Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Food Chem Toxicol ; 161: 112822, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35063473

ABSTRACT

Manganese (Mn) is an essential trace element, but overexposure is associated with toxicity and neurological dysfunction. Accumulation of Mn can be observed in dopamine-rich regions of the brain in vivo and Mn-induced oxidative stress has been discussed extensively. Nevertheless, Mn-induced DNA damage, adverse effects of DNA repair, and possible resulting consequences for the neurite network are not yet characterized. For this, LUHMES cells were used, as they differentiate into dopaminergic-like neurons and form extensive neurite networks. Experiments were conducted to analyze Mn bioavailability and cytotoxicity of MnCl2, indicating a dose-dependent uptake and substantial cytotoxic effects. DNA damage, analyzed by means of 8-oxo-7,8-dihydro-2'-guanine (8oxodG) and single DNA strand break formation, showed significant dose- and time-dependent increase of DNA damage upon 48 h Mn exposure. Furthermore, the DNA damage response was increased which was assessed by analytical quantification of poly(ADP-ribosyl)ation (PARylation). Gene expression of the respective DNA repair genes was not significantly affected. Degradation of the neuronal network is significantly altered by 48 h Mn exposure. Altogether, this study contributes to the characterization of Mn-induced neurotoxicity, by analyzing the adverse effects of Mn on genome integrity in dopaminergic-like neurons and respective outcomes.


Subject(s)
Chlorides/toxicity , Neurons/drug effects , Biological Availability , Cell Line , Cell Survival/drug effects , Chlorides/pharmacokinetics , DNA Damage/drug effects , DNA Repair/drug effects , DNA Repair/physiology , Gene Expression Regulation/drug effects , Humans , Manganese Compounds/pharmacokinetics , Membrane Potential, Mitochondrial/drug effects , Trace Elements , Tubulin/genetics , Tubulin/metabolism
2.
Theranostics ; 11(15): 7439-7449, 2021.
Article in English | MEDLINE | ID: mdl-34158859

ABSTRACT

X-ray excited persistent luminescence (XEPL) imaging has attracted increasing attention in biomedical imaging due to elimination of autofluorescence, high signal-to-noise ratio and repeatable activation with high penetration. However, optical imaging still suffers from limited for high spatial resolution. Methods: Herein, we report Mn3+-rich manganese oxide (MnOx)-coated chromium-doped zinc gallogermanate (ZGGO) nanoparticles (Mn-ZGGOs). Enhanced XEPL and magnetic resonance (MR) imaging were investigated by the decomposition of MnOx shell in the environment of tumors. We also evaluated the tumor cell-killing mechanism by detection of reactive oxygen (ROS), lipid peroxidation and mitochondrial membrane potential changes in vitro. Furthermore, the in vivo biodistribution, imaging and therapy were studied by U87MG tumor-bearing mice. Results: In the tumor region, the MnOx shell is quickly decomposed to produce Mn3+ and oxygen (O2) to directly generate singlet oxygen (1O2). The resulting Mn2+ transforms endogenous H2O2 into highly toxic hydroxyl radical (·OH) via a Fenton-like reaction. The Mn2+ ions and ZGGOs also exhibit excellent T1-weighted magnetic resonance (MR) imaging and ultrasensitive XEPL imaging in tumors. Conclusion: Both the responsive dual-mode imaging and simultaneous self-supplied O2 for the production of 1O2 and oxygen-independent ·OH in tumors allow for more accurate diagnosis of deep tumors and more efficient inhibition of tumor growth without external activation energy.


Subject(s)
Hydroxyl Radical/metabolism , Luminescent Agents , Manganese Compounds , Nanoparticles , Neoplasms, Experimental , Optical Imaging , Oxides , Singlet Oxygen/metabolism , Animals , Cell Line, Tumor , Humans , Luminescent Agents/chemistry , Luminescent Agents/pharmacokinetics , Luminescent Agents/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Oxides/chemistry , Oxides/pharmacokinetics , Oxides/pharmacology , Xenograft Model Antitumor Assays
3.
Radiat Environ Biophys ; 59(4): 683-692, 2020 11.
Article in English | MEDLINE | ID: mdl-32989569

