Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mo Dan is active.

Publication


Featured researches published by Mo Dan.


Toxicological Sciences | 2010

Brain Distribution and Toxicological Evaluation of a Systemically Delivered Engineered Nanoscale Ceria

Sarita S. Hardas; D.A. Butterfield; Rukhsana Sultana; Michael T. Tseng; Mo Dan; Rebecca L. Florence; Jason M. Unrine; Uschi M. Graham; Peng Wu; Eric A. Grulke; Robert A. Yokel

Engineered nanoscale ceria is used as a diesel fuel catalyst. Little is known about its mammalian central nervous system effects. The objective of this paper is to characterize the biodistribution of a 5-nm citrate-stabilized ceria dispersion from blood into brain and its pro- or antioxidant effects. An approximately 4% aqueous ceria dispersion was iv infused into rats (0, 100, and up to 250 mg/kg), which were terminated after 1 or 20 h. Ceria concentration, localization, and chemical speciation in the brain were assessed by inductively coupled plasma mass spectrometry, light and electron microscopy (EM), and electron energy loss spectroscopy (EELS). Pro- or antioxidative stress effects were assessed as protein carbonyls, 3-nitrotyrosine, and protein-bound 4-hydroxy-2-trans-nonenal in hippocampus, cortex, and cerebellum. Glutathione reductase, glutathione peroxidase, manganese superoxide dismutase, and catalase levels and activities were measured in hippocampus. Catalase levels and activities were also measured in cortex and cerebellum. Na fluorescein and horseradish peroxidase (HRP) were given iv as blood-brain barrier (BBB) integrity markers. Mortality was seen after administration of 175-250 mg ceria/kg. Twenty hours after infusion of 100 mg ceria/kg, brain HRP was marginally elevated. EM and EELS revealed mixed Ce(III) and Ce(IV) valence in the freshly synthesized ceria in vitro and in ceria agglomerates in the brain vascular compartment. Ceria was not seen in microvascular endothelial or brain cells. Ceria elevated catalase levels at 1 h and increased catalase activity at 20 h in hippocampus and decreased catalase activity at 1 h in cerebellum. Compared with a previously studied approximately 30-nm ceria, this ceria was more toxic, was not seen in the brain, and produced little oxidative stress effect to the hippocampus and cerebellum. The results are contrary to the hypothesis that a smaller engineered nanomaterial would more readily permeate the BBB.


Toxicological Sciences | 2012

Distribution, Elimination, and Biopersistence to 90 Days of a Systemically Introduced 30 nm Ceria-Engineered Nanomaterial in Rats

Robert A. Yokel; Tu C. Au; Robert C. MacPhail; Sarita S. Hardas; D. Allan Butterfield; Rukhsana Sultana; Michael Goodman; Michael T. Tseng; Mo Dan; Hamed Haghnazar; Jason M. Unrine; Uschi M. Graham; Peng Wu; Eric A. Grulke

Nanoceria is used as a catalyst in diesel fuel, as an abrasive in printed circuit manufacture, and is being pursued as an antioxidant therapeutic. Our objective is to extend previous findings showing that there were no reductions of cerium in organs of the mononuclear phagocyte (reticuloendothelial) system up to 30 days after a single nanoscale ceria administration. An ~5% aqueous dispersion of citrate-stabilized 30 nm ceria, synthesized and characterized in-house, or vehicle, was iv infused into rats terminated 1, 7, 30, or 90 days later. Cageside observations were obtained daily, body weight weekly. Daily urinary and fecal cerium outputs were quantified for 2 weeks. Nine organs were weighed and samples collected from 14 tissues/organs/systems, blood and cerebrospinal fluid for cerium determination. Histology and oxidative stress were assessed. Less than 1% of the nanoceria was excreted in the first 2 weeks, 98% in feces. Body weight gain was initially impaired. Spleen weight was significantly increased in some ceria-treated groups, associated with abnormalities. Ceria was primarily retained in the spleen, liver, and bone marrow. There was little decrease of ceria in any tissue over the 90 days. Granulomas were observed in the liver. Time-dependent oxidative stress changes were seen in the liver and spleen. Nanoscale ceria was persistently retained by organs of the mononuclear phagocyte system, associated with adverse changes. The results support concern about the long-term fate and adverse effects of inert nanoscale metal oxides that distribute throughout the body, are persistently retained, and produce adverse changes.


