1
|
Ammassam Veettil R, Sebastian S, McCallister T, Ghosh S, Hynds DL. Uptake of surface-functionalized thermo-responsive polymeric nanocarriers in corticospinal tract motor neurons. Biochem Biophys Res Commun 2024; 696:149503. [PMID: 38262309 DOI: 10.1016/j.bbrc.2024.149503] [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: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024]
Abstract
Nanocarrier drug delivery systems are attractive options for targeted delivery of survival- and regeneration-enhancing therapeutics to neurons damaged by degenerative or traumatic central nervous system (CNS) lesions. Functional groups on nanocarrier surfaces allow derivatization with molecules to target specific cells but may affect cellular interactions and nanocarrier uptake. We synthesized differently sized -COOH and -NH2 surface functionalized polymeric nanocarriers (SFNCs) by emulsion copolymerization and assessed uptake by different cell types in mixed cortical cultures. Following 60-min incubation with SFNCs, mean intensity measurements of fluorescently labeled SFNCs indicated that corticospinal tract motor neurons (CSMNs) took up more COOH- or NH2- functionalized SFNCs with similar sizes (150 nm), compared to glia. However, larger diameter (750 nm) SFNCs were taken up at higher concentrations compared to smaller COOH-derivatized SFNCs (150 nm). These data suggest that larger SFNCs may provide an advantage for enhanced uptake by targeted neurons.
Collapse
Affiliation(s)
- Remya Ammassam Veettil
- Division of Biology, Texas Woman's University, 1000 Old Main Circle, Denton, TX, 76204, USA.
| | - Sumod Sebastian
- Division of Biology, Texas Woman's University, 1000 Old Main Circle, Denton, TX, 76204, USA.
| | - Thomas McCallister
- Department of Engineering and Technology, Southeast Missouri State University, Cape Girardeau, One University Plaza, MO, 63701, USA
| | - Santaneel Ghosh
- Department of Engineering and Technology, Southeast Missouri State University, Cape Girardeau, One University Plaza, MO, 63701, USA
| | - DiAnna L Hynds
- Division of Biology, Texas Woman's University, 1000 Old Main Circle, Denton, TX, 76204, USA.
| |
Collapse
|
2
|
Irrsack E, Aydin S, Bleckmann K, Schuller J, Dringen R, Koch M. Local Administrations of Iron Oxide Nanoparticles in the Prefrontal Cortex and Caudate Putamen of Rats Do Not Compromise Working Memory and Motor Activity. Neurotox Res 2023; 42:6. [PMID: 38133743 PMCID: PMC10746586 DOI: 10.1007/s12640-023-00684-x] [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: 08/22/2023] [Revised: 11/10/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Iron oxide nanoparticles (IONPs) have come into focus for their use in medical applications although possible health risks for humans, especially in terms of brain functions, have not yet been fully clarified. The present study investigates the effects of IONPs on neurobehavioural functions in rats. For this purpose, we infused dimercaptosuccinic acid-coated IONPs into the medial prefrontal cortex (mPFC) and caudate putamen (CPu). Saline (VEH) and ferric ammonium citrate (FAC) were administered as controls. One- and 4-week post-surgery mPFC-infused animals were tested for their working memory performance in the delayed alternation T-maze task and in the open field (OF) for motor activity, and CPu-infused rats were tested for their motor activity in the OF. After completion of the experiments, the brains were examined histologically and immunohistochemically. We did not observe any behavioural or structural abnormalities in the rats after administration of IONPs in the mPFC and the CPu. In contrast, administration of FAC into the CPu resulted in decreased motor activity and increased the number of microglia in the mPFC. Perls' Prussian blue staining revealed that FAC- and IONP-treated rats had more iron-containing ramified cells than VEH-treated rats, indicating iron uptake by microglia. Our results demonstrate that local infusions of IONPs into selected brain regions have no adverse impact on locomotor behaviour and working memory.
Collapse
Affiliation(s)
- Ellen Irrsack
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany.
| | - Sidar Aydin
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| | - Katja Bleckmann
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| | - Julia Schuller
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen (CBIB), and Centre for Environmental Research and Sustainable, Technology, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| | - Michael Koch
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| |
Collapse
|
3
|
Bechinger P, Serrano Sponton L, Grützner V, Musyanovych A, Jussen D, Krenzlin H, Eldahaby D, Riede N, Kempski O, Ringel F, Alessandri B. In-vivo time course of organ uptake and blood-brain-barrier permeation of poly(L-lactide) and poly(perfluorodecyl acrylate) nanoparticles with different surface properties in unharmed and brain-traumatized rats. Front Neurol 2023; 14:994877. [PMID: 36814997 PMCID: PMC9939480 DOI: 10.3389/fneur.2023.994877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/20/2023] [Indexed: 02/08/2023] Open
Abstract
Background Traumatic brain injury (TBI) has a dramatic impact on mortality and quality of life and the development of effective treatment strategies is of great socio-economic relevance. A growing interest exists in using polymeric nanoparticles (NPs) as carriers across the blood-brain barrier (BBB) for potentially effective drugs in TBI. However, the effect of NP material and type of surfactant on their distribution within organs, the amount of the administrated dose that reaches the brain parenchyma in areas with intact and opened BBB after trauma, and a possible elicited inflammatory response are still to be clarified. Methods The organ distribution, BBB permeation and eventual inflammatory activation of polysorbate-80 (Tw80) and sodiumdodecylsulfate (SDS) stabilized poly(L-lactide) (PLLA) and poly(perfluorodecyl acrylate) (PFDL) nanoparticles were evaluated in rats after intravenous administration. The NP uptake into the brain was assessed under intact conditions and after controlled cortical impact (CCI). Results A significantly higher NP uptake at 4 and 24 h after injection was observed in the liver and spleen, followed by the brain and kidney, with minimal concentrations in the lungs and heart for all NPs. A significant increase of NP uptake at 4 and 24 h after CCI was observed within the traumatized hemisphere, especially in the perilesional area, but NPs were still found in areas away from the injury site and the contralateral hemisphere. NPs were internalized in brain capillary endothelial cells, neurons, astrocytes, and microglia. Immunohistochemical staining against GFAP, Iba1, TNFα, and IL1β demonstrated no glial activation or neuroinflammatory changes. Conclusions Tw80 and SDS coated biodegradable PLLA and non-biodegradable PFDL NPs reach the brain parenchyma with and without compromised BBB by TBI, even though a high amount of NPs are retained in the liver and spleen. No inflammatory reaction is elicited by these NPs within 24 h after injection. Thus, these NPs could be considered as potentially effective carriers or markers of newly developed drugs with low or even no BBB permeation.
Collapse
Affiliation(s)
- Patrick Bechinger
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, Mainz, Germany,Department of Anesthesiology, Helios Dr. Horst Schmidt Clinic, Wiesbaden, Germany
| | - Lucas Serrano Sponton
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, Mainz, Germany,Department of Neurosurgery, Sana Clinic Offenbach, Offenbach, Germany,*Correspondence: Lucas Serrano Sponton ✉
| | - Verena Grützner
- Fraunhofer Institute for Microengineering and Microsystems, Mainz, Germany
| | - Anna Musyanovych
- Fraunhofer Institute for Microengineering and Microsystems, Mainz, Germany
| | - Daniel Jussen
- Department of Neurosurgery, Johann Wolfgang Goethe University Frankfurt am Main, Frankfurt, Germany
| | - Harald Krenzlin
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, Mainz, Germany
| | - Daniela Eldahaby
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, Mainz, Germany,San Paolo Medical School, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nicole Riede
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, Mainz, Germany
| | - Oliver Kempski
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, Mainz, Germany
| | - Florian Ringel
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, Mainz, Germany
| | - Beat Alessandri
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, Mainz, Germany
| |
Collapse
|
4
|
Erythropoietin Nanobots: Their Feasibility for the Controlled Release of Erythropoietin and Their Neuroprotective Bioequivalence in Central Nervous System Injury. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12073351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Erythropoietin (EPO) plays important roles in neuroprotection in central nervous system injury. Due to the limited therapeutic time window and coexistence of hematopoietic/extrahematopoietic receptors displaying heterogenic and phylogenetic differences, fast, targeted delivery agents, such as nanobots, are needed. To confirm the feasibility of EPO-nanobots (ENBs) as therapeutic tools, the authors evaluated controlled EPO release from ENBs and compared the neuroprotective bioequivalence of these substances after preconditioning sonication. Methods: ENBs were manufactured by a nanospray drying technique with preconditioning sonication. SH-SY5Y neuronal cells were cotreated with thapsigargin and either EPO or ENBs before cell viability, EPO receptor activation, and endoplasmic reticulum stress-related pathway deactivation were determined over 24 h. Results: Preconditioning sonication (50–60 kHz) for 1 h increased the cumulative EPO release from the ENBs (84% versus 25% at 24 h). Between EPO and ENBs at 24 h, both neuronal cell viability (both > 65% versus 15% for thapsigargin alone) and the expression of the proapoptotic/apoptotic biomolecular markers JAK2, PDI, PERK, GRP78, ATF6, CHOP, TGF-β, and caspase-3 were nearly the same or similar. Conclusion: ENBs controlled EPO release in vitro after preconditioning sonication, leading to neuroprotection similar to that of EPO at 24 h.
Collapse
|
5
|
Irrsack E, Schuller J, Petters C, Willmann W, Dringen R, Koch M. Effects of Local Administration of Iron Oxide Nanoparticles in the Prefrontal Cortex, Striatum, and Hippocampus of Rats. Neurotox Res 2021; 39:2056-2071. [PMID: 34705254 PMCID: PMC8639550 DOI: 10.1007/s12640-021-00432-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/08/2021] [Accepted: 10/18/2021] [Indexed: 10/26/2022]
Abstract
Iron oxide nanoparticles (IONPs) are used for diverse medical approaches, although the potential health risks, for example adverse effects on brain functions, are not fully clarified. Several in vitro studies demonstrated that the different types of brain cells are able to accumulate IONPs and reported a toxic potential for IONPs, at least for microglia. However, little information is available for the in vivo effects of direct application of IONPs into the brain over time. Therefore, we examined the cellular responses and the distribution of iron in the rat brain at different time points after local infusion of IONPs into selected brain areas. Dispersed IONPs or an equivalent amount of low molecular weight iron complex ferric ammonium citrate or vehicle were infused into the medial prefrontal cortex (mPFC), the caudate putamen (CPu), or the dorsal hippocampus (dHip). Rats were sacrificed 1 day, 1 week, or 4 weeks post-infusion and brain sections were histologically examined for treatment effects on astrocytes, microglia, and neurons. Glial scar formation was observed in the mPFC and CPu 1 week post-infusion independent of the substance and probably resulted from the infusion procedure. Compared to vehicle, IONPs did not cause any obvious additional adverse effects and no additional tissue damage, while the infusion of ferric ammonium citrate enhanced neurodegeneration in the mPFC. Results of iron staining indicate that IONPs were mainly accumulated in microglia. Our results demonstrate that local infusions of IONPs in selected brain areas do not cause any additional adverse effects or neurodegeneration compared to vehicle.
