia.
| | - Samuel Yuen
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW 2065, Australia.
| | - Barry H Rickman
- Sydney School of Veterinary Science, University of Sydney Teaching Hospital Camden, Camden, NSW 2570, Australia.
| | - Nicole S Bryce
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia.
| | | | - Marcel Tanudji
- Sirtex Medical Limited, North Sydney, NSW 2060, Australia.
| | | | - Viive M Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW 2065, Australia.
- Sydney Medical School-Northern, University of Sydney, Sydney, NSW 2006, Australia.
| | - Brian S Hawkett
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
20
|
Kumar A, Tan A, Wong J, Spagnoli JC, Lam J, Blevins BD, G N, Thorne L, Ashkan K, Xie J, Liu H. Nanotechnology for Neuroscience: Promising Approaches for Diagnostics, Therapeutics and Brain Activity Mapping. ADVANCED FUNCTIONAL MATERIALS 2017; 27:1700489. [PMID: 30853878 PMCID: PMC6404766 DOI: 10.1002/adfm.201700489] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Unlocking the secrets of the brain is a task fraught with complexity and challenge - not least due to the intricacy of the circuits involved. With advancements in the scale and precision of scientific technologies, we are increasingly equipped to explore how these components interact to produce a vast range of outputs that constitute function and disease. Here, an insight is offered into key areas in which the marriage of neuroscience and nanotechnology has revolutionized the industry. The evolution of ever more sophisticated nanomaterials culminates in network-operant functionalized agents. In turn, these materials contribute to novel diagnostic and therapeutic strategies, including drug delivery, neuroprotection, neural regeneration, neuroimaging and neurosurgery. Further, the entrance of nanotechnology into future research arenas including optogenetics, molecular/ion sensing and monitoring, and piezoelectric effects is discussed. Finally, considerations in nanoneurotoxicity, the main barrier to clinical translation, are reviewed, and direction for future perspectives is provided.
Collapse
Affiliation(s)
- Anil Kumar
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China
| | - Aaron Tan
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Joanna Wong
- Imperial College School of Medicine, Imperial College London,London, United Kingdom
| | - Jonathan Clayton Spagnoli
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - James Lam
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Brianna Diane Blevins
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Natasha G
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Lewis Thorne
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Keyoumars Ashkan
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, King's College London, London, United Kingdom
| | - Jin Xie
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China
| |
Collapse
|
21
|
Huang L, Xia B, Liu Z, Cao Q, Huang J, Luo Z. Superparamagnetic Iron Oxide Nanoparticle-Mediated Forces Enhance the Migration of Schwann Cells Across the Astrocyte-Schwann Cell Boundary In vitro. Front Cell Neurosci 2017; 11:83. [PMID: 28400720 PMCID: PMC5368970 DOI: 10.3389/fncel.2017.00083] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/10/2017] [Indexed: 12/20/2022] Open
Abstract
Schwann cells (SCs) are one of the most promising cellular candidates for the treatment of spinal cord injury. However, SCs show poor migratory ability within the astrocyte-rich central nervous system (CNS) environment and exhibit only limited integration with host astrocytes. Our strategy for improving the therapeutic potential of SCs was to magnetically drive SCs to migrate across the astrocyte-SC boundary to intermingle with astrocytes. SCs were firstly magnetized with poly-L-lysine-coated superparamagnetic iron oxide nanoparticles (SPIONs). Internalization of SPIONs showed no effect upon the migration of SCs in the absence of a magnetic field (MF). In contrast, magnetized SCs exhibited enhanced migration along the direction of force in the presence of a MF. An inverted coverslip assay showed that a greater number of magnetized SCs migrated longer distances onto astrocytic monolayers under the force of a MF compared to other test groups. More importantly, a confrontation assay demonstrated that magnetized SCs intermingled with astrocytes under an applied MF. Furthermore, inhibition of integrin activation reduced the migration of magnetized SCs within an astrocyte-rich environment under an applied MF. Thus, SPION-mediated forces could act as powerful stimulants to enhance the migration of SCs across the astrocyte-SC boundary, via integrin-mediated mechanotransduction, and could represent a vital way of improving the therapeutic potential of SCs for spinal cord injuries.