ABSTRACT

The experiment was performed in support of a Japanese initiative to investigate the biological effects of irradiation from residual neutron-activated radioactivity that resulted from the A-bombing. Radionuclide 56Mn (T1/2 = 2.58 h) is one of the main neutron-activated emitters during the first hours after neutron activation of soil dust particles. In our previous studies (2016-2017) related to irradiation of male Wistar rats after dispersion of 56MnO2 powder, the internal doses in rats were found to be very inhomogeneous: distribution of doses among different organs ranged from 1.3 Gy in small intestine to less than 0.0015 Gy in some of the other organs. Internal doses in the lungs ranged from 0.03 to 0.1 Gy. The essential pathological changes were found in lung tissue of rats despite a low level of irradiation. In the present study, the dosimetry investigations were extended: internal doses in experimental mice and rats were estimated for various activity levels of dispersed neutron-activated 56MnO2 powder. The following findings were noted: (a) internal radiation doses in mice were several times higher in comparison with rats under similar conditions of exposure to 56MnO2 powder. (b) When 2.74 × 108 Bq of 56MnO2 powder was dispersed over mice, doses of internal irradiation ranged from 0.81 to 4.5 Gy in the gastrointestinal tract (small intestine, stomach, large intestine), from 0.096 to 0.14 Gy in lungs, and doses in skin and eyes ranged from 0.29 to 0.42 Gy and from 0.12 to 0.16 Gy, respectively. Internal radiation doses in other organs of mice were much lower. (c) Internal radiation doses were significantly lower in organs of rats with the same activity of exposure to 56MnO2 powder (2.74 × 108 Bq): 0.09, 0.17, 0.29, and 0.025 Gy in stomach, small intestine, large intestine, and lungs, respectively. (d) Doses of internal irradiation in organs of rats and mice were two to four times higher when they were exposed to 8.0 × 108 Bq of 56MnO2 (in comparison with exposure to 2.74 × 108 Bq of 56MnO2). (e) Internal radiation doses in organs of mice were 7-14 times lower with the lowest 56MnO2 amount (8.0 × 107 Bq) in comparison with the highest amount, 8.0 × 108 Bq, of dispersed 56MnO2 powder. The data obtained will be used for interpretation of biological effects in experimental mice and rats that result from dispersion of various levels of neutron-activated 56MnO2 powder, which is the subject of separate studies.


Subject(s)
Manganese Compounds/pharmacokinetics , Oxides/pharmacokinetics , Radioisotopes/pharmacokinetics , Animals , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Radiation Dosage , Rats, Wistar , Tissue Distribution
4.
PLoS One ; 15(9): e0224414, 2020.
Article in English | MEDLINE | ID: mdl-32931488

ABSTRACT

PROCEDURES: To preliminary assess the relationship between Manganese Enhanced Magnetic Resonance Imaging (MEMRI) and the expression of calcium receptors in human prostate and breast cancer animal models. METHODS: NOD/SCID mice were inoculated with MDA-MB-231 breast cancer cells and prostate PC3 cancer cells to develop orthotopic or pseudometastatic cancer animal models. Mice were studied on a clinical 3T scanner by using a prototype birdcage coil before and after intravenous injection of MnCl2. Assessment of receptor's status was carried out after the MR images acquisition by immunohistochemistry on excised tumours. RESULTS: Manganese contrast enhancement in breast or prostate cancer animal models well correlated with CaSR expression (p<0.01), whereas TRPV6 expression levels appeared not relevant to the Mn uptake. CONCLUSION: Our preliminary results suggest that MEMRI appears an efficient tool to characterize human breast and prostate cancer animal models in the presence of different expression level of calcium receptors.


Subject(s)
Breast Neoplasms/diagnostic imaging , Chlorides/administration & dosage , Contrast Media/administration & dosage , Magnetic Resonance Imaging/methods , Manganese Compounds/administration & dosage , Prostatic Neoplasms/diagnostic imaging , Animals , Breast Neoplasms/pathology , Calcium/metabolism , Cell Line, Tumor , Chlorides/pharmacokinetics , Contrast Media/pharmacokinetics , Feasibility Studies , Female , Humans , Immunohistochemistry , Injections, Intravenous , Male , Manganese Compounds/pharmacokinetics , Mice , Pilot Projects , Prostatic Neoplasms/pathology , Receptors, Calcium-Sensing/metabolism , TRPV Cation Channels/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
5.
Theranostics ; 10(17): 7683-7696, 2020.
Article in English | MEDLINE | ID: mdl-32685013

ABSTRACT

Tumor hypoxia, acidosis, and excessive reactive oxygen species (ROS) were the main characteristics of the bladder tumor microenvironment (TME), and abnormal TME led to autophagy activation, which facilitated cancer cell proliferation. The therapeutic efficacy of autophagy inhibitors might also be impeded by abnormal TME. To address these issues, we proposed a new strategy that utilized manganese dioxide (MnO2) nanoparticles to optimize the abnormal TME and revitalize autophagy inhibitors, and both oxygenation and autophagy inhibition may sensitize the tumor cells to radiation therapy. Methods: By taking advantage of the strong affinity between negatively charged MnO2 and positively charged chloroquine (CQ), the nanoparticles were fabricated by integrating MnO2 and CQ in human serum albumin (HSA)-based nanoplatform (HSA-MnO2-CQ NPs). Results: HSA-MnO2-CQ NPs NPs efficiently generated O2 and increased pH in vitro after reaction with H+/H2O2 and then released the encapsulated CQ in a H+/H2O2 concentration-dependent manner. The NPs restored the autophagy-inhibiting activity of chloroquine in acidic conditions by increasing its intracellular uptake, and markedly blocked hypoxia-induced autophagic flux. In vivo studies showed the NPs improved pharmacokinetic behavior of chloroquine and effectively accumulated in tumor tissues. The NPs exhibited significantly decreased tumor hypoxia areas and increased tumor pH, and had remarkable autophagy inhibition efficacy on bladder tumors. Finally, a significant anti-tumor effect achieved by the enhanced autophagy inhibition and radiation sensitization. Conclusions: HSA-MnO2-CQ NPs synergistically regulated the abnormal TME and inhibited autophagic flux, and effectively sensitized radiation therapy to treat bladder cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Chemoradiotherapy/methods , Drug Carriers/chemistry , Radiation-Sensitizing Agents/administration & dosage , Urinary Bladder Neoplasms/therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Autophagy/drug effects , Autophagy/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Chloroquine/administration & dosage , Chloroquine/pharmacokinetics , Drug Synergism , Humans , Hydrogen-Ion Concentration/drug effects , Male , Manganese Compounds/administration & dosage , Manganese Compounds/pharmacokinetics , Mice , Nanoparticles/chemistry , Oxides/administration & dosage , Oxides/pharmacokinetics , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacokinetics , Reactive Oxygen Species/metabolism , Serum Albumin, Human/chemistry , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Nanoscale ; 12(5): 2966-2972, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-31971210