Collapse
Affiliation(s)
- Ellen Irrsack
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, 28334, Bremen, Germany.
| | - Julia Schuller
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, 28334, Bremen, Germany
| | - Charlotte Petters
- Centre for Biomolecular Interactions Bremen (CBIB), and Centre for Environmental Research and Sustainable Technology, University of Bremen, PO Box 330440, 28334, Bremen, Germany
| | - Wiebke Willmann
- Centre for Biomolecular Interactions Bremen (CBIB), and Centre for Environmental Research and Sustainable Technology, University of Bremen, PO Box 330440, 28334, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen (CBIB), and Centre for Environmental Research and Sustainable Technology, University of Bremen, PO Box 330440, 28334, Bremen, Germany
| | - Michael Koch
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, 28334, Bremen, Germany
| |
Collapse
|
6
|
Keilhoff G, Pinkernelle J, Fansa H. The Ryanodine receptor stabilizer S107 fails to support motor neuronal neuritogenesis in vitro. Tissue Cell 2021; 73:101625. [PMID: 34419737 DOI: 10.1016/j.tice.2021.101625] [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: 06/11/2021] [Revised: 08/02/2021] [Accepted: 08/15/2021] [Indexed: 11/30/2022]
Abstract
Calcium homeostasis is essential for neuronal cell survival/differentiation. Imbalance of the Ca2+ homeostasis due to excessive Ca2+ overload is essential for spinal cord injury (SCI). The overload resulted from Ca2+ flux across the plasma membrane and from internal Ca2+ store release (mitochondria, endoplasmic reticulum, ER). Inositol trisphosphate receptors (IP3R) and ryanodine receptors (RyR) are involved in releasing Ca2+ from ER contributing to axonal degeneration following SCI. In turn, block of both receptors is axoprotective. The calstabin RyR subunit, stabilizing the channel in a state of reduced activity, prevents pathological Ca2+ release too. We investigated whether S107, a RyR-stabilizing compound (Rycal), is beneficial for survival and neuritogenesis of spinal cord motor neurons in vitro. We used a spinal cord slice model and the motor neuron-like NSC-34 cell line. Effects of S107 were tested by propidium iodide/fluorescein diacetate vital staining, mitotic index determination via BrdU-incorporation, and neurite sprouting parameters. Results showed that S107 (i) had no effect on gliosis resulting from slices preparation; (ii) had no effect on motor neuronal survival and proliferation; and (iii) impaired neurite sprouting, no matter whether it was a differentiation (NSC-34 cells) or regeneration (spinal cord slices) process. The results underline the need for a flexible Ca2+homeostasis provided by the ER for re-initiation of neuritogenesis.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, 39120, Magdeburg, Germany.
| | - Josephine Pinkernelle
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, 39120, Magdeburg, Germany
| | - Hisham Fansa
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld, OWL-University, 33604, Bielefeld, Germany; Department of Plastic Surgery, and Breast Centre, Spital Zollikerberg, 8125, Zollikerberg, Switzerland
| |
Collapse
|
7
|
Shin TH, Manavalan B, Lee DY, Basith S, Seo C, Paik MJ, Kim SW, Seo H, Lee JY, Kim JY, Kim AY, Chung JM, Baik EJ, Kang SH, Choi DK, Kang Y, Maral Mouradian M, Lee G. Silica-coated magnetic-nanoparticle-induced cytotoxicity is reduced in microglia by glutathione and citrate identified using integrated omics. Part Fibre Toxicol 2021; 18:42. [PMID: 34819099 PMCID: PMC8614058 DOI: 10.1186/s12989-021-00433-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/25/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Nanoparticles have been utilized in brain research and therapeutics, including imaging, diagnosis, and drug delivery, owing to their versatile properties compared to bulk materials. However, exposure to nanoparticles leads to their accumulation in the brain, but drug development to counteract this nanotoxicity remains challenging. To date, concerns have risen about the potential toxicity to the brain associated with nanoparticles exposure via penetration of the brain blood barrier to address this issue. METHODS Here the effect of silica-coated-magnetic nanoparticles containing the rhodamine B isothiocyanate dye [MNPs@SiO2(RITC)] were assessed on microglia through toxicological investigation, including biological analysis and integration of transcriptomics, proteomics, and metabolomics. MNPs@SiO2(RITC)-induced biological changes, such as morphology, generation of reactive oxygen species, intracellular accumulation of MNPs@SiO2(RITC) using transmission electron microscopy, and glucose uptake efficiency, were analyzed in BV2 murine microglial cells. Each omics data was collected via RNA-sequencing-based transcriptome analysis, liquid chromatography-tandem mass spectrometry-based proteome analysis, and gas chromatography- tandem mass spectrometry-based metabolome analysis. The three omics datasets were integrated and generated as a single network using a machine learning algorithm. Nineteen compounds were screened and predicted their effects on nanotoxicity within the triple-omics network. RESULTS Intracellular reactive oxygen species production, an inflammatory response, and morphological activation of cells were greater, but glucose uptake was lower in MNPs@SiO2(RITC)-treated BV2 microglia and primary rat microglia in a dose-dependent manner. Expression of 121 genes (from 41,214 identified genes), and levels of 45 proteins (from 5918 identified proteins) and 17 metabolites (from 47 identified metabolites) related to the above phenomena changed in MNPs@SiO2(RITC)-treated microglia. A combination of glutathione and citrate attenuated nanotoxicity induced by MNPs@SiO2(RITC) and ten other nanoparticles in vitro and in the murine brain, protecting mostly the hippocampus and thalamus. CONCLUSIONS Combination of glutathione and citrate can be one of the candidates for nanotoxicity alleviating drug against MNPs@SiO2(RITC) induced detrimental effect, including elevation of intracellular reactive oxygen species level, activation of microglia, and reduction in glucose uptake efficiency. In addition, our findings indicate that an integrated triple omics approach provides useful and sensitive toxicological assessment for nanoparticles and screening of drug for nanotoxicity.
Collapse
Affiliation(s)
- Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Balachandran Manavalan
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Da Yeon Lee
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Chan Seo
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon, 57922 Republic of Korea
| | - Man Jeong Paik
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon, 57922 Republic of Korea
| | - Sang-Wook Kim
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Haewoon Seo
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Ju Yeon Lee
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119 Republic of Korea
| | - Jin Young Kim
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119 Republic of Korea
| | - A Young Kim
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Jee Min Chung
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Eun Joo Baik
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Seong Ho Kang
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin-si, Gyeonggi-do 17104 Republic of Korea
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104 Republic of Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, 268 Chungwondaero, Chungju, 27478 Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics, Rutgers Biomedical and Health Sciences, and Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Gwang Lee
- Department of Molecular Science and Technology, Ajou University, Suwon-si, Gyeonggi-do 16499 Republic of Korea
- Department of Physiology, Ajou University School of Medicine, Suwon-si, Gyeonggi-do 16499 Republic of Korea
| |
Collapse
|
8
|
Keilhoff G, Ludwig C, Pinkernelle J, Lucas B. Effects of Gynostemma pentaphyllum on spinal cord motor neurons and microglial cells in vitro. Acta Histochem 2021; 123:151759. [PMID: 34425524 DOI: 10.1016/j.acthis.2021.151759] [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: 04/27/2021] [Revised: 06/25/2021] [Accepted: 07/11/2021] [Indexed: 11/18/2022]
Abstract
The regenerative capability of spinal cord neurons is limited to impossible. Thus, experimental approaches supporting reconstruction/regeneration are in process. This study focused on the evaluation of the protective potency of an extract from Gynostemma pentaphyllum (GP), a plant used in traditional medicine with anti-oxidative and neuroprotective activities, in vitro on organotypic spinal cord cultures, the motor-neuron-like NSC-34 cell line and the microglial cell line BV-2. Organotypic cultures were mechanically stressed by the slicing procedure and the effect of GP on motor neuron survival and neurite sprouting was tested by immunohistochemistry. NSC-34 cells were neuronal differentiated by using special medium. Afterwards, cell survival (propidium iodide/fluorescein diacetate labeling), proliferation (BrdU-incorporation), and neurite sprouting were evaluated. BV-2 cells were stimulated with LPS/interferon γ and subjected to migration assay and nanoparticle uptake. Cell survival, proliferation and the expression pattern of different microglial activation markers (cFOS, iNOS) as well as transcription factors (PPARγ, YB1) were analyzed. In organotypic cultures, high-dose GP supported survival of motor neurons and especially of the neuronal fiber network. Despite reduced neurodegeneration, however, there was a GP-mediated activation of astro- and microglia. In NSC-34 cells, high-dosed GP had degenerative and anti-proliferative effects, but only in normal medium. Moreover, GP supported the neuro-differentiation ability. In BV-2 cells, high-dosed GP was toxic. In lower dosages, GP affected cell survival and proliferation when combined with LPS/interferon γ. Nanoparticle uptake, migration ability, and the transcription factor PPARγ, however, GP affected directly. The data suggest positive effects of GP on injured spinal motor neurons. Moreover, GP activated microglial cells. The dual role of microglia (protective/detrimental) in neurodegenerative processes required further experiments to enhance the knowledge about GP effects. Therefore, a possible clinical use of GP in spinal cord injuries is still a long way off.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany.
| | - Christina Ludwig
- Institute of Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany
| | - Josephine Pinkernelle
- Institute of Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany
| | - Benjamin Lucas
- Dept. of Trauma Surgery, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany
| |
Collapse
|
9
|
Therapeutic Nanoparticles for the Different Phases of Ischemic Stroke. Life (Basel) 2021; 11:life11060482. [PMID: 34073229 PMCID: PMC8227304 DOI: 10.3390/life11060482] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/27/2022] Open
Abstract
Stroke represents the second leading cause of mortality and morbidity worldwide. Ischemic strokes are the most prevalent type of stroke, and they are characterized by a series of pathological events prompted by an arterial occlusion that leads to a heterogeneous pathophysiological response through different hemodynamic phases, namely the hyperacute, acute, subacute, and chronic phases. Stroke treatment is highly reliant on recanalization therapies, which are limited to only a subset of patients due to their narrow therapeutic window; hence, there is a huge need for new stroke treatments. Nonetheless, the vast majority of promising treatments are not effective in the clinical setting due to their inability to cross the blood-brain barrier and reach the brain. In this context, nanotechnology-based approaches such as nanoparticle drug delivery emerge as the most promising option. In this review, we will discuss the current status of nanotechnology in the setting of stroke, focusing on the diverse available nanoparticle approaches targeted to the different pathological and physiological repair mechanisms involved in each of the stroke phases.
Collapse
|
10
|
Tosat-Bitrián C, Avis-Bodas A, Porras G, Borrego-Hernández D, García-Redondo A, Martín-Requero A, Palomo V. CdSe Quantum Dots in Human Models Derived from ALS Patients: Characterization, Nuclear Penetration Studies and Multiplexing. NANOMATERIALS 2021; 11:nano11030671. [PMID: 33803158 PMCID: PMC7998605 DOI: 10.3390/nano11030671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/25/2022]
Abstract
CdSe quantum dots (QDs) are valuable tools for deciphering molecular mechanisms in cells. Their conjugation with antibodies offers a unique staining source with optimal characteristics, including increased photostability and narrow emission spectra, allowing for improved multiplexing capabilities using a single excitation source. In combination with pathology models derived from patients, they have great potential to contribute to quantitative molecular profiling and promote personalized medicine. However, the commercial availability of diverse CdSe QDs is still limited and characterization techniques must be performed to these materials or the conjugates developed in the lab to assure a proper function and reproducibility. Furthermore, while there is significant data of QDs experiments in cell lines, the literature with primary human cells is scarce, and QD behavior in these systems may be different. Rigorous characterization data of commercially available QDs and their conjugates with biomolecules of interest is needed in order to establish their potential for target labelling and expand their use among research labs. Here we compare the characterization and labelling performance of different QD conjugates in SH-SY5Y cell line, fibroblasts and immortalized lymphocytes derived from amyotrophic lateral sclerosis patients.
Collapse
Affiliation(s)
- Carlota Tosat-Bitrián
- Centro de Investigaciones Biológicas Margarita Salas CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain; (C.T.-B.); (A.A.-B.); (G.P.); (A.M.-R.)
| | - Alicia Avis-Bodas
- Centro de Investigaciones Biológicas Margarita Salas CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain; (C.T.-B.); (A.A.-B.); (G.P.); (A.M.-R.)
| | - Gracia Porras
- Centro de Investigaciones Biológicas Margarita Salas CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain; (C.T.-B.); (A.A.-B.); (G.P.); (A.M.-R.)
| | - Daniel Borrego-Hernández
- Neurology Department, ALS Unit, CIBERER U-723, Health Research Institute, 28041 Madrid, Spain; (D.B.-H.); (A.G.-R.)
| | - Alberto García-Redondo
- Neurology Department, ALS Unit, CIBERER U-723, Health Research Institute, 28041 Madrid, Spain; (D.B.-H.); (A.G.-R.)
| | - Angeles Martín-Requero
- Centro de Investigaciones Biológicas Margarita Salas CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain; (C.T.-B.); (A.A.-B.); (G.P.); (A.M.-R.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Valle Palomo
- Centro de Investigaciones Biológicas Margarita Salas CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain; (C.T.-B.); (A.A.-B.); (G.P.); (A.M.-R.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Correspondence:
| |
Collapse
|
11
|
Hwang CH. Targeted Delivery of Erythropoietin Hybridized with Magnetic Nanocarriers for the Treatment of Central Nervous System Injury: A Literature Review. Int J Nanomedicine 2020; 15:9683-9701. [PMID: 33311979 PMCID: PMC7726550 DOI: 10.2147/ijn.s287456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
Although the incidence of central nervous system injuries has continued to rise, no promising treatments have been elucidated. Erythropoietin plays an important role in neuroprotection and neuroregeneration as well as in erythropoiesis. Moreover, the current worldwide use of erythropoietin in the treatment of hematologic diseases allows for its ready application in patients with central nervous system injuries. However, erythropoietin has a very short therapeutic time window (within 6–8 hours) after injury, and it has both hematopoietic and nonhematopoietic receptors, which exhibit heterogenic and phylogenetic differences. These differences lead to limited amounts of erythropoietin binding to in situ erythropoietin receptors. The lack of high-quality evidence for clinical use and the promising results of in vitro/in vivo models necessitate fast targeted delivery agents such as nanocarriers. Among current nanocarriers, noncovalent polymer-entrapping or polymer-adsorbing erythropoietin obtained by nanospray drying may be the most promising. With the incorporation of magnetic nanocarriers into an erythropoietin polymer, spatiotemporal external magnetic navigation is another area of great interest for targeted delivery within the therapeutic time window. Intravenous administration is the most readily used route. Manufactured erythropoietin nanocarriers should be clearly characterized using bioengineering analyses of the in vivo size distribution and the quality of entrapment or adsorption. Further preclinical trials are required to increase the therapeutic bioavailability (in vivo biological identity alteration, passage through the lung capillaries or the blood brain barrier, and timely degradation followed by removal of the nanocarriers from the body) and decrease the adverse effects (hematological complications, neurotoxicity, and cytotoxicity), especially of the nanocarrier.