Collapse
Affiliation(s)
- Liangliang Huang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Bing Xia
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Zhongyang Liu
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Quanliang Cao
- State Key Laboratory of Advanced Electromagnetic Engineering and Technology, Wuhan National High Magnetic Field Center, Huazhong University of Science and Technology Wuhan, China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| |
Collapse
|
22
|
Pham THN, Lengkeek NA, Greguric I, Kim BJ, Pellegrini PA, Bickley SA, Tanudji MR, Jones SK, Hawkett BS, Pham BTT. Tunable and noncytotoxic PET/SPECT-MRI multimodality imaging probes using colloidally stable ligand-free superparamagnetic iron oxide nanoparticles. Int J Nanomedicine 2017; 12:899-909. [PMID: 28184160 PMCID: PMC5291326 DOI: 10.2147/ijn.s127171] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Physiologically stable multimodality imaging probes for positron emission tomography/single-photon emission computed tomography (PET/SPECT)-magnetic resonance imaging (MRI) were synthesized using the superparamagnetic maghemite iron oxide (γ-Fe2O3) nanoparticles (SPIONs). The SPIONs were sterically stabilized with a finely tuned mixture of diblock copolymers with either methoxypolyethylene glycol (MPEG) or primary amine NH2 end groups. The radioisotope for PET or SPECT imaging was incorporated with the SPIONs at high temperature. 57Co2+ ions with a long half-life of 270.9 days were used as a model for the radiotracer to study the kinetics of radiolabeling, characterization, and the stability of the radiolabeled SPIONs. Radioactive 67Ga3+ and Cu2+-labeled SPIONs were also produced successfully using the optimized conditions from the 57Co2+-labeling process. No free radioisotopes were detected in the aqueous phase for the radiolabeled SPIONs 1 week after dispersion in phosphate-buffered saline (PBS). All labeled SPIONs were not only well dispersed and stable under physiological conditions but also noncytotoxic in vitro. The ability to design and produce physiologically stable radiolabeled magnetic nanoparticles with a finely controlled number of functionalizable end groups on the SPIONs enables the generation of a desirable and biologically compatible multimodality PET/SPECT-MRI agent on a single T2 contrast MRI probe.
Collapse
Affiliation(s)
- TH Nguyen Pham
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney
| | - Nigel A Lengkeek
- Radioisotopes and Radiotracers, NSTLI, Australian Nuclear Science and Technology Organisation, Sydney
| | - Ivan Greguric
- Radioisotopes and Radiotracers, NSTLI, Australian Nuclear Science and Technology Organisation, Sydney
| | - Byung J Kim
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney
| | - Paul A Pellegrini
- Radioisotopes and Radiotracers, NSTLI, Australian Nuclear Science and Technology Organisation, Sydney
| | | | | | | | - Brian S Hawkett
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney
| | - Binh TT Pham
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney
| |
Collapse
|
23
|
Guldris N, Argibay B, Gallo J, Iglesias-Rey R, Carbó-Argibay E, Kolen'ko YV, Campos F, Sobrino T, Salonen LM, Bañobre-López M, Castillo J, Rivas J. Magnetite Nanoparticles for Stem Cell Labeling with High Efficiency and Long-Term in Vivo Tracking. Bioconjug Chem 2016; 28:362-370. [PMID: 27977143 DOI: 10.1021/acs.bioconjchem.6b00522] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIO-PAA), ultrasmall iron oxide nanoparticles (USPIO-PAA), and glucosamine-modified iron oxide nanoparticles (USPIO-PAA-GlcN) were studied as mesenchymal stem cell (MSCs) labels for cell tracking applications by magnetic resonance imaging (MRI). Pronounced differences were found in the labeling performance of the three samples in terms of cellular dose and labeling efficiency. In combination with polylysine, SPIO-PAA showed nonhomogeneous cell internalization, while for USPIO-PAA no uptake was found. On the contrary, USPIO-PAA-GlcN featured high cellular uptake and biocompatibility, and sensitive detection in both in vitro and in vivo experiments was found by MRI, showing that glucosamine functionalization can be an efficient strategy to increase cell uptake of ultrasmall iron oxide nanoparticles by MSCs.
