Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 422
Filter
1.
Toxicology ; 467: 153098, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35026344

ABSTRACT

Molybdenum, lithium, and tungsten are constituents of many products, and exposure to these elements potentially occurs at work. Therefore it is important to determine at what levels they are toxic, and thus we set out to review their pulmonary toxicity, genotoxicity, and carcinogenicity. After pulmonary exposure, molybdenum and tungsten are increased in multiple tissues; data on the distribution of lithium are limited. Excretion of all three elements is both via faeces and urine. Molybdenum trioxide exerted pulmonary toxicity in a 2-year inhalation study in rats and mice with a lowest-observed-adverse-effect concentration (LOAEC) of 6.6 mg Mo/m3. Lithium chloride had a LOAEC of 1.9 mg Li/m3 after subacute inhalation in rabbits. Tungsten oxide nanoparticles resulted in a no-observed-adverse-effect concentration (NOAEC) of 5 mg/m3 after inhalation in hamsters. In another study, tungsten blue oxide had a LOAEC of 63 mg W/m3 in rats. Concerning genotoxicity, for molybdenum, the in vivo genotoxicity after inhalation remains unknown; however, there was some evidence of carcinogenicity of molybdenum trioxide. The data on the genotoxicity of lithium are equivocal, and one carcinogenicity study was negative. Tungsten seems to have a genotoxic potential, but the data on carcinogenicity are equivocal. In conclusion, for all three elements, dose descriptors for inhalation toxicity were identified, and the potential for genotoxicity and carcinogenicity was assessed.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Lithium Chloride/toxicity , Lung/drug effects , Molybdenum/toxicity , Neoplasms/chemically induced , Oxides/toxicity , Tungsten/toxicity , Animals , Body Burden , Carcinogenicity Tests , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Dose-Response Relationship, Drug , Humans , Inhalation Exposure , Lithium Chloride/pharmacokinetics , Lung/metabolism , Lung/pathology , Metal Nanoparticles , Molybdenum/pharmacokinetics , Mutagenicity Tests , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oxides/pharmacokinetics , Risk Assessment , Tungsten/pharmacokinetics
2.
Naunyn Schmiedebergs Arch Pharmacol ; 394(10): 1991-2002, 2021 10.
Article in English | MEDLINE | ID: mdl-34415355

ABSTRACT

The current understanding of osteoarthritis is developing from a mechanical disease caused by cartilage wear to a complex biological response involving inflammation, oxidative stress and other aspects. Nanoparticles are widely used in drug delivery due to its good stability in vivo and cell uptake efficiency. In addition to the above advantages, metal/metal oxide NPs, such as cerium oxide and manganese dioxide, can also simulate the activity of antioxidant enzymes and catalyze the degradation of superoxide anions and hydrogen peroxide. Degrading of metal/metal oxide nanoparticles releases metal ions, which may slow down the progression of osteoarthritis by inhibiting inflammation, promoting cartilage repair and inhibiting cartilage ossification. In present review, we focused on recent research works concerning osteoarthritis treating with metal/metal oxide nanoparticles, and introduced some potential nanoparticles that may have therapeutic effects.


Subject(s)
Metal Nanoparticles/therapeutic use , Metals/therapeutic use , Osteoarthritis/drug therapy , Oxides/therapeutic use , Animals , Cartilage/metabolism , Humans , Metals/pharmacokinetics , Osteoarthritis/metabolism , Oxides/pharmacokinetics
3.
Toxicology ; 459: 152859, 2021 07.
Article in English | MEDLINE | ID: mdl-34273449

ABSTRACT

Vanadium dioxide nanoparticles (VO2 NPs) have been massively produced and widely applied due to their excellent metal-insulator transition property, making it extremely urgent to evaluate their safety, especially for low-dose long-term respiratory occupational exposure. Here, we report a comprehensive cytotoxicity and genotoxicity study on VO2 NPs to lung cell lines A549 and BEAS-2B following a long-term exposure. A commercial VO2 NP, S-VO2, was used to treat BEAS-2B (0.15-0.6 µg/mL) and A549 (0.3-1.2 µg/mL) cells for four exposure cycles, and each exposure cycle lasted for 4 consecutive days; then various bioassays were performed after each cycle. Significant proliferation inhibition was observed in both cell lines after long-term exposure of S-VO2 at low doses that did not cause apparent acute cytotoxicity; however, the genotoxicity of S-VO2, characterized by DNA damage and micronuclei, was only observed in A549 cells. These adverse effects of S-VO2 were exposure time-, dose- and cell-dependent, and closely related to the solubility of S-VO2. The oxidative stress in cells, i.e., enhanced reactive oxygen species (ROS) generation and suppressed reduced glutathione, was the main toxicity mechanism of S-VO2. The ROS-associated mitochondrial damage and DNA damage led to the genotoxicity, and cell proliferation retard, resulting in the cellular viability loss. Our results highlight the importance and urgent necessity of the investigation on the long-term toxicity of VO2 NPs.