Collapse
Affiliation(s)
- Chang Ho Hwang
- Department of Physical and Rehabilitation Medicine, Chungnam National University Sejong Hospital, Chungnam National University College of Medicine, Sejong, Republic of Korea
| |
Collapse
|
12
|
Lojk J, Babič L, Sušjan P, Bregar VB, Pavlin M, Hafner-Bratkovič I, Veranič P. Analysis of the Direct and Indirect Effects of Nanoparticle Exposure on Microglial and Neuronal Cells In Vitro. Int J Mol Sci 2020; 21:E7030. [PMID: 32987760 PMCID: PMC7582992 DOI: 10.3390/ijms21197030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/03/2020] [Accepted: 09/21/2020] [Indexed: 11/29/2022] Open
Abstract
Environmental or biomedical exposure to nanoparticles (NPs) can results in translocation and accumulation of NPs in the brain, which can lead to health-related problems. NPs have been shown to induce toxicity to neuronal cells through several direct mechanisms, but only a few studies have also explored the indirect effects of NPs, through consequences due to the exposure of neighboring cells to NPs. In this study, we analysed possible direct and indirect effects of NPs (polyacrylic acid (PAA) coated cobalt ferrite NP, TiO2 P25 and maghemite NPs) on immortalized mouse microglial cells and differentiated CAD mouse neuronal cells in monoculture (direct toxicity) or in transwell co-culture system (indirect toxicity). We showed that although the low NP concentrations (2-25 µg/mL) did not induce changes in cell viability, cytokine secretion or NF-κB activation of microglial cells, even low NP concentrations of 10 µg/mL can affect the cells and change their secretion of protein stress mediators. These can in turn influence neuronal cells in indirect exposure model. Indirect toxicity of NPs is an important and not adequately assessed mechanism of NP toxicity, since it not only affects cells on the exposure sites, but through secretion of signaling mediators, can also affect cells that do not come in direct contact with NPs.
Collapse
Affiliation(s)
- Jasna Lojk
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, 1000 Ljubljana, Slovenia; (L.B.); (V.B.B.); (M.P.)
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Lea Babič
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, 1000 Ljubljana, Slovenia; (L.B.); (V.B.B.); (M.P.)
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova ulica 19, 1000 Ljubljana, Slovenia; (P.S.); (I.H.-B.)
| | - Petra Sušjan
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova ulica 19, 1000 Ljubljana, Slovenia; (P.S.); (I.H.-B.)
| | - Vladimir Boštjan Bregar
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, 1000 Ljubljana, Slovenia; (L.B.); (V.B.B.); (M.P.)
| | - Mojca Pavlin
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, 1000 Ljubljana, Slovenia; (L.B.); (V.B.B.); (M.P.)
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova ulica 19, 1000 Ljubljana, Slovenia; (P.S.); (I.H.-B.)
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, 1000 Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia;
| |
Collapse
|
13
|
Nguyen CT, Kim CR, Le TH, Koo KI, Hwang CH. Magnetically guided targeted delivery of erythropoietin using magnetic nanoparticles: Proof of concept. Medicine (Baltimore) 2020; 99:e19972. [PMID: 32384447 PMCID: PMC7220084 DOI: 10.1097/md.0000000000019972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The objective of this proof-of-concept study was to demonstrate the targeted delivery of erythropoietin (EPO) using magnetically guided magnetic nanoparticles (MNPs).MNPs consisting of a ferric-ferrous mixture (FeCl3·6H2O and FeCl2·4H2O) were prepared using a co-precipitation method. The drug delivery system (DDS) was manufactured via the spray-drying technique using a nanospray-dryer. The DDS comprised 7.5 mg sodium alginate, 150 mg MNPs, and 1000 IU EPO.Scanning electron microscopy revealed DDS particles no more than 500 nm in size. Tiny particles on the rough surfaces of the DDS particles were composed of MNPs and/or EPO, unlike the smooth surfaces of the only alginate particles. Transmission electron microscopy showed the tiny particles from 5 to 20 nm in diameter. Fourier-transform infrared spectroscopy revealed DDS peaks characteristic of MNPs as well as of alginate. Thermal gravimetric analysis presented that 50% of DDS weight was lost in a single step around 500°C. The mode size of the DDS particles was approximately 850 nm under in vivo conditions. Standard soft lithography was applied to DDS particles prepared with fluorescent beads using a microchannel fabricated to have one inlet and two outlets in a Y-shape. The fluorescent DDS particles reached only one outlet reservoir in the presence of a neodymium magnet. The neurotoxicity was evaluated by treating SH-SY5Y cells in 48-well plates (1 × 10 cells/well) with 2 μL of a solution containing sodium alginate (0.075 mg/mL), MNPs (1.5 mg/mL), or sodium alginate + MNPs. A cell viability assay kit was used to identify a 93% cell viability after MNP treatment and a 94% viability after sodium alginate + MNP treatment, compared with the control. As for the DDS particle neurotoxicity, a 95% cell viability was noticed after alginate-encapsulated MNPs treatment and a 93% cell viability after DDS treatment, compared with the control.The DDS-EPO construct developed here can be small under in vivo conditions enough to pass through the lung capillaries with showing the high coating efficiency. It can be guided using magnetic control without displaying significant neurotoxicity in the form of solution or particles.
Collapse
Affiliation(s)
| | - Chung Reen Kim
- Department of Physical Medicine and Rehabilitation, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan
| | - Thi Huong Le
- Department of Biomedical Engineering, University of Ulsan, Ulsan
| | - Kyo-in Koo
- Department of Biomedical Engineering, University of Ulsan, Ulsan
| | - Chang Ho Hwang
- Department of Biomedical Engineering, University of Ulsan, Ulsan
- Department of Physical and Rehabilitation Medicine, Chungnam National University Sejong Hospital, Chungnam National University College of Medicine, Sejong, Republic of Korea
| |
Collapse
|
14
|
Labusca L, Herea DD, Danceanu CM, Minuti AE, Stavila C, Grigoras M, Gherca D, Stoian G, Ababei G, Chiriac H, Lupu N. The effect of magnetic field exposure on differentiation of magnetite nanoparticle-loaded adipose-derived stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 109:110652. [PMID: 32228923 DOI: 10.1016/j.msec.2020.110652] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/26/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023]
Abstract
Magnetic nanoparticles (MNPs) are versatile tools for various applications in biotechnology and nanomedicine. MNPs-mediated cell tracking, targeting and imaging are increasingly studied for regenerative medicine applications in cell therapy and tissue engineering. Mechanical stimulation influences mesenchymal stem cell differentiation. Here we show that MNPs-mediated magneto-mechanical stimulation of human primary adipose derived stem cells (ADSCs) exposed to variable magnetic field (MF) influences their adipogenic and osteogenic differentiation. ADSCs loaded with biocompatible magnetite nanoparticles of 6.6 nm, and with an average load of 21 picograms iron/cell were exposed to variable low intensity (0.5 mT - LMF) and higher intensity magnetic fields (14.7 and 21.6 mT - HMF). Type, duration, intensity and frequency of MF differently affect differentiation. Short time (2 days) intermittent exposure to LMF increases adipogenesis while longer (7 days) intermittent as well as continuous exposure favors osteogenesis. HMF (21.6 mT) short time intermittent exposure favors osteogenesis. Different exposure protocols can be used to increase differentiation dependently on expected results. Magnetic remotely-actuated MNPs up-taken by ADSCs promotes the shift towards osteoblastic lineage. ADSCs-MNPs under MF exposure could be used for enabling osteoblastic conversion during cell therapy for systemic osteoporosis. Current results enable further in vivo studies investigating the role of remotely-controlled magnetically actuated ADSCs-MNPs for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Luminita Labusca
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania
| | - Dumitru-Daniel Herea
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania.
| | - Camelia-Mihaela Danceanu
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania; University "Al. I. Cuza", 11 Carol I Boulevard, 700506, Iasi, Romania
| | - Anca Emanuela Minuti
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania; University "Al. I. Cuza", 11 Carol I Boulevard, 700506, Iasi, Romania
| | - Cristina Stavila
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania; University "Al. I. Cuza", 11 Carol I Boulevard, 700506, Iasi, Romania
| | - Marian Grigoras
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania
| | - Daniel Gherca
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania
| | - George Stoian
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania
| | - Gabriel Ababei
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania
| | - Horia Chiriac
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania
| | - Nicoleta Lupu
- National Institute of Research and Development for Technical Physics, 47 Mangeron Blvd., 700050, Iasi, Romania
| |
Collapse
|
15
|
Kumarasamy M, Sosnik A. The Nose-To-Brain Transport of Polymeric Nanoparticles Is Mediated by Immune Sentinels and Not by Olfactory Sensory Neurons. ADVANCED BIOSYSTEMS 2019; 3:e1900123. [PMID: 32648679 DOI: 10.1002/adbi.201900123] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/02/2019] [Indexed: 11/11/2022]
Abstract
The nose-to-brain (N-to-B) transport mechanism of nanoparticles through the olfactory epithelium (OE) is not fully understood. Most research utilized nasal epithelial cell models completely deprived of olfactory cells. Aiming to shed light into key cellular pathways, in this work, for the first time, the interaction of polymeric nanoparticles in a 17-483 nm size range and with neutral and negatively and positively charged surfaces with primary olfactory sensory neurons, cortical neurons, and microglia isolated from olfactory bulb (OB), OE, and cortex of newborn rats is investigated. After demonstrating the good cell compatibility of the different nanoparticles, the nanoparticle uptake by confocal laser scanning fluorescence microscopy is monitored. Our findings reveal that neither olfactory nor forebrain neurons internalize nanoparticles. Conversely, it is demonstrated that olfactory and cortical microglia phagocytose the nanoparticles independently of their features. Overall, our findings represent the first unambiguous evidence of the possible involvement of microglia in N-to-B nanoparticle transport and the unlikely involvement of neurons. Furthermore, this approach emerges as a completely new experimental tool to screen the biocompatibility, uptake, and transport of nanomaterials by key cellular players of the N-to-B pathway in nanosafety and nanotoxicology and nanomedicine.
