1
|
Morrison L, Smoody B, Woltjer R, Hinds MT, Loftis JM, Wyatt CW, Nguyen KP. Ferumoxytol-enhanced MRI assessment of venous Thrombus resolution and macrophage content in a murine deep vein thrombosis model. Thromb Res 2024; 240:109063. [PMID: 38878741 PMCID: PMC11239555 DOI: 10.1016/j.thromres.2024.109063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Imaging evaluation of acute deep vein thrombosis (DVT) or post-thrombotic syndrome (PTS) in animal or clinical models is limited to anatomical assessment of the location and extent of thrombi. We hypothesize that Fe-MRI, used to evaluate macrophage content in other inflammatory diseases, can be useful to evaluate the thromboinflammatory features after DVT over time. METHODS Nineteen wild-type CD-1 mice underwent surgical IVC ligation to induce DVT. Mice received either saline or 5 mg/kg of 14E11, a Factor XI inhibitor, before the procedure. Fe-MRI was performed on days 6-7 after ligation to evaluate thrombus volume, perfusion, and macrophage content via T2-weighted images. Mice were euthanized at days 3-15 after surgery. The thrombi and adjacent vein walls were excised, weighed, formalin-fixed, and paraffin-embedded for immunohistological analysis. Specimens were stained with specific antibodies to evaluate macrophage content, collagen deposition, neovascularization, and recanalization. Significance was determined using the Mann-Whitney U or Student's t-test. RESULTS After IVC-ligation in control mice, thrombus weights decreased by 59 % from day 3 to 15. Thrombus volumes peaked on day 5 before decreasing by 85 % by day 13. FXI inhibition led to reduced macrophage content in both thrombi (p = .008) and vein walls (p = .01), decreased thrombus volume (p = .03), and decreased thrombus mass (p = .01) compared to control mice. CCR2+ staining corroborated these findings, showing significantly reduced macrophage presence in the thrombi (p = .002) and vein wall (p = .002). CONCLUSIONS Fe-MRI T2 relaxation times can be used to characterize and quantify post-thrombotic changes of perfusion, macrophage content, and thrombus volume over time in a surgical mouse model of venous thrombosis. This approach could lead to better quantification of in vivo inflammation correlating monocyte and macrophage content within resolving thrombi and veins and may serve as a useful tool for research and clinically in the evaluation of the post-thrombotic environment.
Collapse
Affiliation(s)
- L Morrison
- Department of Surgery, Division of Vascular Surgery, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - B Smoody
- Department of Radiology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Advanced Imaging Research Center (AIRC), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - R Woltjer
- Department of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - M T Hinds
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science Advanced Imaging Research Center (AIRC), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - J M Loftis
- Research and Development, Portland VA Health Care System, 3710 SW US Veterans Highway Road, Portland, OR 97239, USA; Departments of Psychiatry and Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - C W Wyatt
- Department of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - K P Nguyen
- Department of Surgery, Division of Vascular Surgery, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Department of Biomedical Engineering, School of Medicine, Oregon Health & Science Advanced Imaging Research Center (AIRC), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Research and Development, Portland VA Health Care System, 3710 SW US Veterans Highway Road, Portland, OR 97239, USA
| |
Collapse
|
2
|
Nakamura M, Araki H, Tsurushima H, Oyane A. Fabrication of water-dispersible and cell-stimulating calcium phosphate nanoparticles immobilizing basic fibroblast growth factor. Colloids Surf B Biointerfaces 2023; 230:113502. [PMID: 37574618 DOI: 10.1016/j.colsurfb.2023.113502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/15/2023]
Abstract
Basic fibroblast growth factor (bFGF) is a therapeutic protein that can enhance angiogenesis, wound healing, and tissue regeneration; however, it is extremely unstable even under a normal physiological environment. Biocompatible calcium phosphate (CaP) nanoparticles (NPs) co-immobilizing bFGF, heparin, and ferucarbotran would be useful as a multifunctional delivery carrier of bFGF. In this study, such NPs were successfully fabricated by a coprecipitation process, using a labile supersaturated CaP solution containing bFGF, heparin, and ferucarbotran. The NPs showed relatively high negative zeta potential (-12 mV) because of the negatively charged heparin, which enabled their stable dispersion in water. The hydrodynamic diameter of the NPs was around 200 nm. Immunoreactive bFGF was released from the NPs in an acellular medium dose-dependently. The NPs promoted proliferation of baby hamster kidney fibroblasts (BHK-21 cells) and mouse osteoblastic MC3T3-E1 cells at a certain dose range, although they inhibited proliferation of rat pheochromocytoma (PC-12) cells. These results demonstrated that the effect of the NPs on cell proliferation was dependent on the cell type and dose, the details of which should be investigated in a future study.
Collapse
Affiliation(s)
- Maki Nakamura
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan.
| | - Hiroko Araki
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Hideo Tsurushima
- Department of Neurosurgery, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Ayako Oyane
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan.
| |
Collapse
|
3
|
Alves Feitosa K, de Oliveira Correia R, Maragno Fattori AC, Albuquerque YR, Brassolatti P, Flores Luna G, de Almeida Rodolpho JM, T Nogueira C, Cancino Bernardi J, Speglich C, de Freitas Anibal F. Toxicological effects of the mixed iron oxide nanoparticle (Fe 3O 4 NP) on murine fibroblasts LA-9. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2022; 85:649-670. [PMID: 35469539 DOI: 10.1080/15287394.2022.2068711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The increase in large-scale production of magnetic nanoparticles (NP) associated with the incomplete comprehensive knowledge regarding the potential risks of their use on environmental and human health makes it necessary to study the biological effects of these particles on organisms at the cellular level. The aim of this study to examine the cellular effects on fibroblast lineage LA-9 after exposure to mixed iron oxide NP (Fe3O4 NP). The following analyses were performed: field emission gun-scanning electron microscopy (SEM-FEG), dynamic light scattering (DLS), zeta potential, ultraviolet/visible region spectroscopy (UV/VIS), and attenuated total reactance-Fourier transform infrared (ATR-FTIR) spectroscopy analyses for characterization of the NP. The assays included cell viability, morphology, clonogenic potential, oxidative stress as measurement of reactive oxygen species (ROS) and nitric oxide (NO) levels, cytokines quantification interleukin 6 (IL-6) and tumor necrosis factor (TNF), NP uptake, and cell death. The size of Fe3O4 NP was 26.3 nm when evaluated in water through DLS. Fe3O4 NP did not reduce fibroblast cell viability until the highest concentration tested (250 µg/ml), which showed a decrease in clonogenic potential as well as small morphological changes after exposure for 48 and 72 hr. The NP concentration of 250 µg/ml induced enhanced ROS and NO production after 24 hr treatment. The uptake assay exhibited time-dependent Fe3O4 NP internalization at all concentrations tested with no significant cell death. Hence, exposure of fibroblasts to Fe3O4 NP-induced oxidative stress but not reduced cell viability or death. However, the decrease in the clonogenic potential at the highest concentration demonstrates cytotoxic effects attributed to Fe3O4 NP which occurred on the 7th day after exposure.
Collapse
Affiliation(s)
- Karina Alves Feitosa
- Department of Morphology and Pathology, Inflammation and Infectious Diseases Laboratory, Federal University of São Carlos, São Carlos, Brazil
| | - Ricardo de Oliveira Correia
- Department of Morphology and Pathology, Inflammation and Infectious Diseases Laboratory, Federal University of São Carlos, São Carlos, Brazil
| | - Ana Carolina Maragno Fattori
- Department of Morphology and Pathology, Inflammation and Infectious Diseases Laboratory, Federal University of São Carlos, São Carlos, Brazil
| | - Yulli Roxenne Albuquerque
- Department of Morphology and Pathology, Inflammation and Infectious Diseases Laboratory, Federal University of São Carlos, São Carlos, Brazil
| | - Patricia Brassolatti
- Department of Morphology and Pathology, Inflammation and Infectious Diseases Laboratory, Federal University of São Carlos, São Carlos, Brazil
| | - Genoveva Flores Luna
- Department of Morphology and Pathology, Inflammation and Infectious Diseases Laboratory, Federal University of São Carlos, São Carlos, Brazil
| | - Joice Margareth de Almeida Rodolpho
- Department of Morphology and Pathology, Inflammation and Infectious Diseases Laboratory, Federal University of São Carlos, São Carlos, Brazil
| | | | - Juliana Cancino Bernardi
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Carlos Speglich
- Leopoldo Américo Miguez de Mello Research Center CENPES/Petrobras, Rio de Janeiro, Brazil
| | - Fernanda de Freitas Anibal
- Department of Morphology and Pathology, Inflammation and Infectious Diseases Laboratory, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
4
|
Lu CH, Hsiao JK. Diagnostic and therapeutic roles of iron oxide nanoparticles in biomedicine. Tzu Chi Med J 2022; 35:11-17. [PMID: 36866343 PMCID: PMC9972926 DOI: 10.4103/tcmj.tcmj_65_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/03/2022] [Accepted: 06/08/2022] [Indexed: 11/04/2022] Open
Abstract
Nanotechnology changed our understanding of physics and chemics and influenced the biomedical field. Iron oxide nanoparticles (IONs) are one of the first emerging biomedical applications of nanotechnology. The IONs are composed of iron oxide core exhibiting magnetism and coated with biocompatible molecules. The small size, strong magnetism, and biocompatibility of IONs facilitate the application of IONs in the medical imaging field. We listed several clinical available IONs including Resovist (Bayer Schering Pharma, Berlin, Germany) and Feridex intravenous (I.V.)/Endorem as magnetic resonance (MR) contrast agents for liver tumor detection. We also illustrated GastroMARK as a gastrointestinal contrast agent for MR imaging. Recently, IONs named Feraheme for treating iron-deficiency anemia have been approved by the Food and Drug Administration. Moreover, tumor ablation by IONs named NanoTherm has also been discussed. In addition to the clinical application, several potential biomedical applications of IONs including cancer-targeting capability by conjugating IONs with cancer-specific ligands, cell trafficking tools, or tumor ablation agents have also been discussed. With the growing awareness of nanotechnology, further application of IONs is still on the horizon that would shed light on biomedicine.
Collapse
Affiliation(s)
- Chia-Hung Lu
- Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan,School of Medicine, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Jong-Kia Hsiao, Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 289, Jianguo Road, Xindian District, New Taipei, Taiwan. E-mail:
| |
Collapse
|
5
|
Pavlin M, Lojk J, Strojan K, Hafner-Bratkovič I, Jerala R, Leonardi A, Križaj I, Drnovšek N, Novak S, Veranič P, Bregar VB. The Relevance of Physico-Chemical Properties and Protein Corona for Evaluation of Nanoparticles Immunotoxicity-In Vitro Correlation Analysis on THP-1 Macrophages. Int J Mol Sci 2022; 23:6197. [PMID: 35682872 PMCID: PMC9181693 DOI: 10.3390/ijms23116197] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Alongside physiochemical properties (PCP), it has been suggested that the protein corona of nanoparticles (NPs) plays a crucial role in the response of immune cells to NPs. However, due to the great variety of NPs, target cells, and exposure protocols, there is still no clear relationship between PCP, protein corona composition, and the immunotoxicity of NPs. In this study, we correlated PCP and the protein corona composition of NPs to the THP-1 macrophage response, focusing on selected toxicological endpoints: cell viability, reactive oxygen species (ROS), and cytokine secretion. We analyzed seven commonly used engineered NPs (SiO2, silver, and TiO2) and magnetic NPs. We show that with the exception of silver NPs, all of the tested TiO2 types and SiO2 exhibited moderate toxicities and a transient inflammatory response that was observed as an increase in ROS, IL-8, and/or IL-1β cytokine secretion. We observed a strong correlation between the size of the NPs in media and IL-1β secretion. The induction of IL-1β secretion was completely blunted in NLR family pyrin domain containing 3 (NLRP3) knockout THP-1 cells, indicating activation of the inflammasome. The correlations analysis also implicated the association of specific NP corona proteins with the induction of cytokine secretion. This study provides new insights toward a better understanding of the relationships between PCP, protein corona, and the inflammatory response of macrophages for different engineered NPs, to which we are exposed on a daily basis.