Subject(s)
Cell Survival/drug effects , Lung/pathology , Metal Nanoparticles/toxicity , Mutagens/toxicity , Oxides/toxicity , Vanadium Compounds/toxicity , A549 Cells , Cell Line , Cell Proliferation/drug effects , DNA Damage , Glutathione/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Micronucleus Tests , Oxidative Stress , Oxides/pharmacokinetics , Reactive Oxygen Species/metabolism , Vanadium Compounds/pharmacokinetics
4.
Biomed Chromatogr ; 35(12): e5207, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34184288

ABSTRACT

Monocrotaline (MCT) is a pyrrolizidine alkaloid that can induce hepatic sinusoidal damage, pulmonary hypertension, renal toxicity, and heart disease. Monocrotaline N-oxide (MNO), the primary metabolite of MCT, is less toxic; however, it can convert back to MCT to exhibit its toxicity. This study developed and validated a rapid and sensitive LC-MS/MS method for the simultaneous determination of MCT and monocrotaline N-oxide in rat plasma. The method has a linearity over the concentration range of 1-2000 ng/mL with correlation coefficients (r) >0.997 for each analyte. The results of selectivity, matrix effect, accuracy and precision, and recovery were all within the acceptance criteria. The validated method has been successfully applied to study pharmacokinetic behaviors and bioavailability of MCT in rats. MCT was rapidly absorbed (Tmax : 0.400 ± 0.149 h) after oral administration, and the absolute bioavailability of MCT was 78.2%.


Subject(s)
Chromatography, Liquid/methods , Monocrotaline , Tandem Mass Spectrometry/methods , Administration, Oral , Animals , Biological Availability , Limit of Detection , Linear Models , Male , Monocrotaline/blood , Monocrotaline/pharmacokinetics , Oxides/blood , Oxides/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reproducibility of Results
5.
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
6.
Sci Rep ; 10(1): 18156, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097778

ABSTRACT

Glioblastoma (GBM) is a malignant brain tumour with a dismal prognosis, despite best treatment by surgical resection, radiation therapy (RT) and chemotherapy with temozolomide (TMZ). Nanoparticle (NP) therapy is an emerging consideration due to the ability of NPs to be formulated and cross the blood brain barrier. Lanthanum oxide (La2O3) NPs are therapeutically advantageous due to the unique chemical properties of lanthanum making it cytotoxic to cancers, and able to enhance existing anti-cancer treatments. However, La2O3 NPs have yet to be thoroughly investigated in brain tumors. We show that these NPs can reach the brain after venous injection, penetrate into GBM cells via endocytosis, dissociate to be cytotoxic, and enhance the therapeutic effects of RT and TMZ. The mechanisms of cell death by La2O3 NPs were found to be multifaceted. Increasing NP concentration was correlated to increased intrinsic and extrinsic apoptosis pathway markers in a radical oxygen species (ROS)-dependent manner, as well as involving direct DNA damage and autophagic pathways within GBM patient-derived cell lines. NP interactions to sensitize GBM to RT and TMZ were shown to involve these pathways by enhancing ROS and apoptotic mechanisms. We therefore demonstrate the therapeutic potential of La2O3 NPs to treat GBM cells in vitro, and encourage translational exploration in the future.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Lanthanum/administration & dosage , Metal Nanoparticles/administration & dosage , Oxides/administration & dosage , Temozolomide/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Brain/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chemoradiotherapy/methods , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Glioblastoma/pathology , Humans , Injections, Intravenous , Lanthanum/pharmacokinetics , Mice , Oxides/pharmacokinetics , Radiation Tolerance/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Temozolomide/therapeutic use , Tissue Distribution , Xenograft Model Antitumor Assays
7.
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
8.
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
9.
Int J Nanomedicine ; 15: 3843-3850, 2020.
Article in English | MEDLINE | ID: mdl-32581534