Collapse
Affiliation(s)
- Murali Kumarasamy
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| |
Collapse
|
16
|
Peviani M, Capasso Palmiero U, Cecere F, Milazzo R, Moscatelli D, Biffi A. Biodegradable polymeric nanoparticles administered in the cerebrospinal fluid: Brain biodistribution, preferential internalization in microglia and implications for cell-selective drug release. Biomaterials 2019; 209:25-40. [PMID: 31026609 DOI: 10.1016/j.biomaterials.2019.04.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 12/31/2022]
Abstract
Cell-selective drug release in the central nervous system (CNS) holds great promise for the treatment of many CNS disorders but it is still challenging. We previously demonstrated that polymeric nanoparticles (NPs) injected intra-parenchyma in the CNS can be internalized specifically in microglia/macrophages surrounding the injection site. Here, we explored NPs administration in the cerebrospinal fluid (CSF) to achieve a wider spreading and increased cell targeting throughout the CNS; we generated new NPs variants and studied the effect of modifying size and surface charge on NPs biodistribution and cellular uptake. Intra-cerebroventricular administration resulted in prevalent localization of the NPs in proximity to stem-cell niches, such as around the lateral ventricles, the subventricular zone and the rostral migratory stream. NPs internalization occurred preferentially in brain myeloid cells/microglia. We demonstrated that brain biodistribution and extent of internalization in microglia are influenced by NPs dimensions and can be improved by applying a transient disruption of the blood-brain barrier with mannitol, leading to NPs internalization in up to 25% of brain myeloid/microglia cells. A fraction of the targeted cells was positive for markers of proliferation or stained positive for stemness/progenitor-cell markers such as Nestin, c-kit, or NG2. Interestingly, through these newly formulated NPs we obtained controlled and selective release of drugs otherwise difficult to formulate (such as busulfan and etoposide) to the target cells, preventing unwanted side effects and the toxicity obtained by direct brain delivery of the not encapsulated drugs. Overall, these data provide proof of concept of the applicability of these novel NP-based drug formulations for achieving internalization not only in mature microglia but also possibly in more immature myeloid cells in the brain and pave the way for brain-restricted microglia-targeted drug delivery regimens.
Collapse
Affiliation(s)
- Marco Peviani
- Gene Therapy Program, Dana Farber/Boston Children's Cancer and Blood Disorders Center, 450 Brookline Ave., 02215, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 48, 20156, Milan, Italy.
| | - Umberto Capasso Palmiero
- Dipartimento di Chimica, Materiali ed Ingegneria Chimica, Politecnico di Milano, 20131, Milan, Italy; Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg 1, 8093 Zürich, Switzerland
| | - Francesca Cecere
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 48, 20156, Milan, Italy
| | - Rita Milazzo
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 48, 20156, Milan, Italy
| | - Davide Moscatelli
- Dipartimento di Chimica, Materiali ed Ingegneria Chimica, Politecnico di Milano, 20131, Milan, Italy
| | - Alessandra Biffi
- Gene Therapy Program, Dana Farber/Boston Children's Cancer and Blood Disorders Center, 450 Brookline Ave., 02215, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 48, 20156, Milan, Italy.
| |
Collapse
|
17
|
Lesniak A, Kilinc D, Blasiak A, Galea G, Simpson JC, Lee GU. Rapid Growth Cone Uptake and Dynein-Mediated Axonal Retrograde Transport of Negatively Charged Nanoparticles in Neurons Is Dependent on Size and Cell Type. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1803758. [PMID: 30565853 DOI: 10.1002/smll.201803758] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/20/2018] [Indexed: 06/09/2023]
Abstract
Nanoparticles (NPs) are now used in numerous technologies and serve as carriers for several new classes of therapeutics. Studies of the distribution of NPs in vivo demonstrate that they can be transported through biological barriers and are concentrated in specific tissues. Here, transport behavior, and final destination of polystyrene NPs are reported in primary mouse cortical neurons and SH-SY5Y cells, cultured in two-compartmental microfluidic devices. In both cell types, negative polystyrene NPs (PS(-)) smaller than 100 nm are taken up by the axons, undergo axonal retrograde transport, and accumulate in the somata. Examination of NP transport reveals different transport mechanisms depending on the cell type, particle charge, and particle internalization by the lysosomes. In cortical neurons, PS(-) inside lysosomes and 40 nm positive polystyrene NPs undergo slow axonal transport, whereas PS(-) outside lysosomes undergo fast axonal transport. Inhibition of dynein in cortical neurons decreases the transport velocity and cause a dose-dependent reduction in the number of accumulated PS(-), suggesting that the fast axonal transport is dynein mediated. These results show that the axonal retrograde transport of NPs depends on the endosomal pathway taken and establishes a means for screening nanoparticle-based therapeutics for diseases that involve neurons.
Collapse
Affiliation(s)
- Anna Lesniak
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Devrim Kilinc
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Agata Blasiak
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - George Galea
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biology and Environmental Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jeremy C Simpson
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biology and Environmental Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gil U Lee
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
18
|
De Simone U, Roccio M, Gribaldo L, Spinillo A, Caloni F, Coccini T. Human 3D Cultures as Models for Evaluating Magnetic Nanoparticle CNS Cytotoxicity after Short- and Repeated Long-Term Exposure. Int J Mol Sci 2018; 19:ijms19071993. [PMID: 29986546 PMCID: PMC6073335 DOI: 10.3390/ijms19071993] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 11/20/2022] Open
Abstract
Since nanoparticles (NPs) can translocate to the brain and impact the highly vulnerable central nervous system (CNS), novel in vitro tools for the assessment of NP-induced neurotoxicity are advocated. In this study, two types of CNS spheroids have been developed from human D384 astrocyte- and SH-SY5Y neuronal-like cells, and optimized in combination with standard assays (viability readout and cell morphology) to test neurotoxic effects caused by Fe3O4NPs, as NP-model, after short- (24–48 h; 1–100µg/ml) and long-term repeated exposure (30days; 0.1–25µg/ml). Short-term exposure of 3D-spheroids to Fe3O4NP induced cytotoxicity at 10 µg/mL in astrocytes and 25 µg/mL neurons. After long-term repeated dose regimen, spheroids showed concentration- and time-dependent cell mortality at 10 µg/mL for D384 and 0.5 µg/mL for SH-SY5Y, indicating a higher susceptibility of neurons than astrocytes. Both spheroid types displayed cell disaggregation after the first week of treatment at ≥0.1 µg/mL and becoming considerably evident at higher concentrations and over time. Recreating the 3D-spatial environment of the CNS allows cells to behave in vitro more closely to the in vivo situations, therefore providing a model that can be used as a stand-alone test or as a part of integrated testing strategies. These models could drive an improvement in the species-relevant predictivity of toxicity testing.
Collapse
Affiliation(s)
- Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, Toxicology Unit, ICS Maugeri SpA-BC, IRCCS Pavia, 27100 Pavia, Italy.
| | - Marianna Roccio
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, 27100 Pavia, Italy.
| | - Laura Gribaldo
- European Commission, Directorate General Joint Research Centre, Directorate F-Health, Consumers and Reference Materials, Chemicals Safety and Alternative Methods Unit, 21027 Ispra, Italy.
| | - Arsenio Spinillo
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, 27100 Pavia, Italy.
| | - Francesca Caloni
- Università degli Studi di Milano, Dipartimento di Medicina Veterinaria (DIMEVET), 20133 Milano, Italy.
| | - Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, Toxicology Unit, ICS Maugeri SpA-BC, IRCCS Pavia, 27100 Pavia, Italy.
| |
Collapse
|
19
|
Gahl TJ, Kunze A. Force-Mediating Magnetic Nanoparticles to Engineer Neuronal Cell Function. Front Neurosci 2018; 12:299. [PMID: 29867315 PMCID: PMC5962660 DOI: 10.3389/fnins.2018.00299] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022] Open
Abstract
Cellular processes like membrane deformation, cell migration, and transport of organelles are sensitive to mechanical forces. Technically, these cellular processes can be manipulated through operating forces at a spatial precision in the range of nanometers up to a few micrometers through chaperoning force-mediating nanoparticles in electrical, magnetic, or optical field gradients. But which force-mediating tool is more suitable to manipulate cell migration, and which, to manipulate cell signaling? We review here the differences in forces sensation to control and engineer cellular processes inside and outside the cell, with a special focus on neuronal cells. In addition, we discuss technical details and limitations of different force-mediating approaches and highlight recent advancements of nanomagnetics in cell organization, communication, signaling, and intracellular trafficking. Finally, we give suggestions about how force-mediating nanoparticles can be used to our advantage in next-generation neurotherapeutic devices.
Collapse
Affiliation(s)
| | - Anja Kunze
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, MT, United States
| |
Collapse
|
20
|
Preferential and Increased Uptake of Hydroxyl-Terminated PAMAM Dendrimers by Activated Microglia in Rabbit Brain Mixed Glial Culture. Molecules 2018; 23:molecules23051025. [PMID: 29702566 PMCID: PMC6102539 DOI: 10.3390/molecules23051025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 01/05/2023] Open
Abstract
Polyamidoamine (PAMAM) dendrimers are multifunctional nanoparticles with tunable physicochemical features, making them promising candidates for targeted drug delivery in the central nervous system (CNS). Systemically administered dendrimers have been shown to localize in activated glial cells, which mediate neuroinflammation in the CNS. These dendrimers delivered drugs specifically to activated microglia, producing significant neurological improvements in multiple brain injury models, including in a neonatal rabbit model of cerebral palsy. To gain further insight into the mechanism of dendrimer cell uptake, we utilized an in vitro model of primary glial cells isolated from newborn rabbits to assess the differences in hydroxyl-terminated generation 4 PAMAM dendrimer (D4-OH) uptake by activated and non-activated glial cells. We used fluorescently-labelled D4-OH (D-Cy5) as a tool for investigating the mechanism of dendrimer uptake. D4-OH PAMAM dendrimer uptake was determined by fluorescence quantification using confocal microscopy and flow cytometry. Our results indicate that although microglial cells in the mixed cell population demonstrate early uptake of dendrimers in this in vitro system, activated microglia take up more dendrimer compared to resting microglia. Astrocytes showed delayed and limited uptake. We also illustrated the differences in mechanism of uptake between resting and activated microglia using different pathway inhibitors. Both resting and activated microglia primarily employed endocytotic pathways, which are enhanced in activated microglial cells. Additionally, we demonstrated that hydroxyl terminated dendrimers are taken up by primary microglia using other mechanisms including pinocytosis, caveolae, and aquaporin channels for dendrimer uptake.
Collapse
|
21
|
In-vitro in-vivo correlation (IVIVC) in nanomedicine: Is protein corona the missing link? Biotechnol Adv 2017; 35:889-904. [DOI: 10.1016/j.biotechadv.2017.08.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/04/2017] [Accepted: 08/19/2017] [Indexed: 12/17/2022]
|
22
|
Cell damage produced by magnetic fluid hyperthermia on microglial BV2 cells. Sci Rep 2017; 7:8627. [PMID: 28819156 PMCID: PMC5561037 DOI: 10.1038/s41598-017-09059-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/19/2017] [Indexed: 02/07/2023] Open
Abstract
We present evidence on the effects of exogenous heating by water bath (WB) and magnetic hyperthermia (MHT) on a glial micro-tumor phantom. To this, magnetic nanoparticles (MNPs) of 30-40 nm were designed to obtain particle sizes for maximum heating efficiency. The specific power absorption (SPA) values (f = 560 kHz, H = 23.9 kA/m) for as prepared colloids (533-605 W/g) dropped to 98-279 W/g in culture medium. The analysis of the intracellular MNPs distribution showed vesicle-trapped MNPs agglomerates spread along the cytoplasm, as well as large (~0.5-0.9 μm) clusters attached to the cell membrane. Immediately after WB and MHT (T = 46 °C for 30 min) the cell viability was ≈70% and, after 4.5 h, decreased to 20-25%, demonstrating that metabolic processes are involved in cell killing. The analysis of the cell structures after MHT revealed a significant damage of the cell membrane that is correlated to the location of MNPs clusters, while local cell damage were less noticeable after WB without MNPs. In spite of the similar thermal effects of WB and MHT on the cell viability, our results suggest that there is an additional mechanism of cell damage related to the presence of MNPs at the intracellular space.
Collapse
|
23
|
Yarjanli Z, Ghaedi K, Esmaeili A, Rahgozar S, Zarrabi A. Iron oxide nanoparticles may damage to the neural tissue through iron accumulation, oxidative stress, and protein aggregation. BMC Neurosci 2017; 18:51. [PMID: 28651647 PMCID: PMC5485499 DOI: 10.1186/s12868-017-0369-9] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 06/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the recent decade, iron oxide nanoparticles (IONPs) have been proposed for several applications in the central nervous system (CNS), including targeting amyloid beta (Aβ) in the arteries, inhibiting the microglial cells, delivering drugs, and increasing contrast in magnetic resonance imaging. Conversely, a notable number of studies have reported the role of iron in neurodegenerative diseases. Therefore, this study has reviewed the recent studies to determine whether IONPs iron can threaten the cellular viability same as iron. RESULTS Iron contributes in Fenton's reaction and produces reactive oxygen species (ROS). ROS cause to damage the macromolecules and organelles of the cell via oxidative stress. Iron accumulation and oxidative stress are able to aggregate some proteins, including Aβ and α-synuclein, which play a critical role in Alzheimer's and Parkinson's diseases, respectively. Iron accumulation, oxidative stress, and protein aggregation make a positive feedback loop, which can be toxic for the cell. The release of iron ions from IONPs may result in iron accumulation in the targeted tissue, and thus, activate the positive feedback loop. However, the levels of IONPs induced toxicity depend on the size, concentration, surface charge, and the type of coating and functional groups of IONPs. CONCLUSION IONPs depending on their properties can lead to iron accumulation, oxidative stress and protein aggregation in the neural cells. Therefore, in order to apply IONPs in the CNS, the consideration of IONPs properties is crucial.