ABSTRACT

Lactate, the main contributor to the acidic tumor microenvironment, not only promotes the proliferation of tumor cells, but also closely relates to tumor invasion and metastasis. Here, a tumor targeting nanoplatform, designated as Me&Flu@MSN@MnO2-FA, was fabricated for effective tumor suppression and anti-metastasis by interfering with lactate metabolism of tumor cells. Metformin (Me) and fluvastatin sodium (Flu) were incorporated into MnO2-coated mesoporous silicon nanoparticles (MSNs), the synergism between Me and Flu can modulate the pyruvate metabolic pathway to produce more lactate, and concurrently inhibit lactate efflux to induce intracellular acidosis to kill tumor cells. As a result of the restricted lactate efflux, the extracellular lactate concentration is reduced, and the ability of the tumor cells to migrate is also weakened. This ingenious strategy based on Me&Flu@MSN@MnO2-FA showed an obvious inhibitory effect on tumor growth and resistance to metastasis.


Subject(s)
Fluvastatin , Lactates/metabolism , Manganese Compounds , Metformin , Nanoparticles , Neoplasms , Tumor Microenvironment/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Fluvastatin/chemistry , Fluvastatin/pharmacokinetics , Fluvastatin/pharmacology , Folic Acid/metabolism , Humans , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Metformin/chemistry , Metformin/pharmacokinetics , Metformin/pharmacology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Porosity , Silicon/chemistry , Silicon/pharmacokinetics , Silicon/pharmacology
7.
Mol Pharm ; 17(3): 837-851, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31977228

ABSTRACT

Delivery efficiencies of theranostic nanoparticles (NPs) based on passive tumor targeting strongly depend either on their blood circulation time or on appropriate modulations of the tumor microenvironment. Therefore, predicting the NP delivery efficiency before and after a tumor microenvironment modulation is highly desirable. Here, we present a new erythrocyte membrane-camouflaged magnetofluorescent nanocarrier (MMFn) with long blood circulation time (92 h) and high delivery efficiency (10% ID for Ehrlich murine tumor model). MMFns owe their magnetic and fluorescent properties to the incorporation of manganese ferrite nanoparticles (MnFe2O4 NPs) and IR-780 (a lipophilic indocyanine fluorescent dye), respectively, to their erythrocyte membrane-derived camouflage. MMFn composition, morphology, and size, as well as optical absorption, zeta potential, and fluorescent, magnetic, and magnetothermal properties, are thoroughly examined in vitro. We then present an analytical pharmacokinetic (PK) model capable of predicting the delivery efficiency (DE) and the time of peak tumor uptake (tmax), as well as changes in DE and tmax due to modulations of the tumor microenvironment, for potentially any nanocarrier. Experimental PK data sets (blood and tumor amounts of MMFns) are simultaneously fit to the model equations using the PK modeling software Monolix. We then validate our model analytical solutions with the numerical solutions provided by Monolix. We also demonstrate how our a priori nonmechanistic model for passive targeting relates to a previously reported mechanistic model for active targeting. All in vivo PK studies, as well as in vivo and ex vivo biodistribution studies, were conducted using two noninvasive techniques, namely, fluorescence molecular tomography (FMT) and alternating current biosusceptometry (ACB). Finally, histopathology corroborates our PK and biodistribution results.


Subject(s)
Drug Carriers/chemistry , Erythrocyte Membrane/chemistry , Ferric Compounds/chemistry , Fluorescent Dyes/chemistry , Magnetic Iron Oxide Nanoparticles/chemistry , Magnets/chemistry , Manganese Compounds/chemistry , Photothermal Therapy/methods , Animals , Carcinoma, Ehrlich Tumor/drug therapy , Disease Models, Animal , Drug Carriers/pharmacokinetics , Female , Ferric Compounds/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Hyperthermia, Induced/methods , Manganese Compounds/pharmacokinetics , Mice , Particle Size , Theranostic Nanomedicine/methods , Tissue Distribution , Tumor Burden/drug effects , Tumor Microenvironment/drug effects
8.
Invest Radiol ; 54(11): 697-703, 2019 11.
Article in English | MEDLINE | ID: mdl-31356382