Collapse
Affiliation(s)
- Noelia Guldris
- International Iberian Nanotechnology Laboratory , Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | | | - Juan Gallo
- International Iberian Nanotechnology Laboratory , Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | | | - Enrique Carbó-Argibay
- International Iberian Nanotechnology Laboratory , Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Yury V Kolen'ko
- International Iberian Nanotechnology Laboratory , Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | | | | | - Laura M Salonen
- International Iberian Nanotechnology Laboratory , Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Manuel Bañobre-López
- International Iberian Nanotechnology Laboratory , Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | | | - José Rivas
- International Iberian Nanotechnology Laboratory , Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| |
Collapse
|
24
|
Nguyen D, Pham BT, Huynh V, Kim BJ, Pham NT, Bickley SA, Jones SK, Serelis A, Davey T, Such C, Hawkett BS. Monodispersed polymer encapsulated superparamagnetic iron oxide nanoparticles for cell labeling. POLYMER 2016. [DOI: 10.1016/j.polymer.2016.08.064] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Jiráková K, Šeneklová M, Jirák D, Turnovcová K, Vosmanská M, Babič M, Horák D, Veverka P, Jendelová P. The effect of magnetic nanoparticles on neuronal differentiation of induced pluripotent stem cell-derived neural precursors. Int J Nanomedicine 2016; 11:6267-6281. [PMID: 27920532 PMCID: PMC5125991 DOI: 10.2147/ijn.s116171] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Introduction Magnetic resonance (MR) imaging is suitable for noninvasive long-term tracking. We labeled human induced pluripotent stem cell-derived neural precursors (iPSC-NPs) with two types of iron-based nanoparticles, silica-coated cobalt zinc ferrite nanoparticles (CZF) and poly-l-lysine-coated iron oxide superparamagnetic nanoparticles (PLL-coated γ-Fe2O3) and studied their effect on proliferation and neuronal differentiation. Materials and methods We investigated the effect of these two contrast agents on neural precursor cell proliferation and differentiation capability. We further defined the intracellular localization and labeling efficiency and analyzed labeled cells by MR. Results Cell proliferation was not affected by PLL-coated γ-Fe2O3 but was slowed down in cells labeled with CZF. Labeling efficiency, iron content and relaxation rates measured by MR were lower in cells labeled with CZF when compared to PLL-coated γ-Fe2O3. Cytoplasmic localization of both types of nanoparticles was confirmed by transmission electron microscopy. Flow cytometry and immunocytochemical analysis of specific markers expressed during neuronal differentiation did not show any significant differences between unlabeled cells or cells labeled with both magnetic nanoparticles. Conclusion Our results show that cells labeled with PLL-coated γ-Fe2O3 are suitable for MR detection, did not affect the differentiation potential of iPSC-NPs and are suitable for in vivo cell therapies in experimental models of central nervous system disorders.
Collapse
Affiliation(s)
- Klára Jiráková
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic
| | - Monika Šeneklová
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Department of Neuroscience, Second Faculty of Medicine, Charles University
| | - Daniel Jirák
- MR-Unit, Radiodiagnostic and Interventional Radiology Department, Institute for Clinical and Experimental Medicine; Department of Biophysics, Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University
| | - Karolína Turnovcová
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic
| | - Magda Vosmanská
- Department of Analytical Chemistry, University of Chemistry and Technology
| | - Michal Babič
- Department of Polymer Particles, Institute of Macromolecular Chemistry
| | - Daniel Horák
- Department of Polymer Particles, Institute of Macromolecular Chemistry
| | - Pavel Veverka
- Department of Magnetics and Superconductors, Institute of Physics, ASCR, Prague, Czech Republic
| | - Pavla Jendelová
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Department of Neuroscience, Second Faculty of Medicine, Charles University
| |
Collapse
|
26
|
Ramos-Gómez M, Martínez-Serrano A. Tracking of iron-labeled human neural stem cells by magnetic resonance imaging in cell replacement therapy for Parkinson's disease. Neural Regen Res 2016; 11:49-52. [PMID: 26981077 PMCID: PMC4774222 DOI: 10.4103/1673-5374.169628] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human neural stem cells (hNSCs) derived from the ventral mesencephalon are powerful research tools and candidates for cell therapies in Parkinson's disease. However, their clinical translation has not been fully realized due, in part, to the limited ability to track stem cell regional localization and survival over long periods of time after in vivo transplantation. Magnetic resonance imaging provides an excellent non-invasive method to study the fate of transplanted cells in vivo. For magnetic resonance imaging cell tracking, cells need to be labeled with a contrast agent, such as magnetic nanoparticles, at a concentration high enough to be easily detected by magnetic resonance imaging. Grafting of human neural stem cells labeled with magnetic nanoparticles allows cell tracking by magnetic resonance imaging without impairment of cell survival, proliferation, self-renewal, and multipotency. However, the results reviewed here suggest that in long term grafting, activated microglia and macrophages could contribute to magnetic resonance imaging signal by engulfing dead labeled cells or iron nanoparticles dispersed freely in the brain parenchyma over time.