Collapse
Grants
- J7-7424, J2-6758, J3-1746, J3-6794, J3-7494, Z4-8229, P1-0055, P3-0108, P1-0207, P4-0220, P2-0087, P4-0176, young researchers program and MRIC UL IP-0510 Infrastructure program Slovenian Research Agency
- ISO-FOOD (FP7-REGPOT) European Commission
Collapse
Affiliation(s)
- Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
- Group for Nano and Biotechnological Application, Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, SI-1000 Ljubljana, Slovenia; (J.L.); (K.S.); (V.B.B.)
| | - Jasna Lojk
- Group for Nano and Biotechnological Application, Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, SI-1000 Ljubljana, Slovenia; (J.L.); (K.S.); (V.B.B.)
| | - Klemen Strojan
- Group for Nano and Biotechnological Application, Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, SI-1000 Ljubljana, Slovenia; (J.L.); (K.S.); (V.B.B.)
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (I.H.-B.); (R.J.)
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, SI-1000 Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia; (I.H.-B.); (R.J.)
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, SI-1000 Ljubljana, Slovenia
| | - Adrijana Leonardi
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (A.L.); (I.K.)
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (A.L.); (I.K.)
| | - Nataša Drnovšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (N.D.); (S.N.)
| | - Saša Novak
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; (N.D.); (S.N.)
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia;
| | - Vladimir Boštjan Bregar
- Group for Nano and Biotechnological Application, Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, SI-1000 Ljubljana, Slovenia; (J.L.); (K.S.); (V.B.B.)
| |
Collapse
|
6
|
Nanoparticle-based drug delivery systems in cancer: A focus on inflammatory pathways. Semin Cancer Biol 2022; 86:860-872. [PMID: 35115226 DOI: 10.1016/j.semcancer.2022.01.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/23/2022] [Accepted: 01/23/2022] [Indexed: 12/16/2022]
Abstract
It has become necessary to accept the clinical reality of therapeutic agents targeting the cancer-associated immune system. In recent decades, several investigations have highlighted the role of inflammation in cancer development. It has now been recognized that inflammatory cells secrete mediators, including enzymes, chemokines, and cytokines. These secreted substances produce an inflammatory microenvironment that is critically involved in cancer growth. Inflammation may enhance genomic instability leading to DNA damage, activation of oncogenes, or compromised tumor suppressor activity, all of which may promote various phases of carcinogenesis. Conventional cancer treatment includes surgery, radiation, and chemotherapy. However, treatment failure occurs because current strategies are unable to achieve complete local control due to metastasis. Nanoparticles (NPs) are a broad spectrum of drug carriers typically below the size of 100 nm, targeting tumor sites while reducing off-target consequences. More importantly, NPs can stimulate innate and adaptive immune systems in the tumor microenvironment (TME); hence, they induce a cancer-fighting immune response. Strikingly, targeting cancer cells with NPs helps eliminate drug resistance and tumor recurrence, as well as prevents inflammation. Throughout this review, we provide recent data on the role of inflammation in cancer and explore nano-therapeutic initiatives to target significant mediators, for example, nuclear factor-kappa B (NF-κB), tumor necrosis factor-α (TNF-α), and interleukins (ILs) associated with cancer-related inflammation, to escort the immunomodulators to cancer cells and associated systemic compartments. We also highlight the necessity of better identifying inflammatory pathways in cancer pathophysiology to develop effective treatment plans.
Collapse
|
7
|
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
|
8
|
Muehe A, Nejadnik H, Muehe H, Rosenberg J, Gharibi H, Saei AA, Lyu SC, Nadeau KC, Mahmoudi M, Daldrup-Link HE. Can the biomolecular corona induce an allergic reaction?-A proof-of-concept study. Biointerphases 2021; 16:011008. [PMID: 33706522 PMCID: PMC7861880 DOI: 10.1116/6.0000755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Ferumoxytol nanoparticles are being used clinically for the treatment of anemia and molecular imaging in patients. It is well documented that while most patients tolerate ferumoxytol well, a small percentage of patients (i.e., 0.01%) develop severe allergic reactions. The purpose of our proof-of-concept study was to determine whether patients with or without hypersensitivity reactions have specific protein corona profiles around ferumoxytol nanoparticles. In a retrospective, institutional review board approved pilot study, we enrolled 13 pediatric patients (5 girls, 8 boys, mean age 16.9 ± 8.2 years) who received a ferumoxytol-enhanced magnetic resonance imaging and who did (group 1, n = 5) or did not (group 2, n = 8) develop an allergic reaction. Blood samples of these patients were incubated with ferumoxytol, and the formation of a hard protein corona around ferumoxytol nanoparticles was measured by dynamic light scattering, zeta potential, and liquid chromatography-mass spectrometry. We also performed in vitro immune response analyses to randomly selected coronas from each group. Our results provide preliminary evidence that ex vivo analysis of the biomolecular corona may provide useful and predictive information on the possibility of severe allergic reactions to ferumoxytol nanoparticles. In the future, patients with predisposition of an allergic reaction to ferumoxytol may be diagnosed based on the proteomic patterns of the corona around ferumoxytol in their blood sample.
Collapse
Affiliation(s)
| | | | | | - Jarrett Rosenberg
- Department of Radiology, Pediatric Molecular Imaging, Molecular Imaging Program at Stanford, Stanford University, Stanford, California 94305
| | - Hassan Gharibi
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden
| | | | - Shu-Chen Lyu
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California 94305
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, California 94305
| | - Morteza Mahmoudi
- Precision Health Program and Department of Radiology, Michigan State University, East Lansing, Michigan 48824
| | | |
Collapse
|
9
|
Frtús A, Smolková B, Uzhytchak M, Lunova M, Jirsa M, Kubinová Š, Dejneka A, Lunov O. Analyzing the mechanisms of iron oxide nanoparticles interactions with cells: A road from failure to success in clinical applications. J Control Release 2020; 328:59-77. [DOI: 10.1016/j.jconrel.2020.08.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/31/2022]
|
10
|
Levada K, Pshenichnikov S, Omelyanchik A, Rodionova V, Nikitin A, Savchenko A, Schetinin I, Zhukov D, Abakumov M, Majouga A, Lunova M, Jirsa M, Smolková B, Uzhytchak M, Dejneka A, Lunov O. Progressive lysosomal membrane permeabilization induced by iron oxide nanoparticles drives hepatic cell autophagy and apoptosis. NANO CONVERGENCE 2020; 7:17. [PMID: 32424769 PMCID: PMC7235155 DOI: 10.1186/s40580-020-00228-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/29/2020] [Indexed: 05/02/2023]
Abstract
Iron oxide nanoparticles (IONs) are frequently used in various biomedical applications, in particular as magnetic resonance imaging contrast agents in liver imaging. Indeed, number of IONs have been withdrawn due to their poor clinical performance. Yet comprehensive understanding of their interactions with hepatocytes remains relatively limited. Here we investigated how iron oxide nanocubes (IO-cubes) and clusters of nanocubes (IO-clusters) affect distinct human hepatic cell lines. The viability of HepG2, Huh7 and Alexander cells was concentration-dependently decreased after exposure to either IO-cubes or IO-clusters. We found similar cytotoxicity levels in three cell lines triggered by both nanoparticle formulations. Our data indicate that different expression levels of Bcl-2 predispose cell death signaling mediated by nanoparticles. Both nanoparticles induced rather apoptosis than autophagy in HepG2. Contrary, IO-cubes and IO-clusters trigger distinct cell death signaling events in Alexander and Huh7 cells. Our data clarifies the mechanism by which cubic nanoparticles induce autophagic flux and the mechanism of subsequent toxicity. These findings imply that the cytotoxicity of ION-based contrast agents should be carefully considered, particularly in patients with liver diseases.
Collapse
Affiliation(s)
- Kateryna Levada
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Stanislav Pshenichnikov
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Alexander Omelyanchik
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Valeria Rodionova
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Aleksey Nikitin
- National University of Science and Technology "MISIS", Moscow, Russia
| | | | - Igor Schetinin
- National University of Science and Technology "MISIS", Moscow, Russia
| | - Dmitry Zhukov
- National University of Science and Technology "MISIS", Moscow, Russia
| | - Maxim Abakumov
- National University of Science and Technology "MISIS", Moscow, Russia
| | - Alexander Majouga
- National University of Science and Technology "MISIS", Moscow, Russia
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic.
| |
Collapse
|
11
|
Iron Oxide Nanoparticle-Induced Autophagic Flux Is Regulated by Interplay between p53-mTOR Axis and Bcl-2 Signaling in Hepatic Cells. Cells 2020; 9:cells9041015. [PMID: 32325714 PMCID: PMC7226334 DOI: 10.3390/cells9041015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023] Open
Abstract
Iron oxide-based nanoparticles have been repeatedly shown to affect lysosomal-mediated signaling. Recently, nanoparticles have demonstrated an ability to modulate autophagic flux via lysosome-dependent signaling. However, the precise underlying mechanisms of such modulation as well as the impact of cellular genetic background remain enigmatic. In this study, we investigated how lysosomal-mediated signaling is affected by iron oxide nanoparticle uptake in three distinct hepatic cell lines. We found that nanoparticle-induced lysosomal dysfunction alters sub-cellular localization of pmTOR and p53 proteins. Our data indicate that alterations in the sub-cellular localization of p53 protein induced by nanoparticle greatly affect the autophagic flux. We found that cells with high levels of Bcl-2 are insensitive to autophagy initiated by nanoparticles. Altogether, our data identify lysosomes as a central hub that control nanoparticle-mediated responses in hepatic cells. Our results provide an important fundamental background for the future development of targeted nanoparticle-based therapies.