Collapse
Affiliation(s)
- Zahra Yarjanli
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Abolghasem Esmaeili
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Soheila Rahgozar
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Ali Zarrabi
- Department of Biotechnology, Faculty of Advanced Sciences and Technologies, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| |
Collapse
|
24
|
Hsiao IL, Hsieh YK, Chuang CY, Wang CF, Huang YJ. Effects of silver nanoparticles on the interactions of neuron- and glia-like cells: Toxicity, uptake mechanisms, and lysosomal tracking. ENVIRONMENTAL TOXICOLOGY 2017; 32:1742-1753. [PMID: 28181394 DOI: 10.1002/tox.22397] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 01/14/2017] [Accepted: 01/15/2017] [Indexed: 05/11/2023]
Abstract
Silver nanoparticles (AgNPs) are commonly used nanomaterials in consumer products. Previous studies focused on its effects on neurons; however, little is known about their effects and uptake mechanisms on glial cells under normal or activated states. Here, ALT astrocyte-like, BV-2 microglia and differentiated N2a neuroblastoma cells were directly or indirectly exposed to 10 nm AgNPs using mono- and co-culture system. A lipopolysaccharide (LPS) was pretreated to activate glial cells before AgNP treatment for mimicking NP exposure under brain inflammation. From mono-culture, ALT took up the most AgNPs and had the lowest cell viability within three cells. Moreover, AgNPs induced H2 O2 and NO from ALT/activated ALT and BV-2, respectively. However, AgNPs did not induce cytokines release (IL-6, TNF-α, MCP-1). LPS-activated BV-2 took up more AgNPs than normal BV-2, while the induction of ROS and cytokines from activated cells were diminished. Ca2+ -regulated clathrin- and caveolae-independent endocytosis and phagocytosis were involved in the AgNP uptake in ALT, which caused more rapid NP translocation to lysosome than in macropinocytosis and clathrin-dependent endocytosis-involved BV-2. AgNPs directly caused apoptosis and necrosis in N2a cells, while by indirect NP exposure to bottom chamber ALT or BV-2 in Transwell, more apoptotic upper chamber N2a cells were observed. Cell viability of BV-2 also decreased in an ALT-BV-2 co-culturing study. The damaged cells correlated to NP-mediated H2 O2 release from ALT or NO from BV-2, which indicates that toxic response of AgNPs to neurons is not direct, but indirectly arises from AgNP-induced soluble factors from other glial cells.
Collapse
Affiliation(s)
- I-Lun Hsiao
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yi-Kong Hsieh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chun-Yu Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chu-Fang Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yuh-Jeen Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| |
Collapse
|
25
|
Giannaccini M, Calatayud MP, Poggetti A, Corbianco S, Novelli M, Paoli M, Battistini P, Castagna M, Dente L, Parchi P, Lisanti M, Cavallini G, Junquera C, Goya GF, Raffa V. Magnetic Nanoparticles for Efficient Delivery of Growth Factors: Stimulation of Peripheral Nerve Regeneration. Adv Healthc Mater 2017; 6. [PMID: 28156059 DOI: 10.1002/adhm.201601429] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/09/2017] [Indexed: 12/19/2022]
Abstract
The only clinically approved alternative to autografts for treating large peripheral nerve injuries is the use of synthetic nerve guidance conduits (NGCs), which provide physical guidance to the regenerating stump and limit scar tissue infiltration at the injury site. Several lines of evidence suggest that a potential future strategy is to combine NGCs with cellular or molecular therapies to deliver growth factors that sustain the regeneration process. However, growth factors are expensive and have a very short half-life; thus, the combination approach has not been successful. In the present paper, we proposed the immobilization of growth factors (GFs) on magnetic nanoparticles (MNPs) for the time- and space-controlled release of GFs inside the NGC. We tested the particles in a rat model of a peripheral nerve lesion. Our results revealed that the injection of a cocktail of MNPs functionalized with nerve growth factor (NGF) and with vascular endothelial growth factor (VEGF) strongly accelerate the regeneration process and the recovery of motor function compared to that obtained using the free factors. Additionally, we found that injecting MNPs in the NGC is safe and does not impair the regeneration process, and the MNPs remain in the conduit for weeks.
Collapse
Affiliation(s)
- Martina Giannaccini
- Department of Biology; Università di Pisa; S.S. 12 Abetone e Brennero 4 56127 Pisa Italy
| | - M. Pilar Calatayud
- Instituto de Nanociencia de Aragon; Universidad de Zaragoza; Mariano Esquillor 50018 Zaragoza Spain
| | - Andrea Poggetti
- Department of Translational Research and of New Surgical and Medical Technologies; Università di Pisa; Via Savi 8 56126 Pisa Italy
| | - Silvia Corbianco
- Department of Translational Research and of New Surgical and Medical Technologies; Università di Pisa; Via Savi 8 56126 Pisa Italy
| | - Michela Novelli
- Department of Translational Research and of New Surgical and Medical Technologies; Università di Pisa; Via Savi 8 56126 Pisa Italy
| | - Melania Paoli
- Institute of Life Science; Scuola Superiore Sant'Anna; Piazza Martiri della Libertà 33 56127 Pisa Italy
| | - Pietro Battistini
- Department of Translational Research and of New Surgical and Medical Technologies; Università di Pisa; Via Savi 8 56126 Pisa Italy
| | - Maura Castagna
- Department of Translational Research and of New Surgical and Medical Technologies; Università di Pisa; Via Savi 8 56126 Pisa Italy
| | - Luciana Dente
- Department of Biology; Università di Pisa; S.S. 12 Abetone e Brennero 4 56127 Pisa Italy
| | - Paolo Parchi
- Department of Translational Research and of New Surgical and Medical Technologies; Università di Pisa; Via Savi 8 56126 Pisa Italy
| | - Michele Lisanti
- Department of Translational Research and of New Surgical and Medical Technologies; Università di Pisa; Via Savi 8 56126 Pisa Italy
| | - Gabriella Cavallini
- Department of Translational Research and of New Surgical and Medical Technologies; Università di Pisa; Via Savi 8 56126 Pisa Italy
| | - Concepción Junquera
- Institute for Health Research Aragon IIS; Faculty of Medicine, C/Domingo Mirals/n; 50009 Zaragoza Spain
| | - Gerardo F. Goya
- Instituto de Nanociencia de Aragon; Universidad de Zaragoza; Mariano Esquillor 50018 Zaragoza Spain
| | - Vittoria Raffa
- Department of Biology; Università di Pisa; S.S. 12 Abetone e Brennero 4 56127 Pisa Italy
- Institute of Life Science; Scuola Superiore Sant'Anna; Piazza Martiri della Libertà 33 56127 Pisa Italy
| |
Collapse
|
26
|
Kunze A, Murray CT, Godzich C, Lin J, Owsley K, Tay A, Di Carlo D. Modulating motility of intracellular vesicles in cortical neurons with nanomagnetic forces on-chip. LAB ON A CHIP 2017; 17:842-854. [PMID: 28164203 PMCID: PMC5400667 DOI: 10.1039/c6lc01349j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Vesicle transport is a major underlying mechanism of cell communication. Inhibiting vesicle transport in brain cells results in blockage of neuronal signals, even in intact neuronal networks. Modulating intracellular vesicle transport can have a huge impact on the development of new neurotherapeutic concepts, but only if we can specifically interfere with intracellular transport patterns. Here, we propose to modulate motion of intracellular lipid vesicles in rat cortical neurons based on exogenously bioconjugated and cell internalized superparamagnetic iron oxide nanoparticles (SPIONs) within microengineered magnetic gradients on-chip. Upon application of 6-126 pN on intracellular vesicles in neuronal cells, we explored how the magnetic force stimulus impacts the motion pattern of vesicles at various intracellular locations without modulating the entire cell morphology. Altering vesicle dynamics was quantified using, mean square displacement, a caging diameter and the total traveled distance. We observed a de-acceleration of intercellular vesicle motility, while applying nanomagnetic forces to cultured neurons with SPIONs, which can be explained by a decrease in motility due to opposing magnetic force direction. Ultimately, using nanomagnetic forces inside neurons may permit us to stop the mis-sorting of intracellular organelles, proteins and cell signals, which have been associated with cellular dysfunction. Furthermore, nanomagnetic force applications will allow us to wirelessly guide axons and dendrites by exogenously using permanent magnetic field gradients.
Collapse
Affiliation(s)
- Anja Kunze
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA. and Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, USA.
| | - Coleman Tylor Murray
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA.
| | - Chanya Godzich
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA.
| | - Jonathan Lin
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA.
| | - Keegan Owsley
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA.
| | - Andy Tay
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA.
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA. and California NanoSystems Institute, University of California, Los Angeles, California 90095, USA and Jonsson Comprehensive Cancer Research Center, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
27
|
Pohland M, Glumm R, Wiekhorst F, Kiwit J, Glumm J. Biocompatibility of very small superparamagnetic iron oxide nanoparticles in murine organotypic hippocampal slice cultures and the role of microglia. Int J Nanomedicine 2017; 12:1577-1591. [PMID: 28280327 PMCID: PMC5339010 DOI: 10.2147/ijn.s127206] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIO) are applied as contrast media for magnetic resonance imaging (MRI) and treatment of neurologic diseases despite the fact that important information concerning their local interactions is still lacking. Due to their small size, SPIO have great potential for magnetically labeling different cell populations, facilitating their MRI tracking in vivo. Before SPIO are applied, however, their effect on cell viability and tissue homoeostasis should be studied thoroughly. We have previously published data showing how citrate-coated very small superparamagnetic iron oxide particles (VSOP) affect primary microglia and neuron cell cultures as well as neuron-glia cocultures. To extend our knowledge of VSOP interactions on the three-dimensional multicellular level, we further examined the influence of two types of coated VSOP (R1 and R2) on murine organotypic hippocampal slice cultures. Our data show that 1) VSOP can penetrate deep tissue layers, 2) long-term VSOP-R2 treatment alters cell viability within the dentate gyrus, 3) during short-term incubation VSOP-R1 and VSOP-R2 comparably modify hippocampal cell viability, 4) VSOP treatment does not affect cytokine homeostasis, 5) microglial depletion decreases VSOP uptake, and 6) microglial depletion plus VSOP treatment increases hippocampal cell death during short-term incubation. These results are in line with our previous findings in cell coculture experiments regarding microglial protection of neurite branching. Thus, we have not only clarified the interaction between VSOP, slice culture, and microglia to a degree but also demonstrated that our model is a promising approach for screening nanoparticles to exclude potential cytotoxic effects.
Collapse
Affiliation(s)
- Martin Pohland
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité - Universitätsmedizin Berlin
| | - Robert Glumm
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité - Universitätsmedizin Berlin; Clinic of Neurology, Jüdisches Krankenhaus
| | - Frank Wiekhorst
- Department 8.2 Biosignals, Physikalisch-Technische Bundesanstalt
| | - Jürgen Kiwit
- Clinic of Neurosurgery, HELIOS Klinikum Berlin Buch, Berlin, Germany
| | - Jana Glumm
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité - Universitätsmedizin Berlin; Clinic of Neurosurgery, HELIOS Klinikum Berlin Buch, Berlin, Germany
| |
Collapse
|
28
|
Healy S, McMahon J, Owens P, FitzGerald U. Significant glial alterations in response to iron loading in a novel organotypic hippocampal slice culture model. Sci Rep 2016; 6:36410. [PMID: 27808258 PMCID: PMC5093415 DOI: 10.1038/srep36410] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/14/2016] [Indexed: 12/31/2022] Open
Abstract
Aberrant iron deposition in the brain is associated with neurodegenerative disorders including Multiple Sclerosis, Alzheimer’s disease and Parkinson’s disease. To study the collective response to iron loading, we have used hippocampal organotypic slices as a platform to develop a novel ex vivo model of iron accumulation. We demonstrated differential uptake and toxicity of iron after 12 h exposure to 10 μM ferrous ammonium sulphate, ferric citrate or ferrocene. Having established the supremacy of ferrocene in this model, the cultures were then loaded with 0.1–100 μM ferrocene for 12 h. One μM ferrocene exposure produced the maximal 1.6-fold increase in iron compared with vehicle. This was accompanied by a 1.4-fold increase in ferritin transcripts and mild toxicity. Using dual-immunohistochemistry, we detected ferritin in oligodendrocytes, microglia, but rarely in astrocytes and never in neurons in iron-loaded slice cultures. Moreover, iron loading led to a 15% loss of olig2-positive cells and a 16% increase in number and greater activation of microglia compared with vehicle. However, there was no appreciable effect of iron loading on astrocytes. In what we believe is a significant advance on traditional mono- or dual-cultures, our novel ex vivo slice-culture model allows characterization of the collective response of brain cells to iron-loading.