Collapse
Affiliation(s)
- Milagros Ramos-Gómez
- Centre for Biomedical Technology, Polytechnic University of Madrid, Madrid, Spain; Biomedical Research Networking Center of Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Alberto Martínez-Serrano
- Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa", Autonomous University of Madrid-C.S.I.C, Madrid, Spain
| |
Collapse
|
27
|
Cao FY, Fan JX, Long Y, Zeng X, Zhang XZ. A smart fluorescence nanoprobe for the detection of cellular alkaline phosphatase activity and early osteogenic differentiation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1313-22. [PMID: 26961462 DOI: 10.1016/j.nano.2016.01.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 12/13/2015] [Accepted: 01/21/2016] [Indexed: 01/22/2023]
Abstract
In the past decades, biomaterials were designed to induce stem cell toward osteogenic differentiation. However, conventional methods for evaluation osteogenic differentiation all required a process of cell fixation or lysis, which induce waste of a large number of cells. In this study, a fluorescence nanoprobe was synthesized by combining phosphorylated fluoresceinamine isomer I (FLA) on the surface of mesoporous silica-coated superparamagnetic iron oxide (Fe3O4@mSiO2) nanoparticles. In the presence of alkaline phosphatase (ALP), the phosphorylated FLA on the nanoprobe would be hydrolyzed, resulting in a fluorescence recovery of FLA. During early osteogenic differentiation, a high-level expression of cellular ALP was induced, which accelerated the hydrolysis of phosphorylated FLA, resulting in an enhancement of cellular fluorescence intensity. This fluorescence nanoprobe provides us a rapid and non-toxic method for the detection of cellular ALP activity and early osteogenic differentiation.
Collapse
Affiliation(s)
- Feng-Yi Cao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, China
| | - Jin-Xuan Fan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, China
| | - Yue Long
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, China.
| |
Collapse
|
28
|
Visualizing myocardial inflammation in a rat model of type 4 cardiorenal syndrome by dual-modality molecular imaging. Biomaterials 2015; 68:67-76. [DOI: 10.1016/j.biomaterials.2015.07.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 07/27/2015] [Accepted: 07/31/2015] [Indexed: 12/21/2022]
|
29
|
Pham BTT, Jain N, Kuchel PW, Chapman BE, Bickley SA, Jones SK, Hawkett BS. The interaction of sterically stabilized magnetic nanoparticles with fresh human red blood cells. Int J Nanomedicine 2015; 10:6645-55. [PMID: 26604741 PMCID: PMC4629969 DOI: 10.2147/ijn.s93225] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Sterically stabilized superparamagnetic iron oxide nanoparticles (SPIONs) were incubated with fresh human erythrocytes (red blood cells [RBCs]) to explore their potential application as magnetic resonance imaging contrast agents. The chemical shift and linewidth of (133)Cs(+) resonances from inside and outside the RBCs in (133)Cs nuclear magnetic resonance spectra were monitored as a function of time. Thus, we investigated whether SPIONs of two different core sizes and with three different types of polymeric stabilizers entered metabolically active RBCs, consuming glucose at 37°C. The SPIONs broadened the extracellular (133)Cs(+) nuclear magnetic resonance, and brought about a small change in its chemical shift to a higher frequency; while the intracellular resonance remained unchanged in both amplitude and chemical shift. This situation pertained over incubation times of up to 90 minutes. If the SPIONs had entered the RBCs, the intracellular resonance would have become broader and possibly even shifted. Therefore, we concluded that our SPIONs did not enter the RBCs. In addition, the T 2 relaxivity of the small and large particles was 368 and 953 mM(-1) s(-1), respectively (three and nine times that of the most effective commercially available samples). This suggests that these new SPIONs will provide a superior performance to any others reported thus far as magnetic resonance imaging contrast agents.