Nanomedicine: Nanotechnology, Biology and Medicine | 2013

Biodistribution and biopersistence of ceria engineered nanomaterials: size dependence

Robert A. Yokel; Michael T. Tseng; Mo Dan; Jason M. Unrine; Uschi M. Graham; Peng Wu; Eric A. Grulke

UNLABELLED The aims were to determine the biodistribution, translocation, and persistence of nanoceria in the brain and selected peripheral organs. Nanoceria is being studied as an anti-oxidant therapeutic. Five, 15, 30, or 55 nm ceria was iv infused into rats which were terminated 1, 20, or 720 h later. Cerium was determined in blood, brain, liver, and spleen. Liver and spleen contained a large percentage of the dose, from which there was no significant clearance over 720 h, associated with adverse changes. Very little nanoceria entered brain parenchyma. The results suggest brain delivery of nanoceria will be a challenge. FROM THE CLINICAL EDITOR This team of investigators revealed that nanoceria, which is being studied as an anti-oxidant, has very limited uptake by the brain regardless of the range of sizes studied, suggesting major challenges in the application of this novel approach in the central nervous system.


Nanomedicine: Nanotechnology, Biology and Medicine | 2012

Ceria-engineered nanomaterial distribution in, and clearance from, blood: size matters

Mo Dan; Peng Wu; Eric A. Grulke; Uschi M. Graham; Jason M. Unrine; Robert A. Yokel

AIMS Characterize different sized ceria-engineered nanomaterial (ENM) distribution in, and clearance from, blood (compared to the cerium ion) following intravenous infusion. MATERIALS & METHODS Cerium (Ce) was quantified in whole blood, serum and clot (the formed elements) up to 720 h. RESULTS Traditional pharmacokinetic modeling showed best fit for 5 nm ceria ENM and the cerium ion. Ceria ENMs larger than 5 nm were rapidly cleared from blood. After initially declining, whole blood 15 and 30 nm ceria increased (results that have not been well-described by traditional pharmacokinetic modeling). The cerium ion and 5 and 55 nm ceria did not preferentially distribute into serum or clot, a mixture of cubic and rod shaped ceria was predominantly in the clot, and 15 and 30 nm ceria migrated into the clot over 4 h. CONCLUSION Reticuloendothelial organs may not readily recognize five nm ceria. Increased ceria distribution into the clot over time may be due to opsonization. Traditional pharmacokinetic analysis was not very informative. Ceria ENM pharmacokinetics are quite different from the cerium ion.


Neurotoxicology | 2012

Rat brain pro-oxidant effects of peripherally administered 5 nm ceria 30 days after exposure

Sarita S. Hardas; Rukhsana Sultana; Govind Warrier; Mo Dan; Rebecca L. Florence; Peng Wu; Eric A. Grulke; Michael T. Tseng; Jason M. Unrine; Uschi M. Graham; Robert A. Yokel; D. Allan Butterfield

The objective of this study was to determine the residual pro-or anti-oxidant effects in rat brain 30 days after systemic administration of a 5 nm citrate-stabilized ceria dispersion. A ∼4% aqueous ceria dispersion was iv-infused (0 or 85 mg/kg) into rats which were terminated 30 days later. Ceria concentration, localization, and chemical speciation in the brain was assessed by inductively coupled plasma mass spectrometry (ICP-MS), light and electron microscopy (EM), and electron energy loss spectroscopy (EELS), respectively. Pro- or anti-oxidant effects were evaluated by measuring levels of protein carbonyls (PC), 3-nitrotyrosine (3NT), and protein-bound-4-hydroxy-2-trans-nonenal (HNE) in the hippocampus, cortex, and cerebellum. Glutathione reductase (GR), glutathione peroxidase (GPx), superoxide dismutase (SOD), and catalase levels and activity were measured in addition to levels of inducible nitric oxide (iNOS), and heat shock protein-70 (Hsp70). The blood brain barrier (BBB) was visibly intact and no ceria was seen in the brain cells. Ceria elevated PC and Hsp70 levels in hippocampus and cerebellum, while 3NT and iNOS levels were elevated in the cortex. Whereas glutathione peroxidase and catalase activity were decreased in the hippocampus, GR levels were decreased in the cortex, and GPx and catalase levels were decreased in the cerebellum. The GSH:GSSG ratio, an index of cellular redox status, was decreased in the hippocampus and cerebellum. The results are in accordance with the observation that this nanoscale material remains in this mammal model up to 30 days after its administration and the hypothesis that it exerts pro-oxidant effects on the brain without crossing the BBB. These results have important implications on the potential use of ceria ENM as therapeutic agents.