ABSTRACT

OBJECTIVES: The goals of this study were to compare the efficacy of the new manganese-based magnetic resonance imaging (MRI) contrast agent Mn-PyC3A to the commercial gadolinium-based agents Gd-DOTA and to Gd-EOB-DTPA to detect tumors in murine models of breast cancer and metastatic liver disease, respectively, and to quantify the fractional excretion and elimination of Mn-PyC3A in rats. METHODS: T1-weighted contrast-enhanced MRI with 0.1 mmol/kg Mn-PyC3A was compared with 0.1 mmol/kg Gd-DOTA in a breast cancer mouse model (n = 8) and to 0.025 mmol/kg Gd-EOB-DTPA in a liver metastasis mouse model (n = 6). The fractional excretion, 1-day biodistribution, and 7-day biodistribution in rats after injection of 2.0 mmol/kg [Mn]Mn-PyC3A or Gd-DOTA were quantified by Mn gamma counting or Gd elemental analysis. Imaging data were compared with a paired t test; biodistribution data were compared with an unpaired t test. RESULTS: The postinjection-preinjection increases in tumor-to-muscle contrast-to-noise ratio (ΔCNR) 3 minutes after injection of Mn-PyC3A and Gd-DOTA (mean ± standard deviation) were 17 ± 3.8 and 20 ± 4.4, respectively (P = 0.34). Liver-to-tumor ΔCNR values at 8 minutes postinjection of Mn-PyC3A and Gd-EOB-DTPA were 28 ± 9.0 and 48 ± 23, respectively (P = 0.11). Mn-PyC3A is eliminated with 85% into the urine and 15% into the feces after administration to rats. The percentage of the injected doses (%ID) of Mn and Gd recovered in tissues after 1 day were 0.32 ± 0.12 and 0.57 ± 0.12, respectively (P = 0.0030), and after 7 days were 0.058 ± 0.051 and 0.19 ± 0.052, respectively (P < 0.0001). CONCLUSIONS: Mn-PyC3A provides comparable tumor contrast enhancement to Gd-DOTA in a mouse breast cancer model and is more completely eliminated than Gd-DOTA; partial hepatobiliary elimination of Mn-PyC3A enables conspicuous delayed phase visualization of liver metastases.


Subject(s)
Breast Neoplasms/diagnostic imaging , Contrast Media/pharmacokinetics , Diamines/pharmacokinetics , Image Enhancement/methods , Liver Neoplasms/diagnostic imaging , Magnetic Resonance Imaging/methods , Manganese Compounds/pharmacokinetics , Manganese/pharmacokinetics , Picolinic Acids/pharmacokinetics , Animals , Disease Models, Animal , Female , Gadolinium/administration & dosage , Gadolinium DTPA/pharmacokinetics , Heterocyclic Compounds/pharmacokinetics , Mice , Mice, Inbred BALB C , Organometallic Compounds/pharmacokinetics , Tissue Distribution
9.
Theranostics ; 9(6): 1764-1776, 2019.
Article in English | MEDLINE | ID: mdl-31037137

ABSTRACT

The development of a highly efficient, low-toxicity, ultrasmall ferrite nanoparticle-based T1 contrast agent for high-resolution magnetic resonance imaging (MRI) is highly desirable. However, the correlations between the chemical compositions, in vitro T1 relaxivities, in vivo nano-bio interactions and toxicities remain unclear, which has been a challenge in optimizing the in vivo T1 contrast efficacy. Methods: Ultrasmall (3 nm) manganese ferrite nanoparticles (MnxFe3-xO4) with different doping concentrations of the manganese ions (x = 0.32, 0.37, 0.75, 1, 1.23 and 1.57) were used as a model system to investigate the composition-dependence of the in vivo T1 contrast efficacy. The efficacy of liver-specific contrast-enhanced MRI was assessed through systematic multiple factor analysis, which included the in vitro T1 relaxivity, in vivo MRI contrast enhancement, pharmacokinetic profiles (blood half-life time, biodistribution) and biosafety evaluations (in vitro cytotoxicity testing, in vivo blood routine examination, in vivo blood biochemistry testing and H&E staining to examine the liver). Results: With increasing Mn doping, the T1 relaxivities initially increased to their highest value of 10.35 mM-1s-1, which was obtained for Mn0.75Fe2.25O4, and then the values decreased to 7.64 m M-1s-1, which was obtained for the Mn1.57Fe1.43O4 nanoparticles. Nearly linear increases in the in vivo MRI signals (ΔSNR) and biodistributions (accumulation in the liver) of the MnxFe3-xO4 nanoparticles were observed for increasing levels of Mn doping. However, both the in vitro and in vivo biosafety evaluations suggested that MnxFe3-xO4 nanoparticles with high Mn-doping levels (x > 1) can induce significant toxicity. Conclusion: The systematic multiple factor assessment indicated that the MnxFe3-xO4 (x = 0.75-1) nanoparticles were the optimal T1 contrast agents with higher in vivo efficacies for liver-specific MRI than those of the other compositions of the MnxFe3-xO4 nanoparticles. Our work provides insight into the optimization of ultrasmall ferrite nanoparticle-based T1 contrast agents by tuning their compositions and promotes the translation of these ultrasmall ferrite nanoparticles for clinical use of high-performance contrast-enhanced MRI.