Collapse
|
12
|
Świętek M, Panchuk R, Skorokhyd N, Černoch P, Finiuk N, Klyuchivska O, Hrubý M, Molčan M, Berger W, Trousil J, Stoika R, Horák D. Magnetic Temperature-Sensitive Solid-Lipid Particles for Targeting and Killing Tumor Cells. Front Chem 2020; 8:205. [PMID: 32328477 PMCID: PMC7161697 DOI: 10.3389/fchem.2020.00205] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/04/2020] [Indexed: 12/14/2022] Open
Abstract
Magnetic and temperature-sensitive solid lipid particles (mag. SLPs) were prepared in the presence of oleic acid-coated iron oxide (IO-OA) nanoparticles with 1-tetradecanol and poly(ethylene oxide)-block-poly(ε-caprolactone) as lipid and stabilizing surfactant-like agents, respectively. The particles, typically ~850 nm in hydrodynamic size, showed heat dissipation under the applied alternating magnetic field. Cytotoxic activity of the mag.SLPs, non-magnetic SLPs, and iron oxide nanoparticles was compared concerning the mammalian cancer cell lines and their drug-resistant counterparts using trypan blue exclusion test and MTT assay. The mag.SLPs exhibited dose-dependent cytotoxicity against human leukemia cell lines growing in suspension (Jurkat and HL-60/wt), as well as the doxorubicin (Dox)- and vincristine-resistant HL-60 sublines. The mag.SLPs showed higher cytotoxicity toward drug-resistant sublines as compared to Dox. The human glioblastoma cell line U251 growing in a monolayer culture was also sensitive to mag.SLPs cytotoxicity. Staining of U251 cells with the fluorescent dyes Hoechst 33342 and propidium iodide (PI) revealed that mag.SLPs treatment resulted in an increased number of cells with condensed chromatin and/or fragmented nuclei as well as with blebbing of the plasma membranes. While the Hoechst 33342 staining of cell suggested the pro-apoptotic activity of the particles, the PI staining indicated the pro-necrotic changes in the target cells. These conclusions were confirmed by Western blot analysis of apoptosis-related proteins, study of DNA fragmentation (DNA laddering due to the inter-nucleosomal cleavage and DNA comets due to single strand breaks), as well as by FACS analysis of the patterns of cell cycle distribution (pre-G1 phase) and Annexin V/PI staining of the treated Jurkat cells. The induction of apoptosis or necrosis by the particles used to treat Jurkat cells depended on the dose of the particles. Production of the reactive oxygen species (ROS) was proposed as a potential mechanism of mag.SLPs-induced cytotoxicity. Accordingly, hydrogen peroxide and superoxide radical levels in mag.SLPs-treated Jurkat leukemic cells were increased by ~20–40 and ~70%, respectively. In contrast, the non-magnetic SLPs and neat iron oxides did not influence ROS levels significantly. Thus, the developed mag.SLPs can be used for effective killing of human tumor cells, including drug-resistant ones.
Collapse
Affiliation(s)
- Małgorzata Świętek
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czechia
| | - Rostyslav Panchuk
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Science of Ukraine, Lviv, Ukraine
| | - Nadia Skorokhyd
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Science of Ukraine, Lviv, Ukraine
| | - Peter Černoch
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czechia
| | - Nataliya Finiuk
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Science of Ukraine, Lviv, Ukraine
| | - Olha Klyuchivska
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Science of Ukraine, Lviv, Ukraine
| | - Martin Hrubý
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czechia
| | - Matúš Molčan
- Institute of Experimental Physics, Slovak Academy of Sciences, Košice, Slovakia
| | - Walter Berger
- Department of Medicine I, Medical University of Vienna, Institute of Cancer Research and Comprehensive Cancer Center, Vienna, Austria
| | - Jirí Trousil
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czechia
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Science of Ukraine, Lviv, Ukraine
| | - Daniel Horák
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
13
|
Kono Y, Gogatsubo S, Ohba T, Fujita T. Enhanced macrophage delivery to the colon using magnetic lipoplexes with a magnetic field. Drug Deliv 2020; 26:935-943. [PMID: 31530198 PMCID: PMC6758636 DOI: 10.1080/10717544.2019.1662515] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Magnetically guided cell delivery systems would be valuable to achieve effective macrophage-based cell therapy for colonic inflammatory diseases. In the current study, we developed a method for the efficient and simultaneous introduction of superparamagnetic iron oxide nanoparticles (SPIONs) and plasmid DNA (pDNA) into RAW264 murine macrophage-like cells using SPION-incorporated cationic liposome/pDNA complexes (magnetic lipoplexes). SPIONs and pDNA were introduced for magnetization and functionalization of the macrophages, respectively. We also evaluated the adhesive properties of magnetized RAW264 cells using magnetic lipoplexes in the murine colon under a magnetic field. Significant cellular association and gene expression without cytotoxicity were observed when magnetic cationic liposomes and pDNA were mixed at a weight ratio of 10:1, and SPION concentration and magnetic field exposure time was 0.1 mg/mL and 10 min, respectively. We also observed that cytokine production in magnetized RAW264 cells was similar to that in non-treated RAW264 cells, whereas nitric oxide production was significantly increased in magnetized RAW264 cells. Furthermore, magnetized RAW264 cells highly adhered to a Caco-2 cell monolayer and colon in mice, under a magnetic field. These results suggest that this magnetic cell delivery system can improve the colonic delivery of macrophages and its therapeutic efficacy against colonic inflammatory diseases.
Collapse
Affiliation(s)
- Yusuke Kono
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University , Kusatsu , Japan.,Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University , Kusatsu , Japan
| | - Serika Gogatsubo
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University , Kusatsu , Japan
| | - Takeshi Ohba
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University , Kusatsu , Japan
| | - Takuya Fujita
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University , Kusatsu , Japan.,Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University , Kusatsu , Japan.,Research Center for Drug Discovery and Development, Ritsumeikan University , Kusatsu , Japan
| |
Collapse
|
14
|
Yamaguchi M, Ohnuki K, Hotta K, Fujii H. MR signal changes in superparamagnetic iron oxide nanoparticle-labeled macrophages in response to X irradiation. NMR IN BIOMEDICINE 2019; 32:e4132. [PMID: 31305958 DOI: 10.1002/nbm.4132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 04/16/2019] [Accepted: 05/21/2019] [Indexed: 06/10/2023]
Abstract
To investigate whether MR signals associated with macrophages labeled with superparamagnetic iron oxide nanoparticles (SPIONs) change in response to X irradiation, we performed in vitro MRI of SPION-labeled macrophage-like J774A.1 cells that were subsequently X irradiated. We labeled the cells with ferucarbotran at a concentration of 10 μg iron/mL in culture medium for 16 h and subsequently performed X irradiation at doses of 0, 2, 10, or 20 Gy using a low-energy X-ray unit. On Days 3 and 6, we suspended the cells in agar at a concentration of 2 × 106 cells/mL and acquired multi-gradient echo and multi-spin echo images of the cell samples using a 3 T scanner to estimate R2 * and R2 . In addition, we microscopically investigated the relationship among the MR signal changes, intracellular SPIONs, and acidic organelles. Our data showed that X irradiation of labeled cells caused increased SPION deposition in lysosomes compared with the non-irradiated control. On Day 3, R2 * and R2 values in the 0 to 10 Gy irradiated samples were dose-dependently increased 5.4- and 1.5-fold compared with 17 ± 2 and 13 ± 1/s, respectively, in the non-irradiated control; these values plateaued at more than 10 Gy. Although the increases in R2 *, R2 , and SPION deposition were still observed in the 10 and 20 Gy samples on Days 6 and 7, the 2 Gy samples showed recovery in these parameters as cell growth was restored. Acidic organelles were temporarily increased in the irradiated cells, which suggests that the reduction in lysosomal acidity was not attributable to SPION deposition. In conclusion, X irradiation of macrophages can cause SPION deposition and R2 * and R2 elevation in a specific dose range. MRI of SPION-labeled and subsequently X-irradiated macrophages may be utilized as a novel technique for investigating macrophage responses to X irradiation.
Collapse
Affiliation(s)
- Masayuki Yamaguchi
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Kazunobu Ohnuki
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Kenji Hotta
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Hirofumi Fujii
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| |
Collapse
|
15
|
Pongrac IM, Radmilović MD, Ahmed LB, Mlinarić H, Regul J, Škokić S, Babič M, Horák D, Hoehn M, Gajović S. D-mannose-Coating of Maghemite Nanoparticles Improved Labeling of Neural Stem Cells and Allowed Their Visualization by ex vivo MRI after Transplantation in the Mouse Brain. Cell Transplant 2019; 28:553-567. [PMID: 31293167 PMCID: PMC7103599 DOI: 10.1177/0963689719834304] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 12/26/2018] [Accepted: 02/05/2019] [Indexed: 12/14/2022] Open
Abstract
Magnetic resonance imaging (MRI) of superparamagnetic iron oxide-labeled cells can be used as a non-invasive technique to track stem cells after transplantation. The aim of this study was to (1) evaluate labeling efficiency of D-mannose-coated maghemite nanoparticles (D-mannose(γ-Fe2O3)) in neural stem cells (NSCs) in comparison to the uncoated nanoparticles, (2) assess nanoparticle utilization as MRI contrast agent to visualize NSCs transplanted into the mouse brain, and (3) test nanoparticle biocompatibility. D-mannose(γ-Fe2O3) labeled the NSCs better than the uncoated nanoparticles. The labeled cells were visualized by ex vivo MRI and their localization subsequently confirmed on histological sections. Although the progenitor properties and differentiation of the NSCs were not affected by labeling, subtle effects on stem cells could be detected depending on dose increase, including changes in cell proliferation, viability, and neurosphere diameter. D-mannose coating of maghemite nanoparticles improved NSC labeling and allowed for NSC tracking by ex vivo MRI in the mouse brain, but further analysis of the eventual side effects might be necessary before translation to the clinic.
Collapse
Affiliation(s)
- Igor M. Pongrac
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | | | - Lada Brkić Ahmed
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | - Hrvoje Mlinarić
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | - Jan Regul
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | - Siniša Škokić
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | - Michal Babič
- Institute of Macromolecular Chemistry, Academy of Sciences, Prague, Czech
Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Academy of Sciences, Prague, Czech
Republic
| | - Mathias Hoehn
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory,
Cologne, Germany
| | - Srećko Gajović
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| |
Collapse
|
16
|
Guo R, Li Q, Yang F, Hu X, Jiao J, Guo Y, Wang J, Zhang Y. In Vivo MR Imaging of Dual MRI Reporter Genes and Deltex-1 Gene-modified Human Mesenchymal Stem Cells in the Treatment of Closed Penile Fracture. Mol Imaging Biol 2019; 20:417-427. [PMID: 28971290 DOI: 10.1007/s11307-017-1128-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE The purpose of this study was to investigate the feasibility of dual magnetic resonance imaging (MRI) reporter genes, including ferritin heavy subunit (Fth) and transferrin receptor (TfR), which provide sufficient MRI contrast for in vivo MRI tracking, and the Deltex-1 (DTX1) gene, which promotes human mesenchymal stem cell (hMSC) differentiation to smooth muscle cells (SMCs), to treat closed penile fracture (CPF). METHODS Multi-gene co-expressing hMSCs were generated. The expression of mRNA and proteins was assessed, and the original biological properties of hMSCs were determined and compared. The intracellular uptake of iron was evaluated, and the ability to differentiate into SMCs was detected. Fifty rabbits with CPF were randomly transplanted with PBS, hMSCs, Fth-TfR-hMSCs, DTX1-hMSCs, and Fth-TfR-DTX1-hMSCs. In vivo MRI was performed to detect the distribution and migration of the grafted cells and healing progress of CPF, and the results were correlated with histology. RESULTS The mRNA and proteins of the multi-gene were highly expressed. The transgenes could not influence the original biological properties of hMSCs. The dual MRI reporter genes increased the iron accumulation capacity, and the DTX1 gene promoted hMSC differentiation into SMCs. The distribution and migration of the dual MRI reporter gene-modified hMSCs, and the healing state of CPF could be obviously detected by MRI and confirmed by histology. CONCLUSION The dual MRI reporter genes could provide sufficient MRI contrast, and the distribution and migration of MSCs could be detected in vivo. The DTX1 gene can promote MSC differentiation into SMCs for the treatment of CPF and effectively inhibit granulation tissue formation.