International Journal of Nanomedicine | 2015

Silver nanoparticles induce tight junction disruption and astrocyte neurotoxicity in a rat blood-brain barrier primary triple coculture model.

Liming Xu; Mo Dan; Anliang Shao; Xiang Cheng; Cuiping Zhang; Robert A. Yokel; Taro Takemura; Nobutaka Hanagata; Masami Niwa; Daisuke Watanabe

Background Silver nanoparticles (Ag-NPs) can enter the brain and induce neurotoxicity. However, the toxicity of Ag-NPs on the blood–brain barrier (BBB) and the underlying mechanism(s) of action on the BBB and the brain are not well understood. Method To investigate Ag-NP suspension (Ag-NPS)-induced toxicity, a triple coculture BBB model of rat brain microvascular endothelial cells, pericytes, and astrocytes was established. The BBB permeability and tight junction protein expression in response to Ag-NPS, NP-released Ag ions, and polystyrene-NP exposure were investigated. Ultrastructural changes of the microvascular endothelial cells, pericytes, and astrocytes were observed using transmission electron microscopy (TEM). Global gene expression of astrocytes was measured using a DNA microarray. Results A triple coculture BBB model of primary rat brain microvascular endothelial cells, pericytes, and astrocytes was established, with the transendothelial electrical resistance values >200 Ω·cm2. After Ag-NPS exposure for 24 hours, the BBB permeability was significantly increased and expression of the tight junction (TJ) protein ZO-1 was decreased. Discontinuous TJs were also observed between microvascular endothelial cells. After Ag-NPS exposure, severe mitochondrial shrinkage, vacuolations, endoplasmic reticulum expansion, and Ag-NPs were observed in astrocytes by TEM. Global gene expression analysis showed that three genes were upregulated and 20 genes were downregulated in astrocytes treated with Ag-NPS. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that the 23 genes were associated with metabolic processes, biosynthetic processes, response to stimuli, cell death, the MAPK pathway, and so on. No GO term and KEGG pathways were changed in the released-ion or polystyrene-NP groups. Ag-NPS inhibited the antioxidant defense of the astrocytes by increasing thioredoxin interacting protein, which inhibits the Trx system, and decreasing Nr4a1 and Dusp1. Meanwhile, Ag-NPS induced inflammation and apoptosis through modulation of the MAPK pathway or B-cell lymphoma-2 expression or mTOR activity in astrocytes. Conclusion These results draw our attention to the importance of Ag-NP-induced toxicity on the neurovascular unit and provide a better understanding of its toxicological mechanisms on astrocytes.


International Journal of Nanomedicine | 2012

Brain microvascular endothelial cell association and distribution of a 5 nm ceria engineered nanomaterial

Mo Dan; Michael T. Tseng; Peng Wu; Jason M. Unrine; Eric A. Grulke; Robert A. Yokel