Collapse
Affiliation(s)
- Binh TT Pham
- School of Chemistry, University of Sydney, NSW, Australia
| | - Nirmesh Jain
- School of Chemistry, University of Sydney, NSW, Australia
| | - Philip W Kuchel
- School of Molecular Bioscience, University of Sydney, NSW, Australia
| | - Bogdan E Chapman
- School of Molecular Bioscience, University of Sydney, NSW, Australia
| | | | | | | |
Collapse
|
30
|
Korchinski DJ, Taha M, Yang R, Nathoo N, Dunn JF. Iron Oxide as an MRI Contrast Agent for Cell Tracking. MAGNETIC RESONANCE INSIGHTS 2015; 8:15-29. [PMID: 26483609 PMCID: PMC4597836 DOI: 10.4137/mri.s23557] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/17/2015] [Accepted: 08/19/2015] [Indexed: 01/07/2023]
Abstract
Iron oxide contrast agents have been combined with magnetic resonance imaging for cell tracking. In this review, we discuss coating properties and provide an overview of ex vivo and in vivo labeling of different cell types, including stem cells, red blood cells, and monocytes/macrophages. Furthermore, we provide examples of applications of cell tracking with iron contrast agents in stroke, multiple sclerosis, cancer, arteriovenous malformations, and aortic and cerebral aneurysms. Attempts at quantifying iron oxide concentrations and other vascular properties are examined. We advise on designing studies using iron contrast agents including methods for validation.
Collapse
Affiliation(s)
- Daniel J. Korchinski
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - May Taha
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Runze Yang
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nabeela Nathoo
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeff F. Dunn
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Experimental Imaging Centre, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,CORRESPONDENCE:
| |
Collapse
|
31
|
Palma SICJ, Rodrigues CAV, Carvalho A, Morales MDP, Freitas F, Fernandes AR, Cabral JMS, Roque ACA. A value-added exopolysaccharide as a coating agent for MRI nanoprobes. NANOSCALE 2015; 7:14272-14283. [PMID: 26186402 DOI: 10.1039/c5nr01979f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Fucopol, a fucose-containing exopolysaccharide (EPS) produced by the bacterium Enterobacter A47 DSM 23139 using glycerol as a carbon source, was employed as a new coating material for iron oxide magnetic nanoparticles (MNPs). The coated particles were assessed as nanoprobes for cell labeling by Magnetic Resonance Imaging (MRI). The MNPs were synthesized by a thermal decomposition method and transferred to an aqueous medium by a ligand-exchange reaction with meso-2,3-dimercaptosuccinic acid (DMSA). Covalent binding of EPS to DMSA-stabilized nanoparticles (MNP-DMSA) resulted in a hybrid magnetic-biopolymeric nanosystem (MNP-DMSA-EPS) with a hydrodynamic size of 170 nm, a negative surface charge under physiological conditions and transverse to longitudinal relaxivity ratio, r2/r1, of 148. In vitro studies with two human cell lines (colorectal carcinoma - HCT116 - and neural stem/progenitor cells - ReNcell VM) showed that EPS promotes internalization of nanoparticles in both cell lines. In vitro MRI cell phantoms showed a superior performance of MNP-DMSA-EPS in ReNcell VM, for which the iron dose-dependent MRI signal drop was obtained at relatively low iron concentrations (12-20 μg Fe per ml) and short incubation times. Furthermore, ReNcell VM multipotency was not affected by culture in the presence of MNP-DMSA or MNP-DMSA-EPS for 14 days. Our study suggests that Fucopol-coated MNPs represent useful cell labeling nanoprobes for MRI.