Collapse
Affiliation(s)
- Ruomi Guo
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qingling Li
- Department of VIP Medical Center, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fei Yang
- Department of Urology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaojun Hu
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ju Jiao
- Department of Nuclear Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yu Guo
- Department of VIP Medical Center, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jin Wang
- Department of Radiology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yong Zhang
- Department of Nuclear Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
17
|
In Vitro Neural Differentiation of Bone Marrow Mesenchymal Stem Cells Carrying the FTH1 Reporter Gene and Detection with MRI. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1978602. [PMID: 30046590 PMCID: PMC6038692 DOI: 10.1155/2018/1978602] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/24/2018] [Accepted: 05/31/2018] [Indexed: 01/15/2023]
Abstract
Magnetic resonance imaging (MRI) based on the ferritin heavy chain 1 (FTH1) reporter gene has been used to trace stem cells. However, whether FTH1 expression is affected by stem cell differentiation or whether cell differentiation is affected by reporter gene expression remains unclear. Here, we explore the relationship between FTH1 expression and neural differentiation in the differentiation of mesenchymal stem cells (MSCs) carrying FTH1 into neuron-like cells and investigate the feasibility of using FTH1 as an MRI reporter gene to detect neurally differentiated cells. By inducing cell differentiation with all-trans retinoic acid and a modified neuronal medium, MSCs and MSCs-FTH1 were successfully differentiated into neuron-like cells (Neurons and Neurons-FTH1), and the neural differentiation rates were (91.56±7.89)% and (92.23±7.64)%, respectively. Neuron-specific markers, including nestin, neuron-specific enolase, and microtubule-associated protein-2, were significantly expressed in Neurons-FTH1 and Neurons without noticeable differences. On the other hand, FTH1 was significantly expressed in MSCs-FTH1 and Neurons-FTH1 cells, and the expression levels were not significantly different. The R2 value was significantly increased in MSCs-FTH1 and Neurons-FTH1 cells, which was consistent with the findings of Prussian blue staining, transmission electron microscopy, and intracellular iron measurements. These results suggest that FTH1 gene expression did not affect MSC differentiation into neurons and was not affected by neural differentiation. Thus, MRI reporter gene imaging based on FTH1 can be used for the detection of neurally differentiated cells from MSCs.
Collapse
|
18
|
Wu Q, Miao T, Feng T, Yang C, Guo Y, Li H. Dextran‑coated superparamagnetic iron oxide nanoparticles activate the MAPK pathway in human primary monocyte cells. Mol Med Rep 2018; 18:564-570. [PMID: 29749448 DOI: 10.3892/mmr.2018.8972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 04/25/2018] [Indexed: 02/05/2023] Open
Abstract
With the increase in applications of superparamagnetic iron oxide nanoparticles (SPIONs) in biomedicine, it is essential to investigate the bio‑security of these nanoparticles, especially with respect to the human immune system. In the present study, the biological effects of dextran‑coated superparamagnetic iron oxide nanoparticles (Dex‑SPIONs) on human primary monocyte cells were evaluated. The results of the present study demonstrated that Dex‑SPIONs can be identified in phagosomes or freed in the cytoplasm and did not affect cell viability or induce apoptosis. Notably, there were certain bulky vacuoles and a number of pseudopodia from the cell membrane, suggesting potential activation of human monocyte cells. In addition, the expression levels of pro‑inflammatory cytokines interleukin (IL)‑1β and tumor necrosis factor (TNF)‑α were also increased following treatment with Dex‑SPIONs. Simultaneously, the phosphorylation levels of mitogen‑activated protein kinase (MAPK) p38, c‑Jun N‑terminal kinase 1 and extracellular signal regulated kinase were markedly enhanced following nanoparticle exposure and MAPK inhibitors could abate the production of IL‑1β and TNF‑α. The results of the present study demonstrated that Dex‑SPIONs could activate human monocyte cells and that activation of MAPK pathway may be involved in these effects.
Collapse
Affiliation(s)
- Qihong Wu
- Key Laboratory of Obstetrics and Gynecology and Pediatric Disease and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Tianyu Miao
- Department of Vascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ting Feng
- Key Laboratory of Obstetrics and Gynecology and Pediatric Disease and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chuan Yang
- Key Laboratory of Obstetrics and Gynecology and Pediatric Disease and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yingkun Guo
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong Li
- Key Laboratory of Obstetrics and Gynecology and Pediatric Disease and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
19
|
Zhao Y, Zhao X, Cheng Y, Guo X, Yuan W. Iron Oxide Nanoparticles-Based Vaccine Delivery for Cancer Treatment. Mol Pharm 2018; 15:1791-1799. [DOI: 10.1021/acs.molpharmaceut.7b01103] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yi Zhao
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Xiaotian Zhao
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Yuan Cheng
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Xiaoshuang Guo
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| | - Weien Yuan
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
20
|
Hsiao YP, Shen CC, Huang CH, Lin YC, Jan TR. Iron oxide nanoparticles attenuate T helper 17 cell responses in vitro and in vivo. Int Immunopharmacol 2018; 58:32-39. [PMID: 29549717 DOI: 10.1016/j.intimp.2018.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 02/15/2018] [Accepted: 03/07/2018] [Indexed: 12/27/2022]
Abstract
Iron oxide nanoparticles (IONPs) have been shown to attenuate T helper (Th)1 and Th2 cell-mediated immunity in ovalbumin (OVA)-sensitized mice. The objective of this study is to investigate the effects of IONPs on the immune responses of Th17 cells, a subset of T cells involved in various inflammatory pathologies. For in vivo study, a murine model of delayed-type hypersensitivity (DTH) was employed. BALB/c mice received a single dose of IONPs (0.2-10 mg iron/kg) via the tail vein 1 h prior to ovalbumin (OVA) sensitization. Their footpads were subcutaneously challenged with OVA to induce DTH reactions. The expression of Th17 cell-related molecules in inflamed footpads were examined by immunohistochemistry. For in vitro study, OVA-primed splenocytes were directly exposed to IONPs (1-100 μg iron/mL), and then re-stimulated with OVA in culture. The expression of Th17 cell-related molecules were measured by reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. IONP administration attenuated the number of interleukin (IL)-6, IL-17, the transcription factor ROR-γ, and chemokine receptor 6 positive cells in OVA-challenged footpads, whereas the number of transforming growth factor-β, IL-23 and chemokine (C-C motif) ligand 20 positive cells was not altered. Direct exposure of OVA-primed splenocytes to IONPs suppressed the production of IL-6 and IL-17, and the mRNA expression of IL-17 and ROR-γt. These data indicate that exposure to IONPs attenuates Th17 cell responses in vivo and in vitro.
Collapse
Affiliation(s)
- Yai-Ping Hsiao
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Chang Shen
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-Hsiung Huang
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan; National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Chin Lin
- Department of Medicinal Botanicals and Health Applications, College of Biotechnology & Bioresources, Da-Yeh University, No.168, University Rd., Dacun, Changhua, Taiwan.
| | - Tong-Rong Jan
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
21
|
Patil RM, Thorat ND, Shete PB, Bedge PA, Gavde S, Joshi MG, Tofail SA, Bohara RA. Comprehensive cytotoxicity studies of superparamagnetic iron oxide nanoparticles. Biochem Biophys Rep 2018; 13:63-72. [PMID: 29349357 PMCID: PMC5766481 DOI: 10.1016/j.bbrep.2017.12.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 11/20/2022] Open
Abstract
Recently lots of efforts have been taken to develop superparamagnetic iron oxide nanoparticles (SPIONs) for biomedical applications. So it is utmost necessary to have in depth knowledge of the toxicity occurred by this material. This article is designed in such way that it covers all the associated toxicity issues of SPIONs. It mainly emphasis on toxicity occurred at different levels including cellular alterations in the form of damage to nucleic acids due to oxidative stress and altered cellular response. In addition focus is been devoted for in vitro and in vivo toxicity of SPIONs, so that a better therapeutics can be designed. At the end the time dependent nature of toxicity and its ultimate faith inside the body is being discussed.
Collapse
Affiliation(s)
- Rakesh M. Patil
- Directorate of Forensic Science Laboratory, Govt. of Maharashtra Kalina, Mumbai, India
- Centre for Interdisciplinary Research, D.Y.Patil University, Kolhapur, India
| | - Nanasaheb D. Thorat
- Material and Surface Science Institute, Bernal Institute, University of Limerick, Ireland
| | - Prajkta B. Shete
- Centre for Interdisciplinary Research, D.Y.Patil University, Kolhapur, India
| | - Poonam A. Bedge
- Department of Stem Cells and Regenerative Medicine, D.Y.Patil University, Kolhapur, India
| | - Shambala Gavde
- Centre for Interdisciplinary Research, D.Y.Patil University, Kolhapur, India
| | - Meghnad G. Joshi
- Department of Stem Cells and Regenerative Medicine, D.Y.Patil University, Kolhapur, India
| | - Syed A.M. Tofail
- Material and Surface Science Institute, Bernal Institute, University of Limerick, Ireland
| | - Raghvendra A. Bohara
- Centre for Interdisciplinary Research, D.Y.Patil University, Kolhapur, India
- Department of Stem Cells and Regenerative Medicine, D.Y.Patil University, Kolhapur, India
- Research and Innovations for Comprehensive Health (RICH), Cell D.Y.Patil University, Kolhapur, India
| |
Collapse
|
22
|
Abdollah MRA, Carter TJ, Jones C, Kalber TL, Rajkumar V, Tolner B, Gruettner C, Zaw-Thin M, Baguña Torres J, Ellis M, Robson M, Pedley RB, Mulholland P, T M de Rosales R, Chester KA. Fucoidan Prolongs the Circulation Time of Dextran-Coated Iron Oxide Nanoparticles. ACS NANO 2018; 12:1156-1169. [PMID: 29341587 DOI: 10.1021/acsnano.7b06734] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The magnetic properties and safety of dextran-coated superparamagnetic iron oxide nanoparticles (SPIONs) have facilitated their clinical use as MRI contrast agents and stimulated research on applications for SPIONs in particle imaging and magnetic hyperthermia. The wider clinical potential of SPIONs, however, has been limited by their rapid removal from circulation via the reticuloendothelial system (RES). We explored the possibility of extending SPION circulatory time using fucoidan, a seaweed-derived food supplement, to inhibit RES uptake. The effects of fucoidan on SPION biodistribution were evaluated using ferucarbotran, which in its pharmaceutical formulation (Resovist) targets the RES. Ferucarbotran was radiolabeled at the iron oxide core with technetium-99m (99mTc; t1/2 = 6 h) or zirconium-89 (89Zr; t1/2 = 3.3 days). Results obtained with 99mTc-ferucarbotran demonstrated that administration of fucoidan led to a 4-fold increase in the circulatory half-life (t1/2 slow) from 37.4 to 150 min (n = 4; P < 0.0001). To investigate whether a longer circulatory half-life could lead to concomitant increased tumor uptake, the effects of fucoidan were tested with 89Zr-ferucarbotran in mice bearing syngeneic subcutaneous (GL261) tumors. In this model, the longer circulatory half-life achieved with fucoidan was associated with a doubling in tumor SPION uptake (n = 5; P < 0.001). Fucoidan was also effective in significantly increasing the circulatory half-life of perimag-COOH, a commercially available SPION with a larger hydrodynamic size (130 nm) than ferucarbotran (65 nm). These findings indicate successful diversion of SPIONs away from the hepatic RES and show realistic potential for future clinical applications.