Purpose: Ceria engineered nanomaterials (ENMs) have current commercial applications and both neuroprotective and toxic effects. Our hypothesis is that ceria ENMs can associate with brain capillary cells and/or cross the blood–brain barrier. Methods: An aqueous dispersion of ∼5 nm ceria ENM was synthesized and characterized in house. Its uptake space in the Sprague Dawley rat brain was determined using the in situ brain perfusion technique at 15 and 20 mL/minute flow rates; 30, 100, and 500 μg/mL ceria perfused for 120 seconds at 20 mL/minute; and 30 μg/mL perfused for 20, 60, and 120 seconds at 20 mL/minute. The capillary depletion method and light and electron microscopy were used to determine its capillary cell and brain parenchymal association and localization. Results: The vascular space was not significantly affected by brain perfusion flow rate or ENM, demonstrating that this ceria ENM did not influence blood–brain barrier integrity. Cerium concentrations, determined by inductively coupled plasma mass spectrometry, were significantly higher in the choroid plexus than in eight brain regions in the 100 and 500 μg/mL ceria perfusion groups. Ceria uptake into the eight brain regions was similar after 120-second perfusion of 30, 100, and 500 μg ceria/mL. Ceria uptake space significantly increased in the eight brain regions and choroid plexus after 60 versus 20 seconds, and it was similar after 60 and 120 seconds. The capillary depletion method showed 99.4% ± 1.1% of the ceria ENM associated with the capillary fraction. Electron microscopy showed the ceria ENM located on the endothelial cell luminal surface. Conclusion: Ceria ENM association with brain capillary endothelial cells saturated between 20 and 60 seconds and ceria ENM brain uptake was not diffusion-mediated. During the 120-second ceria ENM perfusion, ceria ENM predominately associated with the surface of the brain capillary cells, providing the opportunity for its cell uptake or redistribution back into circulating blood.


Nanotoxicology | 2014

Rat hippocampal responses up to 90 days after a single nanoceria dose extends a hierarchical oxidative stress model for nanoparticle toxicity

Sarita S. Hardas; Rukhsana Sultana; Warrier G; Mo Dan; Peng Wu; Eric A. Grulke; Michael T. Tseng; Jason M. Unrine; Uschi M. Graham; Robert A. Yokel; D.A. Butterfield

Abstract Ceria engineered nanomaterials (ENMs) have very promising commercial and therapeutic applications. Few reports address the effects of nanoceria in intact mammals, let alone long term exposure. This knowledge is essential to understand potential therapeutic applications of nanoceria in relation to its hazard assessment. The current study elucidates oxidative stress responses in the rat hippocampus 1 and 20 h, and 1, 7, 30 and 90 days following a single systemic infusion of 30 nm nanoceria. The results are incorporated into a previously described hierarchical oxidative stress (HOS) model. During the 1–20 h period, increases of the GSSG: GSH ratio and cytoprotective phase-II antioxidants were observed. During the 1–7 d period, cytoprotective phase-II antioxidants activities were inhibited with concomitant elevation of protein carbonyl (PC), 3-nitrotyrosine (3NT), heme oxygenase-1 (HO-1), cytokine IL-1β and the autophagy marker LC-3AB. At 30 day post ceria infusion, oxidative stress had its major impact. Phase-II enzyme activities were inhibited; concurrently PC, 3NT, HO-1 and Hsp70 levels were elevated along with augmentation of IL-1β, pro-apoptotic pro-caspase-3 and LC-3AB levels. This progress of escalating oxidative stress was reversed at 90 days when phase-II enzyme levels and activities were restored to normal levels, PC and 3NT levels were reduced to baseline, cytokine and pro-caspase-3 levels were suppressed, and cellular redox balance was restored in the rat hippocampus. This study demonstrates that a single administration of nanoceria induced oxidative stress that escalates to 30 days then terminates, in spite of the previously reported continued presence of nanoceria in peripheral organs. These results for the first time confirm in vivo the HOS model of response to ENM previously posited based on in vitro studies and extends this prior hierarchical oxidative stress model that described three tiers to a 4th tier, characterized by resolution of the oxidative stress and return to normal conditions.


PLOS ONE | 2013

Binding, transcytosis and biodistribution of anti-PECAM-1 iron oxide nanoparticles for brain-targeted delivery.