Subject(s)
Contrast Media/chemistry , Contrast Media/pharmacology , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Magnetic Resonance Imaging/methods , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Animals , Contrast Media/adverse effects , Contrast Media/pharmacokinetics , Ferric Compounds/adverse effects , Ferric Compounds/pharmacokinetics , Liver/diagnostic imaging , Manganese Compounds/adverse effects , Manganese Compounds/pharmacokinetics , Mice, Inbred BALB C , Nanoparticles/adverse effects
10.
IEEE Trans Nanobioscience ; 18(3): 456-462, 2019 07.
Article in English | MEDLINE | ID: mdl-30998477

ABSTRACT

In this paper, the application of a technique to evaluate in vivo biodistribution of magnetic nanoparticles (MNP) is addressed: the Multichannel AC Biosusceptometry System (MC-ACB). It allows real-time assessment of magnetic nanoparticles in both bloodstream clearance and liver accumulation, where a complex network of inter-related cells is responsible for MNP uptake. Based on the acquired MC-ACB images, we propose a mathematical model which helps to understand the distribution and accumulation pharmacokinetics of MNP. The MC-ACB showed a high time resolution to detect and monitor MNP, providing sequential images over the particle biodistribution. Utilizing the MC-ACB instrument, we assessed regions corresponding to the heart and liver, and we determined the MNP transfer rates between the bloodstream and the liver. The pharmacokinetic model resulted in having a strong correlation with the experimental data, suggesting that the MC-ACB is a valuable and accessible imaging device to assess in vivo and real-time pharmacokinetic features of MNP.


Subject(s)
Diagnostic Imaging , Image Processing, Computer-Assisted/methods , Magnetite Nanoparticles , Signal Processing, Computer-Assisted , Animals , Diagnostic Imaging/instrumentation , Diagnostic Imaging/methods , Equipment Design , Ferric Compounds/pharmacokinetics , Male , Manganese Compounds/pharmacokinetics , Particle Size , Rats , Rats, Wistar , Tissue Distribution
11.
ACS Appl Mater Interfaces ; 10(48): 41107-41117, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30403475

ABSTRACT

Nanodrug-delivery systems modified with targeting molecules allow antitumor drugs to localize to tumor sites efficiently. CD147 protein is expressed highly on hepatoma cells. Firstly, we synthesized magnetothermally responsive nanocarriers/doxorubicin (MTRN/DOX) which was composed of manganese zinc (Mn-Zn) ferrite magnetic nanoparticles, amphiphilic and thermosensitivity copolymer drug carriers together with DOX. Then CD147-MTRN/DOX was formed with MTRN/DOX and monoclonal antibody that specifically binds to CD147 protein. It could target hepatoma cells actively and improve the DOX concentration in the tumor sites. Subsequently, an external alternating magnetic field elevated the temperature of the thermomagnetic particles, resulting in structural changes in the thermosensitive copolymer drug carriers, thereby releasing DOX. Hence, CD147-MTRN/DOX could enhance the responsiveness of hepatoma cells to the pre-existing chemotherapy drugs owing to active targeting combined synergistically with thermotherapy and chemotherapy, which has more significant anticancer effects than MTRN/DOX.


Subject(s)
Carcinoma, Hepatocellular , Doxorubicin , Drug Delivery Systems/methods , Hyperthermia, Induced , Liver Neoplasms , Magnetic Fields , Nanoparticles , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Xenograft Model Antitumor Assays , Zinc Compounds/chemistry , Zinc Compounds/pharmacokinetics , Zinc Compounds/pharmacology
12.
ACS Appl Mater Interfaces ; 10(41): 34954-34964, 2018 Oct 17.
Article in English | MEDLINE | ID: mdl-30234287

ABSTRACT

A Mn3O4 nanoparticle (NP)-based dual-modality probe has been developed for tumor positron emission tomography (PET)/magnetic resonance (MR) imaging. The dual-modality imaging probe was constructed by modifying multifunctional polyethyleneimine (PEI)-coated Mn3O4 NPs with folic acid (FA), followed with the radiolabeling with 64Cu. The formed imaging probe was utilized for PET/MR imaging of human cervical cancer mouse xenografts, which overexpress folate receptor (FR). The PEI-coated Mn3O4 NPs were synthesized using a solvothermal approach via decomposition of acetylacetone manganese. Multifunctional groups, including fluorescein isothiocyanate (FI), PEGylated FA, and NOTA chelator, were then sequentially loaded onto the surface of the amine groups of the Mn3O4 NPs. The remaining PEI amines were neutralized by the acetylation reaction. The resulting NOTA-FA-FI-PEG-PEI-Ac-Mn3O4 NPs were fully characterized and evaluated in vitro and successfully radiolabeled with 64Cu for tumor PET/MR imaging in small animals. In vivo blocking experiments were performed to determine the FR binding specificity of NPs. PET imaging results demonstrated that 64Cu-labeled Mn3O4 NPs display good tracer uptake in the FR-expressing HeLa tumors (tumor-to-muscle (T/M) ratio: 5.35 ± 0.31 at 18 h postinjection (pi)) and substantially reduced tracer uptake in the FR-blocked HeLa tumors (T/M ratio: 2.78 ± 0.68 at 18 h pi). The ex vivo data, including PET imaging and biodistribution, further confirmed the tumor binding specificity of the 64Cu-labeled Mn3O4 NPs. Moreover, the FR-targeted Mn3O4 NPs exhibited efficient T1-weighted MR imaging (MRI), leading to the precise tumor MRI at 18 h pi. PET/MR imaging with the 64Cu-NOTA-FA-FI-PEG-PEI-Ac-Mn3O4 NPs may offer a new quantitative approach to precisely measure the FR in tumors. The strategy of incorporating PEI nanotechnology into the construction of new biomaterials may be applied for the construction of novel nanoplatforms for cancer diagnosis and therapy.