Collapse
Affiliation(s)
- Susana I C J Palma
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
MRI tracking of bone marrow mesenchymal stem cells labeled with ultra-small superparamagnetic iron oxide nanoparticles in a rat model of temporal lobe epilepsy. Neurosci Lett 2015; 606:30-5. [PMID: 26318841 DOI: 10.1016/j.neulet.2015.08.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/21/2015] [Accepted: 08/21/2015] [Indexed: 12/25/2022]
Abstract
Transplantation of bone marrow mesenchymal stem cells (BMSCs) is a promising approach for treatment of epilepsy. To our knowledge, there is little research on magnetic resonance imaging (MRI) tracking of BMSCs labeled with ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles in a rat model of temporal lobe epilepsy (TLE). In this study, BMSCs were pre-labeled with USPIO nanoparticles, and then the cell apoptosis, proliferation, surface antigens, and multipotency were investigated. Lithium chloride-pilocarpine induced TLE models were administered by USPIO-labeled BMSCs (U-BMSCs), BMSCs, and saline through lateral ventricle injection as the experimental group, control I group and control II group, respectively, followed by MRI examination, electroencephalography (EEG) and Prussian blue staining. The cell experimental results showed that the labeled USPIO did not affect the biological characteristics and multiple potential of BMSCs. The U-BMSCs can be detected using MRI in vitro and in vivo, and observed in the hippocampus and adjacent parahippocampal cortical areas of the epileptic model. Moreover, electroencephalographic results showed that transplanted U-BMSCs, as well as BMSCs, were capable of reducing the number of epileptiform waves significantly (P<0.01) compared with control II group. All of these findings suggest that it is feasible to track transplanted BMSCs using MRI in a rat model of TLE, and support that USPIO labeling is a valuable tool for cell tracking in the study of seizure disorders.
Collapse
|
33
|
Aswendt M, Henn N, Michalk S, Schneider G, Steiner MS, Bissa U, Dose C, Hoehn M. Novel bimodal iron oxide particles for efficient tracking of human neural stem cells in vivo. Nanomedicine (Lond) 2015; 10:2499-512. [PMID: 26296195 DOI: 10.2217/nnm.15.94] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
AIMS We validated novel bimodal iron oxide particles as substitute of ferumoxides for efficient labeling of human neural stem cells (NSCs). The dextrane-coated FeraTrack Direct (FTD)-Vio particles have additional far-red fluorophores for microscopic cell analysis. METHODS MR relaxometry, spectrophotometric iron determination and microscopy are used for characterization in vitro and in vivo. RESULTS Efficient uptake is not transfection agent-dependent. FTD-Vio594 labeling had no influence on viability, proliferation, migration and differentiation capacity. It allows MRI-based tracking of engrafted NSCs in mouse brain up to 11 days, complemented by bioluminescence imaging of firefly luciferase expressed by the engrafted cells. CONCLUSION Our results highlight the FTD-Vio594 particles as safe and sensitive substitute of ferumoxides for longitudinal tracking of NSCs in preclinical studies.
Collapse
Affiliation(s)
- Markus Aswendt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany
| | - Nadine Henn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany
| | - Stefanie Michalk
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany
| | - Gabriele Schneider
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany
| | - Mark-Steven Steiner
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany
| | - Ursula Bissa
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany
| | - Christian Dose
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany.,Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands.,Percuros BV, Enschede, Drienerlolaan 5-Zuidhorst, 7522 NB Enschede, The Netherlands
| |
Collapse
|
34
|
Bernsen MR, Guenoun J, van Tiel ST, Krestin GP. Nanoparticles and clinically applicable cell tracking. Br J Radiol 2015; 88:20150375. [PMID: 26248872 DOI: 10.1259/bjr.20150375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In vivo cell tracking has emerged as a much sought after tool for design and monitoring of cell-based treatment strategies. Various techniques are available for pre-clinical animal studies, from which much has been learned and still can be learned. However, there is also a need for clinically translatable techniques. Central to in vivo cell imaging is labelling of cells with agents that can give rise to signals in vivo, that can be detected and measured non-invasively. The current imaging technology of choice for clinical translation is MRI in combination with labelling of cells with magnetic agents. The main challenge encountered during the cell labelling procedure is to efficiently incorporate the label into the cell, such that the labelled cells can be imaged at high sensitivity for prolonged periods of time, without the labelling process affecting the functionality of the cells. In this respect, nanoparticles offer attractive features since their structure and chemical properties can be modified to facilitate cellular incorporation and because they can carry a high payload of the relevant label into cells. While these technologies have already been applied in clinical trials and have increased the understanding of cell-based therapy mechanism, many challenges are still faced.