Collapse
Affiliation(s)
- Sinead Healy
- Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Jill McMahon
- Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Peter Owens
- Centre for Microscopy and Imaging, National University of Ireland, Galway, Ireland
| | - Una FitzGerald
- Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland
| |
Collapse
|
29
|
Latronico T, Depalo N, Valente G, Fanizza E, Laquintana V, Denora N, Fasano A, Striccoli M, Colella M, Agostiano A, Curri ML, Liuzzi GM. Cytotoxicity Study on Luminescent Nanocrystals Containing Phospholipid Micelles in Primary Cultures of Rat Astrocytes. PLoS One 2016; 11:e0153451. [PMID: 27097043 PMCID: PMC4838222 DOI: 10.1371/journal.pone.0153451] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/30/2016] [Indexed: 12/14/2022] Open
Abstract
Luminescent colloidal nanocrystals (NCs) are emerging as a new tool in neuroscience field, representing superior optical probes for cellular imaging and medical diagnosis of neurological disorders with respect to organic fluorophores. However, only a limited number of studies have, so far, explored NC applications in primary neurons, glia and related cells. Indeed astrocytes, as resident cells in the central nervous system (CNS), play an important pathogenic role in several neurodegenerative and neuroinflammatory diseases, therefore enhanced imaging tools for their thorough investigation are strongly amenable. Here, a comprehensive and systematic study on the in vitro toxicological effect of core-shell type luminescent CdSe@ZnS NCs incorporated in polyethylene glycol (PEG) terminated phospholipid micelles on primary cultures of rat astrocytes was carried out. Cytotoxicity response of empty micelles based on PEG modified phospholipids was compared to that of their NC containing counterpart, in order to investigate the effect on cell viability of both inorganic NCs and micelles protecting NC surface. Furthermore, since the surface charge and chemistry influence cell interaction and toxicity, effect of two different functional groups terminating PEG-modified phospholipid micelles, namely amine and carboxyl group, respectively, was evaluated against bare micelles, showing that carboxyl group was less toxic. The ability of PEG-lipid micelles to be internalized into the cells was qualitatively and quantitatively assessed by fluorescence microscopy and photoluminescence (PL) assay. The results of the experiments clearly demonstrate that, once incorporated into the micelles, a low, not toxic, concentration of NCs is sufficient to be distinctly detected within cells. The overall study provides essential indications to define the optimal experimental conditions to effectively and profitably use the proposed luminescent colloidal NCs as optical probe for future in vivo experiments.
Collapse
Affiliation(s)
- Tiziana Latronico
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Nicoletta Depalo
- Consiglio Nazionale delle Ricerche, Istituto per i Processi Chimico-Fisici, Bari, Italy c/o Dipartimento di Chimica, Università di Bari, Bari, Italy
| | - Gianpiero Valente
- Consiglio Nazionale delle Ricerche, Istituto per i Processi Chimico-Fisici, Bari, Italy c/o Dipartimento di Chimica, Università di Bari, Bari, Italy
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Elisabetta Fanizza
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Valentino Laquintana
- Dipartimento di Farmacia – Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Nunzio Denora
- Dipartimento di Farmacia – Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Anna Fasano
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Marinella Striccoli
- Consiglio Nazionale delle Ricerche, Istituto per i Processi Chimico-Fisici, Bari, Italy c/o Dipartimento di Chimica, Università di Bari, Bari, Italy
| | - Matilde Colella
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Angela Agostiano
- Consiglio Nazionale delle Ricerche, Istituto per i Processi Chimico-Fisici, Bari, Italy c/o Dipartimento di Chimica, Università di Bari, Bari, Italy
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - M. Lucia Curri
- Consiglio Nazionale delle Ricerche, Istituto per i Processi Chimico-Fisici, Bari, Italy c/o Dipartimento di Chimica, Università di Bari, Bari, Italy
| | - Grazia Maria Liuzzi
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| |
Collapse
|
30
|
Hudson R. Coupling the magnetic and heat dissipative properties of Fe3O4 particles to enable applications in catalysis, drug delivery, tissue destruction and remote biological interfacing. RSC Adv 2016. [DOI: 10.1039/c5ra22260e] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
As interest in nanomaterials continues to grow, and the scope of their applications widens, one subset of materials has set itself apart: magnetic nanoparticles (MNPs).
Collapse
Affiliation(s)
- R. Hudson
- Department of Chemistry
- Colby College
- Waterville
- USA
| |
Collapse
|
31
|
Sanders AW, Jeerage KM, Schwartz CL, Curtin AE, Chiaramonti AN. Gold Nanoparticle Quantitation by Whole Cell Tomography. ACS NANO 2015; 9:11792-9. [PMID: 26563983 DOI: 10.1021/acsnano.5b03815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Many proposed biomedical applications for engineered gold nanoparticles require their incorporation by mammalian cells in specific numbers and locations. Here, the number of gold nanoparticles inside of individual mammalian stem cells was characterized using fast focused ion beam-scanning electron microscopy based tomography. Enhanced optical microscopy was used to provide a multiscale map of the in vitro sample, which allows cells of interest to be identified within their local environment. Cells were then serially sectioned using a gallium ion beam and imaged using a scanning electron beam. To confirm the accuracy of single cross sections, nanoparticles in similar cross sections were imaged using transmission electron microscopy and scanning helium ion microscopy. Complete tomographic series were then used to count the nanoparticles inside of each cell and measure their spatial distribution. We investigated the influence of slice thickness on counting single particles and clusters as well as nanoparticle packing within clusters. For 60 nm citrate stabilized particles, the nanoparticle cluster packing volume is 2.15 ± 0.20 times the volume of the bare gold nanoparticles.
Collapse
Affiliation(s)
- Aric W Sanders
- Quantum Electronics and Photonics Division, National Institute of Standards and Technology (NIST) , Boulder, Colorado 59840, United States
| | - Kavita M Jeerage
- Applied Chemicals and Materials Division, National Institute of Standards and Technology (NIST) , Boulder, Colorado 59840, United States
| | - Cindi L Schwartz
- Department of Molecular, Cell, and Developmental Biology, University of Colorado , Boulder, Colorado 59840, United States
| | - Alexandra E Curtin
- Quantum Electronics and Photonics Division, National Institute of Standards and Technology (NIST) , Boulder, Colorado 59840, United States
- Applied Chemicals and Materials Division, National Institute of Standards and Technology (NIST) , Boulder, Colorado 59840, United States
| | - Ann N Chiaramonti
- Applied Chemicals and Materials Division, National Institute of Standards and Technology (NIST) , Boulder, Colorado 59840, United States
| |
Collapse
|
32
|
Palma SICJ, Rodrigues CAV, Carvalho A, Morales MDP, Freitas F, Fernandes AR, Cabral JMS, Roque ACA. A value-added exopolysaccharide as a coating agent for MRI nanoprobes. NANOSCALE 2015; 7:14272-14283. [PMID: 26186402 DOI: 10.1039/c5nr01979f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Fucopol, a fucose-containing exopolysaccharide (EPS) produced by the bacterium Enterobacter A47 DSM 23139 using glycerol as a carbon source, was employed as a new coating material for iron oxide magnetic nanoparticles (MNPs). The coated particles were assessed as nanoprobes for cell labeling by Magnetic Resonance Imaging (MRI). The MNPs were synthesized by a thermal decomposition method and transferred to an aqueous medium by a ligand-exchange reaction with meso-2,3-dimercaptosuccinic acid (DMSA). Covalent binding of EPS to DMSA-stabilized nanoparticles (MNP-DMSA) resulted in a hybrid magnetic-biopolymeric nanosystem (MNP-DMSA-EPS) with a hydrodynamic size of 170 nm, a negative surface charge under physiological conditions and transverse to longitudinal relaxivity ratio, r2/r1, of 148. In vitro studies with two human cell lines (colorectal carcinoma - HCT116 - and neural stem/progenitor cells - ReNcell VM) showed that EPS promotes internalization of nanoparticles in both cell lines. In vitro MRI cell phantoms showed a superior performance of MNP-DMSA-EPS in ReNcell VM, for which the iron dose-dependent MRI signal drop was obtained at relatively low iron concentrations (12-20 μg Fe per ml) and short incubation times. Furthermore, ReNcell VM multipotency was not affected by culture in the presence of MNP-DMSA or MNP-DMSA-EPS for 14 days. Our study suggests that Fucopol-coated MNPs represent useful cell labeling nanoprobes for MRI.
Collapse
Affiliation(s)
- Susana I C J Palma
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Pinkernelle J, Raffa V, Calatayud MP, Goya GF, Riggio C, Keilhoff G. Growth factor choice is critical for successful functionalization of nanoparticles. Front Neurosci 2015; 9:305. [PMID: 26388717 PMCID: PMC4557102 DOI: 10.3389/fnins.2015.00305] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/12/2015] [Indexed: 12/16/2022] Open
Abstract
Nanoparticles (NPs) show new characteristics compared to the corresponding bulk material. These nanoscale properties make them interesting for various applications in biomedicine and life sciences. One field of application is the use of magnetic NPs to support regeneration in the nervous system. Drug delivery requires a functionalization of NPs with bio-functional molecules. In our study, we functionalized self-made PEI-coated iron oxide NPs with nerve growth factor (NGF) and glial cell-line derived neurotrophic factor (GDNF). Next, we tested the bio-functionality of NGF in a rat pheochromocytoma cell line (PC12) and the bio-functionality of GDNF in an organotypic spinal cord culture. Covalent binding of NGF to PEI-NPs impaired bio-functionality of NGF, but non-covalent approach differentiated PC12 cells reliably. Non-covalent binding of GDNF showed a satisfying bio-functionality of GDNF:PEI-NPs, but turned out to be unstable in conjugation to the PEI-NPs. Taken together, our study showed the importance of assessing bio-functionality and binding stability of functionalized growth factors using proper biological models. It also shows that successful functionalization of magnetic NPs with growth factors is dependent on the used binding chemistry and that it is hardly predictable. For use as therapeutics, functionalization strategies have to be reproducible and future studies are needed.