Mo Dan; David B. Cochran; Robert A. Yokel; Thomas D. Dziubla

Objective Characterize the flux of platelet-endothelial cell adhesion molecule (PECAM-1) antibody-coated superparamagnetic iron oxide nanoparticles (IONPs) across the blood-brain barrier (BBB) and its biodistribution in vitro and in vivo. Methods Anti-PECAM-1 IONPs and IgG IONPs were prepared and characterized in house. The binding affinity of these nanoparticles was investigated using human cortical microvascular endothelial cells (hCMEC/D3). Flux assays were performed using a hCMEC/D3 BBB model. To test their immunospecificity index and biodistribution, nanoparticles were given to Sprague Dawley rats by intra-carotid infusion. The capillary depletion method was used to elucidate their distribution between the BBB and brain parenchyma. Results Anti-PECAM-1 IONPs were ∼130 nm. The extent of nanoparticle antibody surface coverage was 63.6±8.4%. Only 6.39±1.22% of labeled antibody dissociated from IONPs in heparin-treated whole blood over 4 h. The binding affinity of PECAM-1 antibody (KD) was 32 nM with a maximal binding (Bmax) of 17×105 antibody molecules/cell. Anti-PECAM-1 IONP flux across a hCMEC/D3 monolayer was significantly higher than IgG IONPs with 31% of anti-PECAM-1 IONPs in the receiving chamber after 6 h. Anti-PECAM-1 IONPs showed higher concentrations in lung and brain, but not liver or spleen, than IgG IONPs after infusion. The capillary depletion method showed that 17±12% of the anti-PECAM-1 IONPs crossed the BBB into the brain ten minutes after infusion. Conclusions PECAM-1 antibody coating significantly increased IONP flux across the hCMEC/D3 monolayer. In vivo results showed that the PECAM-1 antibody enhanced BBB association and brain parenchymal accumulation of IONPs compared to IgG. This research demonstrates the benefit of anti-PECAM-1 IONPs for association and flux across the BBB into the brain in relation to its biodistribution in peripheral organs. The results provide insight into potential application and toxicity concerns of anti-PECAM-1 IONPs in the central nervous system.


PLOS ONE | 2017

Acute toxicity and genotoxicity of silver nanoparticle in rats

Hairuo Wen; Mo Dan; Ying Yang; Jianjun Lyu; Anliang Shao; Xiang Cheng; Liang Chen; Liming Xu

Objective The potential risk of a nanoparticle as a medical application has raised wide concerns, and this study aims to investigate silver nanoparticle (AgNP)-induced acute toxicities, genotoxicities, target organs and the underlying mechanisms. Methods Sprague-Dawley rats were randomly divided into 4 groups (n = 4 each group), and AgNP (containing Ag nanoparticles and released Ag+, 5 mg/kg), Ag+ (released from the same dose of AgNP, 0.0003 mg/kg), 5% sucrose solution (vechicle control) and cyclophophamide (positive control, 40 mg/kg) were administrated intravenously for 24 h respectively. Clinical signs and body weight of rats were recorded, and the tissues were subsequently collected for biochemical examination, Ag+ distribution detection, histopathological examination and genotoxicity assays. Results The rank of Ag detected in organs from highest to lowest is lung>spleen>liver>kidney>thymus>heart. Administration of AgNP induced a marked increase of ALT, BUN, TBil and Cre. Histopathological examination results showed that AgNP induced more extensive organ damages in liver, kidneys, thymus, and spleen. Bone marrow micronucleus assay found no statistical significance among groups (p > 0.05), but the number of aberration cells and multiple aberration cells were predominately increased from rats dosed with Ag+ and AgNP (p < 0.01), and more polyploidy cells were generated in the AgNP group (4.3%) compared with control. Conclusion Our results indicated that the AgNP accumulated in the immune system organs, and mild irritation was observed in the thymus and spleen of animals treated with AgNP, but not with Ag+. The liver and kidneys could be the most affected organs by an acute i.v. dose of AgNP, and significantly increased chromosome breakage and polyploidy cell rates also implied the potential genotoxicity of AgNP. However, particle-specific toxicities and potential carcinogenic effect remain to be further confirmed in a chronic toxicity study.

Collaboration


Dive into the Mo Dan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peng Wu

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Younsoo Bae

University of Kentucky

View shared research outputs
Researchain Logo
Decentralizing Knowledge