Collapse
Affiliation(s)
- Maha R A Abdollah
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE) , El Shorouk City, Misr- Ismalia Desert Road, Cairo 11837, Egypt
| | - Thomas J Carter
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Clare Jones
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London , London WC1E 6DD, U.K
| | - Vineeth Rajkumar
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Berend Tolner
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Cordula Gruettner
- Micromod Partikeltechnologie GmbH , Friedrich-Barnewitz-Str. 4, D-18119 Rostock, Germany
| | - May Zaw-Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London , London WC1E 6DD, U.K
| | - Julia Baguña Torres
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Matthew Ellis
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Institute of Neurology (ION), University College London (UCL) , Queen Square, London WC1N 3BG, U.K
| | - Mathew Robson
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - R Barbara Pedley
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Paul Mulholland
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Kerry Ann Chester
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| |
Collapse
|
23
|
Nedyalkova M, Donkova B, Romanova J, Tzvetkov G, Madurga S, Simeonov V. Iron oxide nanoparticles - In vivo/in vitro biomedical applications and in silico studies. Adv Colloid Interface Sci 2017; 249:192-212. [PMID: 28499604 DOI: 10.1016/j.cis.2017.05.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/28/2017] [Accepted: 05/02/2017] [Indexed: 12/22/2022]
Abstract
The review presents a broad overview of the biomedical applications of surface functionalized iron oxide nanoparticles (IONPs) as magnetic resonance imaging (MRI) agents for sensitive and precise diagnosis tool and synergistic combination with other imaging modalities. Then, the recent progress in therapeutic applications, such as hyperthermia is discussed and the available toxicity data of magnetic nanoparticles concerning in vitro and in vivo biomedical applications are addressed. This review also presents the available computer models using molecular dynamics (MD), Monte Carlo (MC) and density functional theory (DFT), as a basis for a complete understanding of the behaviour and morphology of functionalized IONPs, for improving NPs surface design and expanding the potential applications in nanomedicine.
Collapse
Affiliation(s)
- Miroslava Nedyalkova
- Faculty of Chemistry and Pharmacy, University of Sofia "St. Kl. Okhridski". J. Bourchier Blvd. 1, 1164 Sofia, Bulgaria.
| | - Borjana Donkova
- Faculty of Chemistry and Pharmacy, University of Sofia "St. Kl. Okhridski". J. Bourchier Blvd. 1, 1164 Sofia, Bulgaria
| | - Julia Romanova
- Faculty of Chemistry and Pharmacy, University of Sofia "St. Kl. Okhridski". J. Bourchier Blvd. 1, 1164 Sofia, Bulgaria
| | - George Tzvetkov
- Faculty of Chemistry and Pharmacy, University of Sofia "St. Kl. Okhridski". J. Bourchier Blvd. 1, 1164 Sofia, Bulgaria
| | - Sergio Madurga
- Materials Science and Physical Chemistry Department & Research Institute of Theoretical and Computational Chemistry (IQTCUB) of Barcelona University (UB), C/Martí i Franquès, 1, 08028 Barcelona, Catalonia, Spain
| | - Vasil Simeonov
- Faculty of Chemistry and Pharmacy, University of Sofia "St. Kl. Okhridski". J. Bourchier Blvd. 1, 1164 Sofia, Bulgaria
| |
Collapse
|
24
|
Pandey RK, Prajapati VK. Molecular and immunological toxic effects of nanoparticles. Int J Biol Macromol 2017; 107:1278-1293. [PMID: 29017884 DOI: 10.1016/j.ijbiomac.2017.09.110] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/21/2017] [Accepted: 09/27/2017] [Indexed: 02/07/2023]
Abstract
Nanoparticles have emerged as a boon for the public health applications such as drug delivery, diagnostic, and imaging. Biodegradable and non-bio degradable nanoparticles have been used at a large scale level to increase the efficiency of the biomedical process at the cellular, animal and human level. Exponential use of nanoparticles reinforces the adverse immunological changes at the human health level. Physical and chemical properties of nanoparticles often lead to a variety of immunotoxic effects such as activation of stress-related genes, membrane disruption, and release of pro-inflammatory cytokines. Delivered nanoparticles in animal or human interact with various components of the immune system such as lymphocytes, macrophages, neutrophils etc. Nanoparticles delivered above the threshold level damages the cellular physiology by the generation of reactive oxygen and nitrogen species. This review article represents the potential of nanoparticles in the field of nanomedicine and provides the critical evidence which leads to develop immunotoxicity in living cells and organisms by altering immunological responses.
Collapse
Affiliation(s)
- Rajan Kumar Pandey
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Kishangarh, 305817, Ajmer, Rajasthan, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Kishangarh, 305817, Ajmer, Rajasthan, India.
| |
Collapse
|
25
|
Wu Q, Jin R, Feng T, Liu L, Yang L, Tao Y, Anderson JM, Ai H, Li H. Iron oxide nanoparticles and induced autophagy in human monocytes. Int J Nanomedicine 2017; 12:3993-4005. [PMID: 28603414 PMCID: PMC5457122 DOI: 10.2147/ijn.s135189] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Superparamagnetic iron oxide nanoparticles have been widely used in biomedical applications, but understanding of their interactions with the biological immune system is relatively limited. This work is focused on dextran-coated iron oxide nanoparticles and their induced autophagy in human monocytes. We found that these nanoparticles can be taken up by human monocytes, followed by localization within vesicles or free in cytoplasm. Autophagosome formation was observed with increased expression of LC3II protein, the specific marker of autophagy. The autophagy substrate p62 was degraded in a dose-dependent manner, and autophagy was blocked by autophagy (or lysosome) inhibitors alone or along with iron oxide nanoparticles, indicating that autophagosome accumulation was mainly due to autophagy induction, rather than blockade of autophagy flux. Interestingly, iron oxide nanoparticles increased the viability of human monocytes, but the mechanism was not clear. We further found that inhibition of autophagy mostly attenuated the survival of cells, with acceleration of the inflammation induced by these nanoparticles. Taken together, autophagic activation in human monocytes may play a protective role against the cytotoxicity of iron oxide nanoparticles.
Collapse
Affiliation(s)
- QiHong Wu
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital
| | - RongRong Jin
- National Engineering Research Center for Biomaterials
| | - Ting Feng
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital
| | - Li Liu
- National Engineering Research Center for Biomaterials
| | - Li Yang
- National Engineering Research Center for Biomaterials
| | - YuHong Tao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - James M Anderson
- Department of Biomedical Engineering.,Department of Pathology, Case Western Reserve University, Cleveland, OH, US
| | - Hua Ai
- National Engineering Research Center for Biomaterials.,Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital.,Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Cheng S, Mi R, Xu Y, Jin G, Zhang J, Zhou Y, Chen Z, Liu F. Ferritin heavy chain as a molecular imaging reporter gene in glioma xenografts. J Cancer Res Clin Oncol 2017; 143:941-951. [PMID: 28247036 DOI: 10.1007/s00432-017-2356-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 01/27/2017] [Indexed: 01/22/2023]
Abstract
PURPOSE The development of glioma therapy in clinical practice (e.g., gene therapy) calls for efficiently visualizing and tracking glioma cells in vivo. Human ferritin heavy chain is a novel gene reporter in magnetic resonance imaging. This study proposes hFTH as a reporter gene for MR molecular imaging in glioma xenografts. METHODS Rat C6 glioma cells were infected by packaged lentivirus carrying hFTH and EGFP genes and obtained by fluorescence-activated cell sorting. The iron-loaded ability was analyzed by the total iron reagent kit. Glioma nude mouse models were established subcutaneously and intracranially. Then, in vivo tumor bioluminescence was performed via the IVIS spectrum imaging system. The MR imaging analysis was analyzed on a 7T animal MRI scanner. Finally, the expression of hFTH was analyzed by western blotting and histological analysis. RESULTS Stable glioma cells carrying hFTH and EGFP reporter genes were successfully obtained. The intracellular iron concentration was increased without impairing the cell proliferation rate. Glioma cells overexpressing hFTH showed significantly decreased signal intensity on T2-weighted MRI both in vitro and in vivo. EGFP fluorescent imaging could also be detected in the subcutaneous and intracranial glioma xenografts. Moreover, the expression of the transferritin receptor was significantly increased in glioma cells carrying the hFTH reporter gene. CONCLUSION Our study illustrated that hFTH generated cellular MR imaging contrast efficiently in glioma via regulating the expression of transferritin receptor. This might be a useful reporter gene in cell tracking and MR molecular imaging for glioma diagnosis, gene therapy and tumor metastasis.
Collapse
Affiliation(s)
- Sen Cheng
- Brain Tumor Research Center, Beijing Laboratory of Biomedical Materials, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Tiantan Xili 6, Dongcheng District, Beijing, 100050, People's Republic of China
| | - Ruifang Mi
- Brain Tumor Research Center, Beijing Laboratory of Biomedical Materials, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Tiantan Xili 6, Dongcheng District, Beijing, 100050, People's Republic of China
| | - Yu Xu
- Radiology Department, Dongzhimen Hospital Beijing University of Chinese Medicine, No. 5 Hai Yun Cang, Dong Cheng District, Beijing, 100700, People's Republic of China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Laboratory of Biomedical Materials, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Tiantan Xili 6, Dongcheng District, Beijing, 100050, People's Republic of China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Laboratory of Biomedical Materials, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Tiantan Xili 6, Dongcheng District, Beijing, 100050, People's Republic of China
| | - Yiqiang Zhou
- Brain Tumor Research Center, Beijing Laboratory of Biomedical Materials, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Tiantan Xili 6, Dongcheng District, Beijing, 100050, People's Republic of China
| | - Zhengguang Chen
- Radiology Department, Dongzhimen Hospital Beijing University of Chinese Medicine, No. 5 Hai Yun Cang, Dong Cheng District, Beijing, 100700, People's Republic of China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Laboratory of Biomedical Materials, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Tiantan Xili 6, Dongcheng District, Beijing, 100050, People's Republic of China.
| |
Collapse
|
27
|
Qiao C, Yang J, Chen L, Weng J, Zhang X. Intracellular accumulation and immunological responses of lipid modified magnetic iron nanoparticles in mouse antigen processing cells. Biomater Sci 2017; 5:1603-1611. [DOI: 10.1039/c7bm00244k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Lipid modified magnetic nanoparticles could enhance the intracellular accumulation and immune responses of mouse antigen processing cells.