Collapse
Affiliation(s)
- Josephine Pinkernelle
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
- Institute for Biochemistry and Cell Biology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
| | - Vittoria Raffa
- Department of Biology, University of PisaPisa, Italy
- Institute of Life Science, Scuola Superiore Sant' AnnaPisa, Italy
| | | | - Gerado F. Goya
- Aragon Institute of Nanosciences, University of ZaragozaZaragoza, Spain
- Department of Condensed Matter Physics, University of ZaragozaSpain
| | - Cristina Riggio
- Institute of Life Science, Scuola Superiore Sant' AnnaPisa, Italy
| | - Gerburg Keilhoff
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
| |
Collapse
|
34
|
Petters C, Thiel K, Dringen R. Lysosomal iron liberation is responsible for the vulnerability of brain microglial cells to iron oxide nanoparticles: comparison with neurons and astrocytes. Nanotoxicology 2015; 10:332-42. [DOI: 10.3109/17435390.2015.1071445] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Charlotte Petters
- Center for Biomedical Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany,
- Center for Environmental Research and Sustainable Technology, Bremen, Germany, and
| | - Karsten Thiel
- Fraunhofer Institute for Manufacturing Technology and Advanced Materials, Bremen, Germany
| | - Ralf Dringen
- Center for Biomedical Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany,
- Center for Environmental Research and Sustainable Technology, Bremen, Germany, and
| |
Collapse
|
35
|
Keilhoff G, Titze M, Esser T, Langnaese K, Ebmeyer U. Constitutive and functional expression of YB-1 in microglial cells. Neuroscience 2015; 301:439-53. [DOI: 10.1016/j.neuroscience.2015.06.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 12/28/2022]
|
36
|
Simkó M, Mattsson MO. Interactions between nanosized materials and the brain. Curr Med Chem 2015; 21:4200-14. [PMID: 25039776 PMCID: PMC4435026 DOI: 10.2174/0929867321666140716100449] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/04/2014] [Accepted: 07/11/2014] [Indexed: 12/21/2022]
Abstract
The current rapid development of nanotechnologies and engineered nanomaterials (ENM) will impact the society in a major fashion during the coming decades. This development also causes substantial safety concerns. Among the many promising applications of ENM, products that can be used for diagnosis and treatment of diseases, including conditions that affect the nervous system, are under development. ENM can pass the blood brain barrier (BBB) and accumulate within the brain. It seems that the nano-form rather than the bulk form of the chemicals pass the BBB, and that there is an inverse relationship between particle size and the ability to penetrate the BBB. Although translocation of ENM to the brain is possible during experimental conditions, the health relevance for real-life situations is far from clear. One major reason for this is that studies have been using nanoparticle concentrations that are far higher than the ones that can be expected during realistic exposures. However, very high exposure to the CNS can cause effects on neurotransmission, redox homeostasis and behavior. Available studies have been focusing on possible effects of the first generation of ENM. It will be necessary to study possible health effects also of expected novel sophisticated materials, independent of the outcome of present studies. The prospects for intended or targeted medical applications are promising since it has been shown that ENM can be made to pass the BBB and reach specific regions or cells within the brain.
Collapse
Affiliation(s)
| | - Mats-Olof Mattsson
- Health and Environment Department, Environmental Resources and Technologies, Austrian Institute of Technology, Konrad-Lorenz-Strasse 24, A-3430 Tulln, Austria.
| |
Collapse
|
37
|
Fernandes AR, Chari DM. A multicellular, neuro-mimetic model to study nanoparticle uptake in cells of the central nervous system. Integr Biol (Camb) 2015; 6:855-61. [PMID: 25017718 DOI: 10.1039/c4ib00085d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Evaluating the uptake and handling of biomedically relevant nanoparticles by cells of the nervous system critically underpins the effective use of nanoparticle platforms for neuro-regenerative therapies. The lack of biologically relevant and 'neuromimetic' models for nanomaterials testing (that can simulate the cellular complexity of neural tissue) currently represents a bottleneck. Further, propagation of individual cell types, in different neural cell-specific media (as commonly occurs in the nanotechnology field), can result in non-standardised corona formation around particles, confounding analyses of intercellular differences between neural cells in nanoparticle uptake. To address these challenges, we have developed a facile multicellular model that broadly simulates the ratios of neurons, astrocytes and oligodendrocytes found in vivo. All cell types in the model are derived from a single neural stem cell source, and propagated in the same medium overcoming the issue of variant corona formation. Using a fluorescent transfection-grade magnetic particle (MP), we demonstrate dramatic differences in particle uptake and resultant gene transfer between neural cell subtypes, with astrocytes being the dominant population in terms of particle uptake and transfection. We demonstrate the compatibility of the model with a high resolution scanning electron microscopy technique, allowing for membrane features of MP stimulated cells to be examined. Using this approach, astrocytes displayed high membrane activity in line with extensive particle uptake/transfection, relative to neurons and oligodendrocytes. We consider that the stem cell based model described here can provide a simple and versatile tool to evaluate interactions of neural cells with nanoparticle systems developed for neurological applications. Models of greater complexity can be evolved from this basic system, to further enhance its neuromimetic capacity.
Collapse
Affiliation(s)
- A R Fernandes
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, UK.
| | | |
Collapse
|
38
|
Neubert J, Wagner S, Kiwit J, Bräuer AU, Glumm J. New findings about iron oxide nanoparticles and their different effects on murine primary brain cells. Int J Nanomedicine 2015; 10:2033-49. [PMID: 25792834 PMCID: PMC4364595 DOI: 10.2147/ijn.s74404] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The physicochemical properties of superparamagnetic iron oxide nanoparticles (SPIOs) enable their application in the diagnostics and therapy of central nervous system diseases. However, since crucial information regarding side effects of particle–cell interactions within the central nervous system is still lacking, we investigated the influence of novel very small iron oxide particles or the clinically approved ferucarbotran or ferumoxytol on the vitality and morphology of brain cells. We exposed primary cell cultures of microglia and hippocampal neurons, as well as neuron–glia cocultures to varying concentrations of SPIOs for 6 and/or 24 hours, respectively. Here, we show that SPIO accumulation by microglia and subsequent morphological alterations strongly depend on the respective nanoparticle type. Microglial viability was severely compromised by high SPIO concentrations, except in the case of ferumoxytol. While ferumoxytol did not cause immediate microglial death, it induced severe morphological alterations and increased degeneration of primary neurons. Additionally, primary neurons clearly degenerated after very small iron oxide particle and ferucarbotran exposure. In neuron–glia cocultures, SPIOs rather stimulated the outgrowth of neuronal processes in a concentration- and particle-dependent manner. We conclude that the influence of SPIOs on brain cells not only depends on the particle type but also on the physiological system they are applied to.
Collapse
Affiliation(s)
- Jenni Neubert
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Susanne Wagner
- Institute for Radiology, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Jürgen Kiwit
- Clinic for Neurosurgery, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Anja U Bräuer
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Jana Glumm
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitaetsmedizin Berlin, Berlin, Germany ; Clinic for Neurosurgery, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| |
Collapse
|
39
|
Petters C, Dringen R. Accumulation of iron oxide nanoparticles by cultured primary neurons. Neurochem Int 2015; 81:1-9. [DOI: 10.1016/j.neuint.2014.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 12/03/2014] [Accepted: 12/09/2014] [Indexed: 01/13/2023]
|
40
|
Kunze A, Tseng P, Godzich C, Murray C, Caputo A, Schweizer FE, Di Carlo D. Engineering cortical neuron polarity with nanomagnets on a chip. ACS NANO 2015; 9:3664-76. [PMID: 25801533 DOI: 10.1021/nn505330w] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Intra- and extracellular signaling play critical roles in cell polarity, ultimately leading to the development of functional cell-cell connections, tissues, and organs. In the brain, pathologically oriented neurons are often the cause for disordered circuits, severely impacting motor function, perception, and memory. Aside from control through gene expression and signaling pathways, it is known that nervous system development can be manipulated by mechanical stimuli (e.g., outgrowth of axons through externally applied forces). The inverse is true as well: intracellular molecular signals can be converted into forces to yield axonal outgrowth. The complete role played by mechanical signals in mediating single-cell polarity, however, remains currently unclear. Here we employ highly parallelized nanomagnets on a chip to exert local mechanical stimuli on cortical neurons, independently of the amount of superparamagnetic nanoparticles taken up by the cells. The chip-based approach was utilized to quantify the effect of nanoparticle-mediated forces on the intracellular cytoskeleton as visualized by the distribution of the microtubule-associated protein tau. While single cortical neurons prefer to assemble tau proteins following poly-L-lysine surface cues, an optimal force range of 4.5-70 pN by the nanomagnets initiated a tau distribution opposed to the pattern cue. In larger cell clusters (groups comprising six or more cells), nanoparticle-mediated forces induced tau repositioning in an observed range of 190-270 pN, and initiation of magnetic field-directed cell displacement was observed at forces above 300 pN. Our findings lay the groundwork for high-resolution mechanical encoding of neural networks in vitro, mechanically driven cell polarization in brain tissues, and neurotherapeutic approaches using functionalized superparamagnetic nanoparticles to potentially restore disordered neural circuits.
Collapse
Affiliation(s)
- Anja Kunze
- †Department of Bioengineering, ‡California NanoSystems Institute, and §Department of Neurobiology, University of California, Los Angeles, California 90095, United States
| | - Peter Tseng
- †Department of Bioengineering, ‡California NanoSystems Institute, and §Department of Neurobiology, University of California, Los Angeles, California 90095, United States
| | - Chanya Godzich
- †Department of Bioengineering, ‡California NanoSystems Institute, and §Department of Neurobiology, University of California, Los Angeles, California 90095, United States
| | - Coleman Murray
- †Department of Bioengineering, ‡California NanoSystems Institute, and §Department of Neurobiology, University of California, Los Angeles, California 90095, United States
| | - Anna Caputo
- †Department of Bioengineering, ‡California NanoSystems Institute, and §Department of Neurobiology, University of California, Los Angeles, California 90095, United States
| | - Felix E Schweizer
- †Department of Bioengineering, ‡California NanoSystems Institute, and §Department of Neurobiology, University of California, Los Angeles, California 90095, United States
| | - Dino Di Carlo
- †Department of Bioengineering, ‡California NanoSystems Institute, and §Department of Neurobiology, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
41
|
Development of a nanomaterial bio-screening platform for neurological applications. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:77-87. [DOI: 10.1016/j.nano.2014.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 06/05/2014] [Accepted: 07/22/2014] [Indexed: 11/23/2022]
|
42
|
Jenkins SI, Yiu HHP, Rosseinsky MJ, Chari DM. Magnetic nanoparticles for oligodendrocyte precursor cell transplantation therapies: progress and challenges. MOLECULAR AND CELLULAR THERAPIES 2014; 2:23. [PMID: 26056590 PMCID: PMC4452053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/20/2014] [Indexed: 11/21/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) have shown high promise as a transplant population to promote regeneration in the central nervous system, specifically, for the production of myelin - the protective sheath around nerve fibers. While clinical trials for these cells have commenced in some areas, there are currently key barriers to the translation of neural cell therapies. These include the ability to (a) image transplant populations in vivo; (b) genetically engineer transplant cells to augment their repair potential; and (c) safely target cells to sites of pathology. Here, we review the evidence that magnetic nanoparticles (MNPs) are a 'multifunctional nanoplatform' that can aid in safely addressing these translational challenges in neural cell/OPC therapy: by facilitating real-time and post-mortem assessment of transplant cell biodistribution, and biomolecule delivery to transplant cells, as well as non-invasive 'magnetic cell targeting' to injury sites by application of high gradient fields. We identify key issues relating to the standardization and reporting of physicochemical and biological data in the field; we consider that it will be essential to systematically address these issues in order to fully evaluate the utility of the MNP platform for neural cell transplantation, and to develop efficacious neurocompatible particles for translational applications.
Collapse
Affiliation(s)
- Stuart I Jenkins
- />Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| | - Humphrey H P Yiu
- />School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS UK
| | | | - Divya M Chari
- />Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| |
Collapse
|
43
|
Jenkins SI, Yiu HHP, Rosseinsky MJ, Chari DM. Magnetic nanoparticles for oligodendrocyte precursor cell transplantation therapies: progress and challenges. MOLECULAR AND CELLULAR THERAPIES 2014; 2:23. [PMID: 26056590 PMCID: PMC4452053 DOI: 10.1186/2052-8426-2-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/20/2014] [Indexed: 01/12/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) have shown high promise as a transplant population to promote regeneration in the central nervous system, specifically, for the production of myelin – the protective sheath around nerve fibers. While clinical trials for these cells have commenced in some areas, there are currently key barriers to the translation of neural cell therapies. These include the ability to (a) image transplant populations in vivo; (b) genetically engineer transplant cells to augment their repair potential; and (c) safely target cells to sites of pathology. Here, we review the evidence that magnetic nanoparticles (MNPs) are a ‘multifunctional nanoplatform’ that can aid in safely addressing these translational challenges in neural cell/OPC therapy: by facilitating real-time and post-mortem assessment of transplant cell biodistribution, and biomolecule delivery to transplant cells, as well as non-invasive ‘magnetic cell targeting’ to injury sites by application of high gradient fields. We identify key issues relating to the standardization and reporting of physicochemical and biological data in the field; we consider that it will be essential to systematically address these issues in order to fully evaluate the utility of the MNP platform for neural cell transplantation, and to develop efficacious neurocompatible particles for translational applications.