Collapse
Affiliation(s)
- Monique R Bernsen
- 1 Department of Radiology, Erasmus MC, Rotterdam, Netherlands.,2 Department of Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Jamal Guenoun
- 1 Department of Radiology, Erasmus MC, Rotterdam, Netherlands
| | | | | |
Collapse
|
35
|
Soenen SJ, Parak WJ, Rejman J, Manshian B. (Intra)cellular stability of inorganic nanoparticles: effects on cytotoxicity, particle functionality, and biomedical applications. Chem Rev 2015; 115:2109-35. [PMID: 25757742 DOI: 10.1021/cr400714j] [Citation(s) in RCA: 297] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Stefaan J Soenen
- Biomedical MRI Unit/MoSAIC, Department of Medicine, KULeuven , B3000 Leuven, Belgium
| | | | | | | |
Collapse
|
36
|
Sadhasivam S, Savitha S, Wu CJ, Lin FH, Stobiński L. Carbon encapsulated iron oxide nanoparticles surface engineered with polyethylene glycol-folic acid to induce selective hyperthermia in folate over expressed cancer cells. Int J Pharm 2015; 480:8-14. [PMID: 25601197 DOI: 10.1016/j.ijpharm.2015.01.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/06/2015] [Accepted: 01/16/2015] [Indexed: 11/29/2022]
Abstract
Carbon encapsulated iron oxide nanoparticles (CEIO-NPs) prepared by carbon arc method were successfully applied for in vitro magnetic hyperthermia. The CEIO-NPs were chemically oxidized and surface modified with PEG-FA for selective tumor localization in cancer cells that over expresses the folate receptors (FR(+)). The size, morphology, heating efficiency, biocompatibility and in vitro cell uptake of CEIO-PEG-FA NPs are extensively characterized. The as-prepared nanoparticles have generated quick heating (43-45°C) upon exposure to an alternating magnetic field (AMF) with the saturation magnetization of 25emu/g. The LDH cytotoxic assay demonstrated that the nanoparticle did not affect the viability of normal human fibroblast. The quantitative and cellular uptake studies by TEM confirmed the selective and increased uptake of CEIO-PEG-FA NPs when compared to the CEIO-nanoparticles. In conclusion, CEIO-PEG-FA NPs have the potential to induce magnetic hyperthermia in FR(+) cells via the receptor mediated endocytosis uptake mechanism.
Collapse
Affiliation(s)
- S Sadhasivam
- Biomedical Engineering and Nanomedicine Research, National Health Research Institutes, Taiwan
| | - S Savitha
- Department of Biotechnology, Sree Sastha Institute of Engineering and Technology, Chennai, India
| | - Chun-Jen Wu
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Feng-Huei Lin
- Biomedical Engineering and Nanomedicine Research, National Health Research Institutes, Taiwan; Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.
| | - Leszek Stobiński
- Institute of Physical Chemistry, Polish Academy of Sciences Warsaw, Poland
| |
Collapse
|
37
|
Weinberg D, Adams CF, Chari DM. Deploying clinical grade magnetic nanoparticles with magnetic fields to magnetolabel neural stem cells in adherent versus suspension cultures. RSC Adv 2015. [DOI: 10.1039/c5ra07481a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
This is the first demonstration that oscillating magnetic fields safely promote the uptake of a clinical-grade magnetic nanoparticle (Lumirem/Ferumoxsil) into neural stem cells for non-invasive cell tracking capabilities.
Collapse
Affiliation(s)
- D. Weinberg
- Cellular and Neural Engineering Group
- Institute for Science and Technology in Medicine
- Keele University
- Staffordshire
- UK
| | - C. F. Adams
- Cellular and Neural Engineering Group
- Institute for Science and Technology in Medicine
- Keele University
- Staffordshire
- UK
| | - D. M. Chari
- Cellular and Neural Engineering Group
- Institute for Science and Technology in Medicine
- Keele University
- Staffordshire
- UK
| |
Collapse
|