Subject(s)
Coated Materials, Biocompatible , Drug Delivery Systems , Magnetic Resonance Imaging , Manganese Compounds , Neoplasms, Experimental , Oxides , Positron-Emission Tomography , Animals , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/pharmacology , Female , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/chemistry , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescein-5-isothiocyanate/pharmacology , Folic Acid/chemistry , Folic Acid/pharmacokinetics , Folic Acid/pharmacology , HeLa Cells , Humans , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Mice, Nude , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oxides/chemistry , Oxides/pharmacokinetics , Oxides/pharmacology , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacokinetics , Polyethyleneimine/pharmacology
13.
J Inorg Biochem ; 186: 257-263, 2018 09.
Article in English | MEDLINE | ID: mdl-29990749

ABSTRACT

With the advent of positron emission tomography/magnetic resonance imaging (PET/MRI) scanner, PET/MRI dual-modal imaging will play more and more important role in the diagnosis of cancers and other diseases. Until now, there is no an approved PET/MRI dual-modal imaging probe. The goal of this work is to design and synthesize potential PET/MRI dual-modal imaging probe based on superparamagnetic manganese ferrite nanoparticles. We have developed superparamagnetic nanoparticles that have uniform size with 5 nm and can be further functionalized through surface coating with dopamine and polyethylene glycol derivatives, which provide functional groups for conjugating tumor-targeting biomolecules and bifunctional chelators. The nanoparticles conjugated with integrin αvß3 over-expressed targeting cyclic arginine-glycine-aspartic acid (RGD)-peptide and labeled with positron radionuclide copper-64 were intravenously injected into glioblastoma xenograft nude mice. In vivo MRI and PET imaging of mice implied that the PET/MRI dual-modal imaging probe can precisely locate the tumor site with αvß3 over expression.


Subject(s)
Copper Radioisotopes , Drug Delivery Systems , Ferric Compounds , Glioblastoma/diagnostic imaging , Integrin alphaVbeta3/metabolism , Magnetic Resonance Imaging , Manganese Compounds , Nanoparticles , Positron-Emission Tomography , Radiopharmaceuticals , Animals , Copper Radioisotopes/chemistry , Copper Radioisotopes/pharmacokinetics , Copper Radioisotopes/pharmacology , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Heterografts , Humans , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasm Transplantation , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology
14.
J Biomed Mater Res B Appl Biomater ; 106(7): 2544-2552, 2018 10.
Article in English | MEDLINE | ID: mdl-29345749

ABSTRACT

Photodynamic therapy (PDT) holds promise in biochemical study and tumor treatment. A novel multifunctional nanosystem CaO2 /MnO2 @polydopamine (PDA)-methylene blue (MB) nanosheet (CMP-MB) was designed. CaO2 nanoparticles were encapsulated by MnO2 nanosheet, and then PDA was coated on the surface of CaO2 /MnO2 nanosheets, which could adsorb photosensitizer MB through hydrophobic interaction or π-π stacking. In this nanosystem, CaO2 /MnO2 had the ability of self-production of oxygen, which solved the problem of tumor hypoxia largely. Moreover, it is worth mentioning that the fluorescence of MB was suppressed by MnO2 , while its emission was triggered in the simulated tumor microenvironment. Therefore, CMP-MB nanosheet could be used to switch-control cell imaging potentially. 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide testing and Live/Dead assay confirmed CMP-MB nanosheet had fewer side effects without illumination while it destroyed Hela cell with the illumination of light. Vitro cell experiment demonstrated CMP-MB nanosheet could achieve tumor microenvironment responsive imaging and inhibit tumor cell growth under illumination effectively. Therefore, the system has great potential for PDT application and switch-control tumor cell imaging. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 2544-2552, 2018.


Subject(s)
Calcium Compounds , Indoles , Manganese Compounds , Methylene Blue , Nanostructures , Neoplasms , Oxides , Photochemotherapy , Polymers , Calcium Compounds/chemistry , Calcium Compounds/pharmacokinetics , Calcium Compounds/pharmacology , HeLa Cells , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Methylene Blue/chemistry , Methylene Blue/pharmacokinetics , Methylene Blue/pharmacology , Nanostructures/chemistry , Nanostructures/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Oxides/chemistry , Oxides/pharmacokinetics , Oxides/pharmacology , Oxygen/chemistry , Oxygen/metabolism , Polymers/chemistry , Polymers/pharmacokinetics , Polymers/pharmacology , Tumor Microenvironment/drug effects
15.
Nanotechnology ; 29(1): 015102, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29205171

ABSTRACT

Tagging recognition group(s) on superparamagnetic iron oxide is known to aid localisation (imaging), stimulation and separation of biological entities using magnetic resonance imaging (MRI) and magnetic agitation/separation (MAS) techniques. Despite the wide applicability of iron oxide nanoparticles in T 2-weighted MRI and MAS, the quality of the images and safe manipulation of the exceptionally delicate neural cells in a live brain are currently the key challenges. Here, we demonstrate the engineered manganese oxide clusters-iron oxide core-shell nanoparticle as an MR dual-modal contrast agent for neural stem cells (NSCs) imaging and magnetic manipulation in live rodents. As a result, using this engineered nanoparticle and associated technologies, identification, stimulation and transportation of labelled potentially multipotent NSCs from a specific location of a live brain to another by magnetic means for self-healing therapy can therefore be made possible.