Collapse
Affiliation(s)
- Stuart I Jenkins
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| | - Humphrey H P Yiu
- School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS UK
| | | | - Divya M Chari
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine Keele University, Stoke-on-Trent, Staffordshire ST5 5BG UK
| |
Collapse
|
44
|
Petters C, Irrsack E, Koch M, Dringen R. Uptake and metabolism of iron oxide nanoparticles in brain cells. Neurochem Res 2014; 39:1648-60. [PMID: 25011394 DOI: 10.1007/s11064-014-1380-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 01/29/2023]
Abstract
Magnetic iron oxide nanoparticles (IONPs) are used for various applications in biomedicine, for example as contrast agents in magnetic resonance imaging, for cell tracking and for anti-tumor treatment. However, IONPs are also known for their toxic effects on cells and tissues which are at least in part caused by iron-mediated radical formation and oxidative stress. The potential toxicity of IONPs is especially important concerning the use of IONPs for neurobiological applications as alterations in brain iron homeostasis are strongly connected with human neurodegenerative diseases. Since IONPs are able to enter the brain, potential adverse consequences of an exposure of brain cells to IONPs have to be considered. This article describes the pathways that allow IONPs to enter the brain and summarizes the current knowledge on the uptake, the metabolism and the toxicity of IONPs for the different types of brain cells in vitro and in vivo.
Collapse
Affiliation(s)
- Charlotte Petters
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | | | | | | |
Collapse
|
45
|
Keilhoff G, Lucas B, Pinkernelle J, Steiner M, Fansa H. Effects of cerebrolysin on motor-neuron-like NSC-34 cells. Exp Cell Res 2014; 327:234-55. [PMID: 24997385 DOI: 10.1016/j.yexcr.2014.06.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/12/2014] [Accepted: 06/26/2014] [Indexed: 01/01/2023]
Abstract
Although the peripheral nervous system is capable of regeneration, this capability is limited. As a potential means of augmenting nerve regeneration, the effects of cerebrolysin (CL)--a proteolytic peptide fraction--were tested in vitro on the motor-neuron-like NSC-34 cell line and organotypic spinal cord cultures. Therefore, NSC-34 cells were subjected to mechanical stress by changing media and metabolic stress by oxygen glucose deprivation. Afterwards, cell survival/proliferation using MTT and BrdU-labeling (FACS) and neurite sprouting using ImageJ analysis were evaluated. Calpain-1, Src and α-spectrin protein expression were analyzed by Western blot. In organotypic cultures, the effect of CL on motor neuron survival and neurite sprouting was tested by immunohistochemistry. CL had a temporary anti-proliferative but initially neuroprotective effect on OGD-stressed NSC-34 cells. High-dosed or repeatedly applied CL was deleterious for cell survival. CL amplified neurite reconstruction to limited extent, affected calpain-1 protein expression and influenced calpain-mediated spectrin cleavage as a function of Src expression. In organotypic spinal cord slice cultures, CL was not able to support motor neuron survival/neurite sprouting. Moreover, it hampered astroglia and microglia activities. The data suggest that CL may have only isolated positive effects on injured spinal motor neurons. High-dosed or accumulated CL seemed to have adverse effects in treatment of spinal cord injury. Further experiments are required to optimize the conditions for a safe clinical administration of CL in spinal cord injuries.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Benjamin Lucas
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Josephine Pinkernelle
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Michael Steiner
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Hisham Fansa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld, Teutoburger Str. 50, D-33604 Bielefeld, Germany
| |
Collapse
|
46
|
Koch F, Möller AM, Frenz M, Pieles U, Kuehni-Boghenbor K, Mevissen M. An in vitro toxicity evaluation of gold-, PLLA- and PCL-coated silica nanoparticles in neuronal cells for nanoparticle-assisted laser-tissue soldering. Toxicol In Vitro 2014; 28:990-8. [PMID: 24768613 DOI: 10.1016/j.tiv.2014.04.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/03/2014] [Accepted: 04/14/2014] [Indexed: 12/14/2022]
Abstract
The uptake of silica (Si) and gold (Au) nanoparticles (NPs) engineered for laser-tissue soldering in the brain was investigated using microglial cells and undifferentiated and differentiated SH-SY5Y cells. It is not known what effects NPs elicit once entering the brain. Cellular uptake, cytotoxicity, apoptosis, and the potential induction of oxidative stress by means of depletion of glutathione levels were determined after NP exposure at concentrations of 10(3) and 10(9)NPs/ml. Au-, silica poly (ε-caprolactone) (Si-PCL-) and silica poly-L-lactide (Si-PLLA)-NPs were taken up by all cells investigated. Aggregates and single NPs were found in membrane-surrounded vacuoles and the cytoplasm, but not in the nucleus. Both NP concentrations investigated did not result in cytotoxicity or apoptosis, but reduced glutathione (GSH) levels predominantly at 6 and 24h, but not after 12 h of NP exposure in the microglial cells. NP exposure-induced GSH depletion was concentration-dependent in both cell lines. Si-PCL-NPs induced the strongest effect of GSH depletion followed by Si-PLLA-NPs and Au-NPs. NP size seems to be an important characteristic for this effect. Overall, Au-NPs are most promising for laser-assisted vascular soldering in the brain. Further studies are necessary to further evaluate possible effects of these NPs in neuronal cells.
Collapse
Affiliation(s)
- Franziska Koch
- Veterinary Pharmacology & Toxicology, Vetsuisse Faculty University Bern, Länggassstrasse 124, 3012 Bern, Switzerland
| | - Anja-M Möller
- Veterinary Pharmacology & Toxicology, Vetsuisse Faculty University Bern, Länggassstrasse 124, 3012 Bern, Switzerland
| | - Martin Frenz
- Institute of Applied Physics, University Bern, Sidlerstrasse 5, 3012 Bern, Switzerland
| | - Uwe Pieles
- School of Life Sciences, Gruendenstrasse 40, 4132 Muttenz, Switzerland
| | - Kathrin Kuehni-Boghenbor
- Veterinary Anatomy, Vetsuisse Faculty University Bern, Laenggassstrasse 120, 3012 Bern, Switzerland
| | - Meike Mevissen
- Veterinary Pharmacology & Toxicology, Vetsuisse Faculty University Bern, Länggassstrasse 124, 3012 Bern, Switzerland.
| |
Collapse
|
47
|
Petters C, Bulcke F, Thiel K, Bickmeyer U, Dringen R. Uptake of Fluorescent Iron Oxide Nanoparticles by Oligodendroglial OLN-93 Cells. Neurochem Res 2013; 39:372-83. [DOI: 10.1007/s11064-013-1234-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 12/30/2022]
|
48
|
Diana V, Bossolasco P, Moscatelli D, Silani V, Cova L. Dose dependent side effect of superparamagnetic iron oxide nanoparticle labeling on cell motility in two fetal stem cell populations. PLoS One 2013; 8:e78435. [PMID: 24244310 PMCID: PMC3820601 DOI: 10.1371/journal.pone.0078435] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022] Open
Abstract
Multipotent stem cells (SCs) could substitute damaged cells and also rescue degeneration through the secretion of trophic factors able to activate the endogenous SC compartment. Therefore, fetal SCs, characterized by high proliferation rate and devoid of ethical concern, appear promising candidate, particularly for the treatment of neurodegenerative diseases. Super Paramagnetic Iron Oxide nanoparticles (SPIOn), routinely used for pre-clinical cell imaging and already approved for clinical practice, allow tracking of transplanted SCs and characterization of their fate within the host tissue, when combined with Magnetic Resonance Imaging (MRI). In this work we investigated how SPIOn could influence cell migration after internalization in two fetal SC populations: human amniotic fluid and chorial villi SCs were labeled with SPIOn and their motility was evaluated. We found that SPIOn loading significantly reduced SC movements without increasing production of Reactive Oxygen Species (ROS). Moreover, motility impairment was directly proportional to the amount of loaded SPIOn while a chemoattractant-induced recovery was obtained by increasing serum levels. Interestingly, the migration rate of SPIOn labeled cells was also significantly influenced by a degenerative surrounding. In conclusion, this work highlights how SPIOn labeling affects SC motility in vitro in a dose-dependent manner, shedding the light on an important parameter for the creation of clinical protocols. Establishment of an optimal SPIOn dose that enables both a good visualization of grafted cells by MRI and the physiological migration rate is a main step in order to maximize the effects of SC therapy in both animal models of neurodegeneration and clinical studies.
Collapse
Affiliation(s)
- Valentina Diana
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Italy
| | - Patrizia Bossolasco
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Italy
| | - Davide Moscatelli
- Department of Chimica Materiali e Ingegneria Chimica G. Natta, Politecnico di Milano, Milan, Italy
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Italy
- Department of Fisiopatologia Medico-Chirurgica e dei Trapianti, “Dino Ferrari” Center, Università degli Studi di Milano, Milan, Italy
| | - Lidia Cova
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Italy
- * E-mail:
| |
Collapse
|
49
|
Petters C, Dringen R. Comparison of primary and secondary rat astrocyte cultures regarding glucose and glutathione metabolism and the accumulation of iron oxide nanoparticles. Neurochem Res 2013; 39:46-58. [PMID: 24190598 DOI: 10.1007/s11064-013-1189-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 10/24/2013] [Accepted: 10/25/2013] [Indexed: 12/31/2022]
Abstract
Astrocyte-rich primary cultures (APCs) are frequently used as a model system for the investigation of properties of brain astrocytes. However, as APCs contain a substantial number of microglial and oligodendroglial cells, biochemical parameters determined for such cultures may at least in part reflect also the presence of the contaminating cell types. To lower the potential contributions of microglial and oligodendroglial cells on properties of the astrocytes in APCs we prepared rat astrocyte-rich secondary cultures (ASCs) by subculturing of APCs and compared these ASCs with APCs regarding basal metabolic parameters, specific enzyme activities and the accumulation of iron oxide nanoparticles. Immunocytochemical characterization revealed that ASCs contained only minute amounts of microglial and oligodendroglial cells. ASCs and APCs did not significantly differ in their specific glucose consumption and lactate production rates, in their specific iron and glutathione contents, in their specific activities of various enzymes involved in glucose and glutathione metabolism nor in their accumulation of iron oxide nanoparticles. Thus, the absence or presence of some contaminating microglial and oligodendroglial cells appears not to substantially modulate the investigated metabolic parameters of astrocyte cultures.
Collapse
Affiliation(s)
- Charlotte Petters
- Center for Biomolecular Interactions Bremen, University of Bremen, PO. Box 330440, 28334, Bremen, Germany
| | | |
Collapse
|
50
|
Luther EM, Petters C, Bulcke F, Kaltz A, Thiel K, Bickmeyer U, Dringen R. Endocytotic uptake of iron oxide nanoparticles by cultured brain microglial cells. Acta Biomater 2013; 9:8454-65. [PMID: 23727247 DOI: 10.1016/j.actbio.2013.05.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Revised: 05/17/2013] [Accepted: 05/21/2013] [Indexed: 11/29/2022]
Abstract
Microglia are the phagocytotic cells of the brain that respond rapidly to alterations in brain homeostasis. Since iron oxide nanoparticles (IONPs) are used for diagnostic and therapeutic applications in the brain, the consequences of an exposure of microglial cells to IONPs are of particular interest. To address this topic we have synthesized and characterized fluorescent BODIPY®-labelled IONPs (BP-IONPs). The average hydrodynamic diameter and the ζ-potential of BP-IONPs in water were ∼65 nm and -49 mV, respectively. Both values increased after dispersion of the particles in serum containing incubation medium to ∼130 nm and -8 mV. Exposure of cultured rat microglial cells with BP-IONPs caused a time-, concentration- and temperature-dependent uptake of the particles, as demonstrated by strong increases in cellular iron contents and cellular fluorescence. Incubation for 3h with 150 and 450 μM iron as BP-IONPs increased the cellular iron content from a low basal level of ∼50 nmol iron mg(-1) to 219±52 and 481±28 nmol iron (mg protein)(-1), respectively. These conditions did not affect cell viability, but exposure to higher concentrations of BP-IONPs or for longer incubation periods severely compromised cell viability. The BP-IONP fluorescence in viable microglial cells was co-localized with lysosomes. In addition, BP-IONP accumulation was lowered by 60% in the presence of the endocytosis inhibitors 5-(N-ethyl-N-isopropyl)amiloride, tyrphostin23 and chlorpromazin. These results suggest that the rapid accumulation of BP-IONPs by microglial cells is predominantly mediated by macropinocytosis and clathrin-mediated endocytosis, which direct the accumulated particles into the lysosomal compartment.
Collapse
Affiliation(s)
- Eva M Luther
- Center for Biomolecular Interactions Bremen, University of Bremen, P.O. Box 330440, D-28334 Bremen, Germany
| | | | | | | | | | | | | |
Collapse
|