Collapse
Affiliation(s)
- Chenmeng Qiao
- Key Laboratory of Advanced Technologies of Materials
- Ministry of Education
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
| | - Jun Yang
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- PR China
| | - Lei Chen
- Department of Obstetrics and Gynecology
- Navy General Hospital of People Liberation Army
- Beijing 100048
- PR China
| | - Jie Weng
- Key Laboratory of Advanced Technologies of Materials
- Ministry of Education
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- PR China
| |
Collapse
|
28
|
Ghalamfarsa G, Hojjat-Farsangi M, Mohammadnia-Afrouzi M, Anvari E, Farhadi S, Yousefi M, Jadidi-Niaragh F. Application of nanomedicine for crossing the blood–brain barrier: Theranostic opportunities in multiple sclerosis. J Immunotoxicol 2016; 13:603-19. [DOI: 10.3109/1547691x.2016.1159264] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ghasem Ghalamfarsa
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene Therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
- Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mousa Mohammadnia-Afrouzi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Enayat Anvari
- Department of Physiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Shohreh Farhadi
- Department of Agricultural Engineering, Islamic Azad University, Tehran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Tracking inflammation in the epileptic rat brain by bi-functional fluorescent and magnetic nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1335-45. [DOI: 10.1016/j.nano.2016.01.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/07/2016] [Accepted: 01/25/2016] [Indexed: 11/21/2022]
|
30
|
Zhukova GV, Goroshinskaya IA, Shikhliarova AI, Kit OI, Kachesova PS, Polozhentsev OE. On the self-dependent effect of metal nanoparticles on malignant tumors. Biophysics (Nagoya-shi) 2016. [DOI: 10.1134/s0006350916030234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
31
|
Tong HI, Kang W, Shi Y, Zhou G, Lu Y. Physiological function and inflamed-brain migration of mouse monocyte-derived macrophages following cellular uptake of superparamagnetic iron oxide nanoparticles-Implication of macrophage-based drug delivery into the central nervous system. Int J Pharm 2016; 505:271-82. [PMID: 27001531 DOI: 10.1016/j.ijpharm.2016.03.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/05/2016] [Accepted: 03/18/2016] [Indexed: 02/05/2023]
Abstract
This study was designed to use superparamagnetic iron oxide nanoparticles (SPIONs) as evaluating tools to study monocyte-derived macrophages (MDM)-mediated delivery of small molecular agents into the diseased brains. MDM were tested with different-configured SPIONs at selected concentrations for their impacts on carrier cells' physiological and migratory properties, which were found to depend largely on particle size, coating, and treatment concentrations. SHP30, a SPION of 30-nm core size with oleic acids plus amphiphilic polymer coating, was identified to have high cellular uptake efficiency and cause little cytotoxic effects on MDM. At lower incubation dose (25μg/mL), few alteration was observed in carrier cells' physiological and in vivo migratory functions, as tested in a lipopolysaccharide-induced acute neuroinflammation mouse model. Nevertheless, significant increase in monocyte-to-macrophage differentiation, and decrease in in vivo carrier MDM inflamed-brain homing ability were found in groups treated with a higher dose of SHP30at 100μg/mL. Overall, our results have identified MDM treatment at 25μg/mL SHP30 resulted in little functional changes, provided valuable parameters for using SPIONs as evaluating tools to study MDM-mediated therapeutics carriage and delivery, and supported the concepts of using monocytes-macrophages as cellular vehicles to transport small molecular agents to the brain.
Collapse
Affiliation(s)
- Hsin-I Tong
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA; Department of Microbiology, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Wen Kang
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Yingli Shi
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Guangzhou Zhou
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Yuanan Lu
- Office of Public Health Studies, University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| |
Collapse
|
32
|
Xu Y, Sherwood JA, Lackey KH, Qin Y, Bao Y. The responses of immune cells to iron oxide nanoparticles. J Appl Toxicol 2016; 36:543-53. [PMID: 26817529 DOI: 10.1002/jat.3282] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/20/2015] [Accepted: 12/03/2015] [Indexed: 12/15/2022]
Abstract
Immune cells play an important role in recognizing and removing foreign objects, such as nanoparticles. Among various parameters, surface coatings of nanoparticles are the first contact with biological system, which critically affect nanoparticle interactions. Here, surface coating effects on nanoparticle cellular uptake, toxicity and ability to trigger immune response were evaluated on a human monocyte cell line using iron oxide nanoparticles. The cells were treated with nanoparticles of three types of coatings (negatively charged polyacrylic acid, positively charged polyethylenimine and neutral polyethylene glycol). The cells were treated at various nanoparticle concentrations (5, 10, 20, 30, 50 μg ml(-1) or 2, 4, 8, 12, 20 μg cm(-2)) with 6 h incubation or treated at a nanoparticle concentration of 50 μg ml(-1) (20 μg cm(-2)) at different incubation times (6, 12, 24, 48 or 72 h). Cell viability over 80% was observed for all nanoparticle treatment experiments, regardless of surface coatings, nanoparticle concentrations and incubation times. The much lower cell viability for cells treated with free ligands (e.g. ~10% for polyethylenimine) suggested that the surface coatings were tightly attached to the nanoparticle surfaces. The immune responses of cells to nanoparticles were evaluated by quantifying the expression of toll-like receptor 2 and tumor necrosis factor-α. The expression of tumor necrosis factor-α and toll-like receptor 2 were not significant in any case of the surface coatings, nanoparticle concentrations and incubation times. These results provide useful information to select nanoparticle surface coatings for biological and biomedical applications.
Collapse
Affiliation(s)
- Yaolin Xu
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Jennifer A Sherwood
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Kimberly H Lackey
- Department of Biological Science, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Ying Qin
- Alabama Innovation and Mentoring of Entrepreneurs, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Yuping Bao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
33
|
Li J, Liu Y, Cha R, Ran B, Mou K, Wang H, Xie Q, Sun J, Jiang X. The biocompatibility evaluation of iron oxide nanoparticles synthesized by a one pot process for intravenous iron supply. RSC Adv 2016. [DOI: 10.1039/c5ra25729h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This paper reports a new synthesis method to control the size of iron oxide nanoparticles (IONs) by adding sodium citrate during fabrication to obtain sodium citrate-modified iron oxide nanoparticles (SCIONs).
Collapse
Affiliation(s)
- Juanjuan Li
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety
- National Center for NanoScience and Technology
- Beijing 100190
- China
- School of Chemical Engineering and Material Science
| | - Yang Liu
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety
- National Center for NanoScience and Technology
- Beijing 100190
- China
- School of Chemical Engineering and Material Science
| | - Ruitao Cha
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety
- National Center for NanoScience and Technology
- Beijing 100190
- China
| | - Bei Ran
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety
- National Center for NanoScience and Technology
- Beijing 100190
- China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy
| | - Kaiwen Mou
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety
- National Center for NanoScience and Technology
- Beijing 100190
- China
- College of Material Science and Engineering
| | - Huashan Wang
- School of Chemical Engineering and Material Science
- Tianjin University of Science and Technology
- Tianjin 300457
- China
| | - Qian Xie
- Division of Nephrology
- Peking University Third Hospital
- Beijing 100191
- China
| | - Jiashu Sun
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety
- National Center for NanoScience and Technology
- Beijing 100190
- China
| | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety
- National Center for NanoScience and Technology
- Beijing 100190
- China
| |
Collapse
|
34
|
Dolci S, Domenici V, Vidili G, Orecchioni M, Bandiera P, Madeddu R, Farace C, Peana M, Tiné MR, Manetti R, Sgarrella F, Delogu LG. Immune compatible cystine-functionalized superparamagnetic iron oxide nanoparticles as vascular contrast agents in ultrasonography. RSC Adv 2016. [DOI: 10.1039/c5ra19652c] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been investigated for biomedical applications.
Collapse
Affiliation(s)
- Sara Dolci
- Department of Chemistry and Industrial Chemistry
- University of Pisa
- 56124 Pisa
- Italy
| | - Valentina Domenici
- Department of Chemistry and Industrial Chemistry
- University of Pisa
- 56124 Pisa
- Italy
| | - Gianpaolo Vidili
- Department of Clinical and Experimental Medicine
- University of Sassari
- 07100 Sassari
- Italy
| | - Marco Orecchioni
- Department of Chemistry and Pharmacy
- University of Sassari
- 07100 Sassari
- Italy
| | - Pasquale Bandiera
- Department of Biomedical Sciences
- University of Sassari
- 07100 Sassari
- Italy
| | - Roberto Madeddu
- Department of Biomedical Sciences
- University of Sassari
- 07100 Sassari
- Italy
| | - Cristiano Farace
- Department of Biomedical Sciences
- University of Sassari
- 07100 Sassari
- Italy
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy
- University of Sassari
- 07100 Sassari
- Italy
| | - Maria Rosaria Tiné
- Department of Chemistry and Industrial Chemistry
- University of Pisa
- 56124 Pisa
- Italy
| | - Roberto Manetti
- Department of Clinical and Experimental Medicine
- University of Sassari
- 07100 Sassari
- Italy
| | | | - Lucia Gemma Delogu
- Department of Chemistry and Pharmacy
- University of Sassari
- 07100 Sassari
- Italy
| |
Collapse
|
35
|
Abstract
Nanoparticles (NPs) present in the environment and in consumer products can cause immunotoxic effects. The immune system is very complex, and in vivo studies are the gold standard for evaluation. Due to the increased amount of NPs that are being developed, cellular screening assays to decrease the amount of NPs that have to be tested in vivo are highly needed. Effects on the unspecific immune system, such as effects on phagocytes, might be suitable for screening for immunotoxicity because these cells mediate unspecific and specific immune responses. They are present at epithelial barriers, in the blood, and in almost all organs. This review summarizes the effects of carbon, metal, and metal oxide NPs used in consumer and medical applications (gold, silver, titanium dioxide, silica dioxide, zinc oxide, and carbon nanotubes) and polystyrene NPs on the immune system. Effects in animal exposures through different routes are compared to the effects on isolated phagocytes. In addition, general problems in the testing of NPs, such as unknown exposure doses, as well as interference with assays are mentioned. NPs appear to induce a specific immunotoxic pattern consisting of the induction of inflammation in normal animals and aggravation of pathologies in disease models. The evaluation of particle action on several phagocyte functions in vitro may provide an indication on the potency of the particles to induce immunotoxicity in vivo. In combination with information on realistic exposure levels, in vitro studies on phagocytes may provide useful information on the health risks of NPs.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Graz, Austria
| |
Collapse
|
36
|
In vivo analysis of neuroinflammation in the late chronic phase after experimental stroke. Neuroscience 2015; 292:71-80. [PMID: 25701708 DOI: 10.1016/j.neuroscience.2015.02.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND PURPOSE In vivo imaging of inflammatory processes is a valuable tool in stroke research. We here investigated the combination of two imaging modalities in the chronic phase after cerebral ischemia: magnetic resonance imaging (MRI) using intravenously applied ultra small supraparamagnetic iron oxide particles (USPIO), and positron emission tomography (PET) with the tracer [(11)C]PK11195. METHODS Rats were subjected to permanent middle cerebral artery occlusion (pMCAO) by the macrosphere model and monitored by MRI and PET for 28 or 56 days, followed by immunohistochemical endpoint analysis. To our knowledge, this is the first study providing USPIO-MRI data in the chronic phase up to 8 weeks after stroke. RESULTS Phagocytes with internalized USPIOs induced MRI-T2(∗) signal alterations in the brain. Combined analysis with [(11)C]PK11195-PET allowed quantification of phagocytic activity and other neuroinflammatory processes. From 4 weeks after induction of ischemia, inflammation was dominated by phagocytes. Immunohistochemistry revealed colocalization of Iba1+ microglia with [(11)C]PK11195 and ED1/CD68 with USPIOs. USPIO-related iron was distinguished from alternatively deposited iron by assessing MRI before and after USPIO application. Tissue affected by non-phagocytic inflammation during the first week mostly remained in a viably vital but remodeled state after 4 or 8 weeks, while phagocytic activity was associated with severe injury and necrosis accordingly. CONCLUSIONS We conclude that the combined approach of USPIO-MRI and [(11)C]PK11195-PET allows to observe post-stroke inflammatory processes in the living animal in an intraindividual and longitudinal fashion, predicting long-term tissue fate. The non-invasive imaging methods do not affect the immune system and have been applied to human subjects before. Translation into clinical applications is therefore feasible.