Subject(s)
Cell Tracking/methods , Ependyma/diagnostic imaging , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/ultrastructure , Animals , Cell Survival , Contrast Media/administration & dosage , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Ependyma/cytology , Ependyma/metabolism , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Magnetite Nanoparticles/chemistry , Male , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oxides/chemistry , Oxides/pharmacokinetics , Rats , Rats, Sprague-Dawley
16.
Chemosphere ; 186: 374-380, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28802129

ABSTRACT

Several efficient stabilizing amendments have been recently proposed for the remediation of metal(loid)-contaminated soils. However, information on their interactions with plants, which is a crucial factor in soil environments, are still scarce. An amorphous manganese oxide (AMO) synthesized from organic compounds and nano zerovalent iron (nZVI) have been previously tested as promising stabilizing agents usable both for the stabilization of metals and As. Experiments with rhizoboxes were performed in order to evaluate their influence on the mobility of metal(loid)s in the bulk soil and rhizosphere of sunflower (Helianthus annuus L.) together with their impact on metal uptake and biomass yield. Generally, AMO proved more efficient than nZVI in all stages of experiment. Furthermore, the AMO effectively reduced water- and 0.01 M CaCl2-extractable fractions of Cd, Pb and Zn. The decreased bioavailability of contaminating metal(loid)s resulted in significant increase of microbial activity in AMO-amended soil. Together with metal(loid) extractability, the AMO was also able to significantly reduce the uptake of metals and ameliorate plant growth, especially in the case of Zn, since this metal was taken up in excessive amounts from the control soil causing strong phytotoxicity and even death of young seedlings. On the other hand, AMO application lead to significant release of Mn that was readily taken up by plants. Resulting Mn concentrations in biomass exceeded toxicity thresholds while plants were showing emergent Mn phytotoxicity symptoms. We highlight the need of such complex studies involving plants and soil biota when evaluating the efficiency of stabilizing amendments in contaminated soils.


Subject(s)
Environmental Restoration and Remediation/methods , Helianthus , Manganese Compounds/pharmacology , Oxides/pharmacology , Soil Pollutants/analysis , Bacteria/drug effects , Bacteria/growth & development , Biomass , Helianthus/growth & development , Helianthus/metabolism , Iron/chemistry , Manganese Compounds/chemical synthesis , Manganese Compounds/pharmacokinetics , Metals, Heavy/analysis , Metals, Heavy/pharmacokinetics , Metals, Heavy/toxicity , Oxides/chemical synthesis , Oxides/pharmacokinetics , Soil , Soil Pollutants/pharmacokinetics , Soil Pollutants/toxicity
17.
Int J Toxicol ; 36(3): 239-251, 2017.
Article in English | MEDLINE | ID: mdl-28460583

ABSTRACT

Manganese (Mn) is neurotoxic and can induce manganism, a Parkinson-like disease categorized as being a serious central nervous system irreversible neurodegenerative disease. An increased risk of developing symptoms of Parkinson disease has been linked to work-related exposure, for example, for workers in agriculture, horticulture, and people living near areas with frequent use of Mn-containing pesticides. In this study, the focus was placed on neurochemical effects of Mn. Rats were dosed intraperitoneally with 0.9% NaCl (control), 1.22 mg Mn (as MnO2)/kg bodyweight (bw)/day, or 2.5 mg Mn (as MnCl2)/kg bw/day for 7 d/wk for 8 or 12 weeks. This dosing regimen adds relevant new knowledge about Mn neurotoxicity as a consequence of low-dose subchronic Mn dosing. Manganese concentrations increased in the striatum, the rest of the brain, and in plasma, and regional brain neurotransmitter concentrations, including noradrenaline, dopamine (DA), 5-hydroxytrytamine, glutamate, taurine, and γ-amino butyric acid, and the activity of acetylcholinesterase changed. Importantly, a target parameter for Parkinson disease and manganism, the striatal DA concentration, was reduced after 12 weeks of dosing with MnCl2. Plasma prolactin concentration was not significantly affected due to a potentially reduced dopaminergic inhibition of the prolactin release from the anterior hypophysis. No effects on the striatal α-synuclein and synaptophysin protein levels were detected.