Collapse
|
37
|
Couto D, Sousa R, Andrade L, Leander M, Lopez-Quintela MA, Rivas J, Freitas P, Lima M, Porto G, Porto B, Carvalho F, Fernandes E. Polyacrylic acid coated and non-coated iron oxide nanoparticles are not genotoxic to human T lymphocytes. Toxicol Lett 2015; 234:67-73. [PMID: 25683033 DOI: 10.1016/j.toxlet.2015.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/31/2015] [Accepted: 02/11/2015] [Indexed: 12/17/2022]
Abstract
The use of iron oxide nanoparticles (ION) for diagnostic and therapeutic purposes requires a clear favorable risk-benefit ratio. This work was performed with the aim of studying the ability of polyacrylic acid (PAA)-coated and non-coated ION to induce genotoxicity in human T lymphocytes. For that purpose, their influence on cell cycle progression and on the induction of chromosome aberrations was evaluated. Blood samples collected from healthy human donors were exposed to PAA-coated and non-coated ION, at different concentrations, for 48h. The obtained results showed that, for all culture conditions, the tested ION are not genotoxic and do not influence the cell cycle arrest. Their possible cumulative effect with the iron-dependent genotoxic agent BLM was also evaluated. Blood samples collected from healthy human donors were exposed to ION, at different concentrations, for 48h, in the presence of a pre-determined toxic concentration of BLM. The obtained results showed that, for all culture conditions, the tested ION do not potentiate the clastogenic effects of BLM.
Collapse
Affiliation(s)
- Diana Couto
- UCIBIO/REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Rosa Sousa
- Laboratory of Cytogenetics, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Lara Andrade
- Laboratory of Cytogenetics, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Magdalena Leander
- Service of Clinical Hematology, Santo António Hospital, Porto, Portugal
| | - M Arturo Lopez-Quintela
- Laboratory of Nanotechnology and Magnetism, Institute of Technological Research, IIT, University of Santiago de Compostela (USC), Spain
| | - José Rivas
- International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Paulo Freitas
- International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Margarida Lima
- Service of Clinical Hematology, Santo António Hospital, Porto, Portugal
| | - Graça Porto
- Service of Clinical Hematology, Santo António Hospital, Porto, Portugal
| | - Beatriz Porto
- Laboratory of Cytogenetics, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Félix Carvalho
- UCIBIO/REQUIMTE Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| | - Eduarda Fernandes
- UCIBIO/REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| |
Collapse
|
38
|
Kim HS, Woo J, Choi Y, Hwang EH, Choi SK, Cho KW, Moon WK. Noninvasive MRI and multilineage differentiation capability of ferritin-transduced human mesenchymal stem cells. NMR IN BIOMEDICINE 2015; 28:168-179. [PMID: 25448225 DOI: 10.1002/nbm.3236] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 10/14/2014] [Accepted: 10/25/2014] [Indexed: 06/04/2023]
Abstract
Molecular imaging can be a breakthrough tool for the investigation of the behavior and ultimate feasibility of transplanted human mesenchymal stem cells (hMSCs) inside the body, and for the development of guidelines and recommendations based on the treatment and evaluation of stem cell therapy for patients. The goals of this study were to evaluate the multilineage differentiation ability of hMSCs expressing an MRI reporter, human ferritin heavy chain (FTH) and to investigate the feasibility of using FTH-based MRI to provide noninvasive imaging of transplanted hMSCs. The transduction of FTH and green fluorescence protein (GFP) did not influence the expression of the mesenchymal stem cell surface markers (CD29+/CD105+/CD34-/CD45-) or the self-renewal marker genes [octamer-binding transcription factor 4 (OCT-4) and SRY (sex determining region Y)-box 2 (Sox-2)], cell viability, migration ability and the release of cytokines [interleukin-5 (IL-5), IL-10, IL-12p70, tumor necrosis factor-α (TNF-α)]. FTH-hMSCs retained the capacity to differentiate into adipogenic, chondrogenic, osteogenic and neurogenic lineages. The transduction of FTH led to a significant enhancement in cellular iron storage capacity and caused hypointensity and a significant increase in R2 * values of FTH-hMSC-collected phantoms and FTH-hMSC-transplanted sites of the brain, as shown by in vitro and in vivo MRI performed at 9.4 T, compared with control hMSCs. This study revealed no differences in biological characteristics between hMSCs and FTH-hMSCs and, therefore, these cells could be used for noninvasive monitoring with MRI during stem cell therapy for brain injury. Our study suggests the use of FTH for in vivo long-term tracking and ultimate fate of hMSCs without alteration of their characteristics and multidifferentiation potential.
Collapse
Affiliation(s)
- Hoe Suk Kim
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
39
|
Doyle KP, Quach LN, Arceuil HED, Buckwalter MS. Ferumoxytol administration does not alter infarct volume or the inflammatory response to stroke in mice. Neurosci Lett 2014; 584:236-40. [PMID: 25449870 DOI: 10.1016/j.neulet.2014.10.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/16/2014] [Accepted: 10/23/2014] [Indexed: 10/24/2022]
Abstract
Ferumoxytol is an ultrasmall superparamagnetic iron oxide (USPIO) nanoparticle that is FDA-approved as an intravenous iron replacement therapy for the treatment of iron deficiency anemia in patients with chronic kidney disease. Ferumoxytol has also been used as a contrast agent for cerebral blood volume mapping by magnetic resonance imaging (MRI), which suggests it could be used for imaging hemodynamic abnormalities after stroke. However, circulating macrophages can internalize USPIOs, and recent data indicate that the accumulation of iron in macrophages can lead them to adopt the M1 pro-inflammatory phenotype. Therefore, the uptake of intravenously administered iron particles by circulating macrophages that home to the stroke core could potentially alter the inflammatory response to stroke. To test this possibility in vivo we administered a dose of ferumoxytol previously used to obtain cerebral blood volume maps in healthy humans by steady-state susceptibility contrast (SSC) MRI to BALB/cJ mice 48h after stroke and examined cytokine levels, microglial/macrophage activation, and lesion volume in the brain 5 days later. Treatment with ferumoxytol did not lead to any differences in these parameters. These data indicate that the use of ferumoxytol as a contrast agent for brain imaging after stroke does not alter the inflammatory response to stroke in mice, and is therefore unlikely to do so in human subjects.
Collapse
Affiliation(s)
- Kristian P Doyle
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Department of Immunobiology, Department of Neurology, and the Arizona Center on Aging, University of Arizona, Tucson, AZ, USA
| | - Lisa N Quach
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen E D' Arceuil
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
40
|
Brown DM, Johnston H, Gubbins E, Stone V. Serum enhanced cytokine responses of macrophages to silica and iron oxide particles and nanomaterials: a comparison of serum to lung lining fluid and albumin dispersions. J Appl Toxicol 2014; 34:1177-87. [DOI: 10.1002/jat.2998] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/29/2014] [Accepted: 01/29/2014] [Indexed: 12/14/2022]
Affiliation(s)
- David M. Brown
- School of Life sciences; Heriot-Watt University; Riccarton Edinburgh EH14 4AS UK
| | - Helinor Johnston
- School of Life sciences; Heriot-Watt University; Riccarton Edinburgh EH14 4AS UK
| | - Eva Gubbins
- School of Life sciences; Heriot-Watt University; Riccarton Edinburgh EH14 4AS UK
| | - Vicki Stone
- School of Life sciences; Heriot-Watt University; Riccarton Edinburgh EH14 4AS UK
| |
Collapse
|
41
|
Clemente-Casares X, Santamaria P. Nanomedicine in autoimmunity. Immunol Lett 2014; 158:167-74. [PMID: 24406504 DOI: 10.1016/j.imlet.2013.12.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/09/2013] [Accepted: 12/20/2013] [Indexed: 11/15/2022]
Abstract
The application of nanotechnology to the diagnosis and therapy of human diseases is already a reality and is causing a real revolution in how we design new therapies and vaccines. In this review we focus on the applications of nanotechnology in the field of autoimmunity. First, we review scenarios in which iron oxide nanoparticles have been used in the diagnosis of autoimmune diseases, mostly through magnetic resonance imaging (MRI), both in animal models and patients. Second, we discuss the potential of nanoparticles as an immunotherapeutic platform for autoimmune diseases, for now exclusively in pre-clinical models. Finally, we discuss the potential of this field to generate the 'perfect drug' with the capacity to report on its therapeutic efficacy in real time, that is, the birth of theranostics in autoimmunity.
Collapse
Affiliation(s)
- Xavier Clemente-Casares
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada; Institut D'Investigacions Biomediques August Pi i Sunyer, Barcelona, Spain.
| |
Collapse
|
42
|
Prietl B, Meindl C, Roblegg E, Pieber TR, Lanzer G, Fröhlich E. Nano-sized and micro-sized polystyrene particles affect phagocyte function. Cell Biol Toxicol 2013; 30:1-16. [PMID: 24292270 DOI: 10.1007/s10565-013-9265-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/15/2013] [Indexed: 12/22/2022]
Abstract
Adverse effect of nanoparticles may include impairment of phagocyte function. To identify the effect of nanoparticle size on uptake, cytotoxicity, chemotaxis, cytokine secretion, phagocytosis, oxidative burst, nitric oxide production and myeloperoxidase release, leukocytes isolated from human peripheral blood, monocytes and macrophages were studied. Carboxyl polystyrene (CPS) particles in sizes between 20 and 1,000 nm served as model particles. Twenty nanometers CPS particles were taken up passively, while larger CPS particles entered cells actively and passively. Twenty nanometers CPS were cytotoxic to all phagocytes, ≥500 nm CPS particles only to macrophages. Twenty nanometers CPS particles stimulated IL-8 secretion in human monocytes and induced oxidative burst in monocytes. Five hundred nanometers and 1,000 nm CPS particles stimulated IL-6 and IL-8 secretion in monocytes and macrophages, chemotaxis towards a chemotactic stimulus of monocytes and phagocytosis of bacteria by macrophages and provoked an oxidative burst of granulocytes. At very high concentrations, CPS particles of 20 and 500 nm stimulated myeloperoxidase release of granulocytes and nitric oxide generation in macrophages. Cytotoxic effect could contribute to some of the observed effects. In the absence of cytotoxicity, 500 and 1,000 nm CPS particles appear to influence phagocyte function to a greater extent than particles in other sizes.
Collapse
Affiliation(s)
- B Prietl
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | | | | | | | | | | |
Collapse
|
43
|
Tamames-Tabar C, Cunha D, Imbuluzqueta E, Ragon F, Serre C, Blanco-Prieto MJ, Horcajada P. Cytotoxicity of nanoscaled metal-organic frameworks. J Mater Chem B 2013; 2:262-271. [PMID: 32261505 DOI: 10.1039/c3tb20832j] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A series of fourteen porous Metal-Organic Frameworks (MOFs) with different compositions (Fe, Zn, and Zr; carboxylates or imidazolates) and structures have been successfully synthesised at the nanoscale and fully characterised by XRPD, FTIR, TGA, N2 porosimetry, TEM, DLS and ζ-potential. Their toxicological assessment was performed using two different cell lines: human epithelial cells from foetal cervical carcinoma (HeLa) and murine macrophage cell line (J774). It appears that MOF nanoparticles (NPs) exhibit low cytotoxicity, comparable to those of other commercialised nanoparticulate systems, the less toxic being the Fe carboxylate and the more toxic being the zinc imidazolate NPs. The cytotoxicity values, higher in J774 cells than in HeLa cells, are mainly function of their composition and cell internalisation capacity. Finally, cell uptake of one of the most relevant Fe-MOF-NPs for drug vectorisation has been investigated by confocal microscopy studies, and indicates a faster kinetics of cell penetration within J774 compared to HeLa cells.