Subject(s)
Brain Chemistry/drug effects , Brain/drug effects , Chlorides/toxicity , Oxides/toxicity , Acetylcholinesterase/metabolism , Animals , Brain/metabolism , Chlorides/blood , Chlorides/pharmacokinetics , Dopamine/metabolism , Glutamic Acid/metabolism , Injections, Intraperitoneal , Male , Manganese/blood , Manganese/metabolism , Manganese Compounds/blood , Manganese Compounds/pharmacokinetics , Norepinephrine/metabolism , Oxides/blood , Oxides/pharmacokinetics , Rats, Sprague-Dawley , Serotonin/metabolism , Taurine/metabolism , gamma-Aminobutyric Acid/metabolism
18.
Neuroimage ; 156: 146-154, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28506873

ABSTRACT

There has been a growing interest in the use of manganese-enhanced MRI (MEMRI) for neuronal tract tracing in mammals, especially in rodents. For this MEMRI application, manganese solutions are usually directly injected into specific brain regions. Recently it was reported that manganese ions can diffuse through intact rat skull. Here the local manganese concentrations in the brain tissue after transcranial manganese application were quantified and the effectiveness of tracing from the area under the skull where delivery occurred was determined. It was established that transcranially applied manganese yields brain tissue enhancement dependent on the location of application on the skull and that manganese that enters the brain transcranially can trace to deeper brain areas.


Subject(s)
Chlorides/administration & dosage , Chlorides/pharmacokinetics , Magnetic Resonance Imaging/methods , Manganese Compounds/administration & dosage , Manganese Compounds/pharmacokinetics , Neuronal Tract-Tracers/administration & dosage , Neuronal Tract-Tracers/pharmacokinetics , Animals , Brain/diagnostic imaging , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Diffusion , Image Enhancement , Image Processing, Computer-Assisted/methods , Male , Rats , Rats, Sprague-Dawley , Skull , Tissue Distribution
19.
Mater Sci Eng C Mater Biol Appl ; 71: 504-511, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27987737

ABSTRACT

Magnetic nanoparticles (MNPs) are the major class of nanoparticles (NPs) with specific functional properties that make them good candidates for biomedical applications. Due to their response to the magnetic field, they can be used in targeted drug delivery systems. In current research, the MNPs were synthesized with the general formula of Fe1-xMnxFe2O4 by the co-precipitation technique. First, the effect of the Fe2+ ions in the system was investigated. Succinic anhydride was used as the first stabilizer to prepare surface for binding two types of polymer, including Polyethylene glycol (PEG) and palmitoylated Polyethylene glycol-grafted Chitosan (Cs-PEG-PA) were introduced as a polymeric shell. The composition, size, structure and magnetic properties of NPs were determined by the particle size analysis (PSA), X-ray diffractometry (XRD), Fourier transform infrared spectroscopy (FTIR) and vibrating sample magnetometer (VSM). Determining the well-defined properties of MNPs, methotrexate (MTX), as a common anticancer drug, was encapsulated into the coated MNPs. The drug encapsulation efficiency was as high as 92.8% with the magnetization value of 19.7emu/g. The in-vitro release pattern was studied, showing only 6% of the drug release in pH=7.4 (as a model of the physiological environment) and 25% in pH=5.4 (as a model of the tumor tissue environment) after 72h. Based on these results, we may be able to introduce this specific system as a novel pH sensitive MNP system for MTX targeting to tumor tissues in cancer chemotherapy.


Subject(s)
Chitosan , Coated Materials, Biocompatible , Ferric Compounds , Manganese Compounds , Methotrexate , Nanoparticles/chemistry , Polyethylene Glycols , Chitosan/chemistry , Chitosan/pharmacokinetics , Chitosan/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Humans , MCF-7 Cells , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Methotrexate/chemistry , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology
20.
Mater Sci Eng C Mater Biol Appl ; 71: 620-628, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27987753

ABSTRACT

Pharmaceutical industries spend more money in developing new and efficient methods for delivering successful drugs for anticancer therapy. Electrospun nanofibers and nanoparticles loaded with drugs have versatile biomedical applications ranging from wound healing to anticancer therapy. We aimed to attempt for fabricating elastomeric poly (l-lactic acid-co-ε-caprolactone) (PLACL) with Aloe Vera (AV), magnesium oxide (MgO) nanoparticles, curcumin (CUR) and ß-cyclodextrin (ß-CD) composite nanofibers to control the growth of MCF-7 cells for breast cancer therapy. The study focused on the interaction of MgO nanoparticle with CUR and ß-CD inhibiting the proliferation of Michigan Cancer Foundation-7 (MCF-7) breast cancer cells. FESEM micrographs of fabricated electrospun PLACL, PLACL/AV, PLACL/AV/MgO, PLACL/AV/MgO/CUR and PLACL/AV/MgO/ß-CD nanofibrous scaffolds achieved bead free, random and uniform nanofibers with fiber diameter in the range of 786±286, 507±171, 334±95, 360±94 and 326±80nm respectively. Proliferation of MCF-7 cells was decreased by 65.92% in PLACL/AV/MgO/CUR with respect to PLACL/AV/MgO nanofibrous scaffolds on day 9. The obtained results proved that 1% CUR interacting with MgO nanoparticles showed higher inhibition of MCF-7 cells among all other nanofibrous scaffolds thus serving as a promising biocomposite material system for the breast cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic , Breast Neoplasms/drug therapy , Curcumin , Manganese Compounds , Nanofibers/chemistry , Nanoparticles/chemistry , Oxides , Polyesters , beta-Cyclodextrins , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Curcumin/chemistry , Curcumin/pharmacokinetics , Curcumin/pharmacology , Female , Humans , MCF-7 Cells , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Oxides/chemistry , Oxides/pharmacokinetics , Oxides/pharmacology , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/pharmacology , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacokinetics , beta-Cyclodextrins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...