Collapse
Affiliation(s)
- Cristina Tamames-Tabar
- Institut Lavoisier, UMR CNRS 8180, Université de Versailles Saint-Quentin-en-Yvelines, 45 Avenue des Etats-Unis, 78035 Versailles Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
44
|
Wu HY, Chung MC, Wang CC, Huang CH, Liang HJ, Jan TR. Iron oxide nanoparticles suppress the production of IL-1beta via the secretory lysosomal pathway in murine microglial cells. Part Fibre Toxicol 2013; 10:46. [PMID: 24047432 PMCID: PMC3851143 DOI: 10.1186/1743-8977-10-46] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 09/09/2013] [Indexed: 11/17/2022] Open
Abstract
Background Superparamagnetic iron oxide nanoparticles (IONPs) have been used as magnetic resonance imaging contrast agents for various research and diagnostic purposes, such as the detection of neuroinflammation and blood-brain-barrier integrity. As the central resident macrophage-like cells, microglia are responsible for managing foreign agents invading the CNS. The present study investigated the direct effect of IONPs on the production of pro-inflammatory cytokines by murine microglia stimulated with lipopolysaccharide (LPS). Methods Primary murine microglial cells were pretreated with IONPs (1–50 μg Fe/mL) for 30 min and then stimulated with LPS (100 ng/mL) for 24 h. Confocal microscopy is used to visualize the intracellular IONP distribution and secretory lysosomes after staining with LysoTracker and Rab27a, respectively. The production of interleukin (IL)-1β and tumor necrosis factor (TNF)-α was quantified by ELISA. The activity of IL-1β converting enzyme (ICE) and TNF-α converting enzyme (TACE) was measured by fluorescent microplate assay using specific substrates. The lysosomal number, alkalinity, permeability and cathepsin B activity were determined by flow cytometry with ectodermal dysplasia-1, lysosensor and acridine orange staining, and using cathepsin B specific substrate, respectively. Results Confocal imaging revealed that IONPs were markedly engulfed by microglia. Exposure to IONPs attenuated the production of IL-1β, but not TNF-α. Concordantly, the activity of ICE, but not the TACE, was suppressed in IONP-treated cells. Mechanistic studies showed that IONPs accumulated in lysosomes and the number of lysosomes was increased in IONP-treated cells. In addition, exposure to IONPs increased lysosomal permeability and alkalinity, but decreased the activity of cathepsin B, a secretory lysosomal enzyme involved in the activation of ICE. Conclusions Our results demonstrated a contrasting effect of IONPs on the production of IL-1β and TNF-α by LPS-stimulated microglia, in which the attenuation of IL-1β by IONPs was mediated by inhibiting the secretory lysosomal pathway of cytokine processing.
Collapse
Affiliation(s)
- Hsin-Ying Wu
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, No,1, Sec, 4, Roosevelt Road, Taipei 10617, Taiwan.
| | | | | | | | | | | |
Collapse
|
45
|
Triboulet S, Aude-Garcia C, Carrière M, Diemer H, Proamer F, Habert A, Chevallet M, Collin-Faure V, Strub JM, Hanau D, Van Dorsselaer A, Herlin-Boime N, Rabilloud T. Molecular responses of mouse macrophages to copper and copper oxide nanoparticles inferred from proteomic analyses. Mol Cell Proteomics 2013; 12:3108-22. [PMID: 23882024 DOI: 10.1074/mcp.m113.030742] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The molecular responses of macrophages to copper-based nanoparticles have been investigated via a combination of proteomic and biochemical approaches, using the RAW264.7 cell line as a model. Both metallic copper and copper oxide nanoparticles have been tested, with copper ion and zirconium oxide nanoparticles used as controls. Proteomic analysis highlighted changes in proteins implicated in oxidative stress responses (superoxide dismutases and peroxiredoxins), glutathione biosynthesis, the actomyosin cytoskeleton, and mitochondrial proteins (especially oxidative phosphorylation complex subunits). Validation studies employing functional analyses showed that the increases in glutathione biosynthesis and in mitochondrial complexes observed in the proteomic screen were critical to cell survival upon stress with copper-based nanoparticles; pharmacological inhibition of these two pathways enhanced cell vulnerability to copper-based nanoparticles, but not to copper ions. Furthermore, functional analyses using primary macrophages derived from bone marrow showed a decrease in reduced glutathione levels, a decrease in the mitochondrial transmembrane potential, and inhibition of phagocytosis and of lipopolysaccharide-induced nitric oxide production. However, only a fraction of these effects could be obtained with copper ions. In conclusion, this study showed that macrophage functions are significantly altered by copper-based nanoparticles. Also highlighted are the cellular pathways modulated by cells for survival and the exemplified cross-toxicities that can occur between copper-based nanoparticles and pharmacological agents.
Collapse
Affiliation(s)
- Sarah Triboulet
- Pro-MD team, Laboratoire de Chimie et Biologie des Métaux, UMR CNRS-CEA-UJF, Université Joseph Fourier, Grenoble 38054, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bernsen MR, Ruggiero A, van Straten M, Kotek G, Haeck JC, Wielopolski PA, Krestin GP. Computed tomography and magnetic resonance imaging. Recent Results Cancer Res 2013. [PMID: 23179877 DOI: 10.1007/978-3-642-10853-2_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Imaging in Oncology is rapidly moving from the detection and size measurement of a lesion to the quantitative assessment of metabolic processes and cellular and molecular interactions. Increasing insights into cancer as a complex disease with involvement of the tumor stroma in tumor pathobiological processes have made it clear that for successful control of cancer, treatment strategies should not only be directed at the tumor cells but also targeted at the tumor microenvironment. This requires understanding of the complex molecular and cellular interactions in cancer tissue. Recent developments in imaging technology have increased the possibility to image various pathobiological processes in cancer development and response to treatment. For computed tomography (CT) and magnetic resonance imaging (MRI) various improvements in hardware, software, and imaging probes have lifted these modalities from classical anatomical imaging techniques to techniques suitable to image and quantify various physiological processes and molecular and cellular interactions. Next to a more general overview of possible imaging targets in oncology this chapter provides an overview of the various developments in CT and MRI technology and some specific applications.
Collapse
Affiliation(s)
- Monique R Bernsen
- Department of Radiology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
47
|
Tourdias T, Dousset V. Neuroinflammatory imaging biomarkers: relevance to multiple sclerosis and its therapy. Neurotherapeutics 2013; 10:111-23. [PMID: 23132327 PMCID: PMC3557362 DOI: 10.1007/s13311-012-0155-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Magnetic resonance imaging is an established tool in the management of multiple sclerosis (MS). Loss of blood brain barrier integrity assessed by gadolinium (Gd) enhancement is the current standard marker of MS activity. To explore the complex cascade of the inflammatory events, other magnetic resonance imaging, but also positron emission tomographic markers reviewed in this article are being developed to address active neuroinflammation with increased sensitivity and specificity. Alternative magnetic resonance contrast agents, positron emission tomographic tracers and imaging techniques could be more sensitive than Gd to early blood brain barrier alteration, and they could assess the inflammatory cell recruitment and/or the associated edema accumulation. These markers of active neuroinflammation, although some of them are limited to experimental studies, could find great relevance to complete Gd information and thereby increase our understanding of acute lesion pathophysiology and its noninvasive follow-up, especially to monitor treatment efficacy. Furthermore, such accurate markers of inflammation combined with those of neurodegeneration hold promise to provide a more complete picture of MS, which will be of great benefit for future therapeutic strategies.
Collapse
Affiliation(s)
- Thomas Tourdias
- INSERM Unit 1049 Neuroinflammation, Imagerie et Thérapie de la Sclérose en Plaques, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux, F-33076, France.
| | | |
Collapse
|
48
|
Kim HS, Tian L, Lin S, Cha JH, Jung HS, Park KS, Moon WK. Magnetic labeling of pancreaticβ-cells modulates the glucose- and insulin-induced phosphorylation of ERK1/2 and AKT. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 8:20-6. [DOI: 10.1002/cmmi.1490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Hoe Suk Kim
- Department of Radiology; Seoul National University Hospital; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
- Institute of Radiation Medicine, Medical Research Center; Seoul National University; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
| | - Lianji Tian
- Department of Radiology; Seoul National University Hospital; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
- Department of Biomedical Science, College of Medicine; Seoul National University; Seoul, 101 Daehangno, Jongno-gu Seoul 110-744 Korea
| | - Shunmei Lin
- Department of Radiology; Seoul National University Hospital; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
| | - Joo Hee Cha
- Department of Radiology; Seoul National University Hospital; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
| | - Hye Seung Jung
- Department of Internal Medicine; Seoul National University Hospital; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
| | - Kyong Soo Park
- Department of Internal Medicine; Seoul National University Hospital; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
| | - Woo Kyung Moon
- Department of Radiology; Seoul National University Hospital; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
- Institute of Radiation Medicine, Medical Research Center; Seoul National University; 101 Daehangno, Jongno-gu Seoul 110-744 Korea
- Department of Biomedical Science, College of Medicine; Seoul National University; Seoul, 101 Daehangno, Jongno-gu Seoul 110-744 Korea
| |
Collapse
|
49
|
Impact of Nanomaterials on Health and Environment. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2012. [DOI: 10.1007/s13369-012-0324-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Shen CC, Liang HJ, Wang CC, Liao MH, Jan TR. Iron oxide nanoparticles suppressed T helper 1 cell-mediated immunity in a murine model of delayed-type hypersensitivity. Int J Nanomedicine 2012; 7:2729-37. [PMID: 22701318 PMCID: PMC3373298 DOI: 10.2147/ijn.s31054] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background It was recently reported that iron oxide nanoparticles attenuated antigen-specific humoral responses and T cell cytokine expression in ovalbumin-sensitized mice. It is presently unclear whether iron oxide nanoparticles influence T helper 1 cell-mediated immunity. The present study aimed to investigate the effect of iron oxide nanoparticles on delayed-type hypersensitivity (DTH), whose pathophysiology requires the participation of T helper 1 cells and macrophages. Methods DTH was elicited by a subcutaneous challenge with ovalbumin to the footpads of mice sensitized with ovalbumin. Iron oxide nanoparticles (0.2–10 mg iron/kg) were administered intravenously 1 hour prior to ovalbumin sensitization. Local inflammatory responses were examined by footpad swelling and histological analysis. The expression of cytokines by splenocytes was measured by enzyme-linked immunosorbent assay. Results Administration of iron oxide nanoparticles, in a dose-dependent fashion, significantly attenuated inflammatory reactions associated with DTH, including the footpad swelling, the infiltration of T cells and macrophages, and the expression of interferon-γ, interleukin-6, and tumor necrosis factor-α in the inflammatory site. Iron oxide nanoparticles also demonstrated a suppressive effect on ovalbumin-stimulated production of interferon-γ by splenocytes and the phagocytic activity of splenic CD11b+ cells. Conclusion These results demonstrated that a single dose of iron oxide nanoparticles attenuated DTH reactions by suppressing the infiltration and functional activity of T helper 1 cells and macrophages in response to antigen stimulation.
Collapse
Affiliation(s)
- Chien-Chang Shen
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei
| | | | | | | | | |
Collapse
|