ABSTRACT

PURPOSE: Despite tremendous results achieved by immune checkpoint inhibitors, most patients are not responders, mainly because of the lack of a pre-existing anti-tumor immune response. Thus, solutions to efficiently prime this immune response are currently under intensive investigations. Radiotherapy elicits cancer cell death, generating an antitumor-specific T cell response, turning tumors in personalized in situ vaccines, with potentially systemic effects (abscopal effect). Nonetheless, clinical evidence of sustained anti-tumor immunity as abscopal effect are rare. METHODS: Hafnium oxide nanoparticles (NBTXR3) have been designed to increase energy dose deposit within cancer cells. We examined the effect of radiotherapy-activated NBTXR3 on anti-tumor immune response activation and abscopal effect production using a mouse colorectal cancer model. RESULTS: We demonstrate that radiotherapy-activated NBTXR3 kill more cancer cells than radiotherapy alone, significantly increase immune cell infiltrates both in treated and in untreated distant tumors, generating an abscopal effect dependent on CD8+ lymphocyte T cells. CONCLUSION: These data show that radiotherapy-activated NBTXR3 could increase local and distant tumor control through immune system priming. Our results may have important implications for immunotherapeutic agent combination with radiotherapy.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Hafnium/pharmacology , Oxides/pharmacology , Animals , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Biological Availability , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Female , Hafnium/chemistry , Hafnium/pharmacokinetics , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/radiotherapy , Oxides/chemistry , Oxides/pharmacokinetics
10.
Biomed Chromatogr ; 34(10): e4918, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32533577

ABSTRACT

From the point of view of drug efficacy and safety, pharmacokinetic profiles of both In this work, a sensitive and reliable liquid chromatographic-tandem mass spectrometric method was established for simultaneous determination of sutetinib and N-oxide metabolite (SNO) in human plasma and further applied to a pharmacokinetic study. Analytes were extracted from plasma samples (100 µl) via acetonitrile-induced protein precipitation and separated on a C18 column using ammonium acetate with ammonium hydroxide and acetonitrile as the mobile phase. Positive electrospray ionization was carried out through multiple reaction monitoring with transitions of m/z 440.2 → 367.1 and 446.2 → 367.1 for sutetinib and SNO, respectively. The method was linear within the concentration range of 0.5-100 ng/ml for both analytes. The precision, accuracy, selectivity, recovery and matrix effect of this method all met the requirements of bioanalytical guidance. In addition, a plasma stability assessment demonstrated unexpected results. Sutetinib was prone to form covalent conjugates with plasma albumin in vitro. The degree of covalent binding increased with increasing temperature, resulting in a significant decrease in its plasma concentrations. However, SNO could not easily bind with albumin owing to steric hindrance or electronegativity. Furthermore, sutetinib and SNO remained stable when blood and plasma samples were kept on wet ice. The validated method was successfully employed for the pharmacokinetic evaluation of sutetinib in patients with advanced malignant solid tumors.


Subject(s)
Amides/blood , Antineoplastic Agents/blood , Chromatography, Liquid/methods , Oxides/blood , Protein Kinase Inhibitors/blood , Amides/pharmacokinetics , Amides/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Humans , Limit of Detection , Linear Models , Neoplasms/drug therapy , Oxides/pharmacokinetics , Oxides/therapeutic use , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Reproducibility of Results , Tandem Mass Spectrometry/methods
11.
Toxicol Sci ; 174(2): 311-325, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32058562

ABSTRACT

Based on the wide use of cobalt substances in a range of important technologies, it has become important to predict the toxicological properties of new or lesser-studied substances as accurately as possible. We studied a group of 6 cobalt substances with inorganic ligands, which were tested for their bioaccessibility (surrogate measure of bioavailability) through in vitro bioelution in simulated gastric and intestinal fluids. Representatives of the group also underwent in vivo blood kinetics and mass balance tests, and both oral acute and repeated dose toxicity (RDT) testing. We were able to show a good correlation between high in vitro bioaccessibility with high in vivo bioavailability and subsequent high in vivo toxicity; consequently, low in vitro bioaccessibility correlated well with low in vivo bioavailability and low in vivo toxicity. In vitro bioelution in simulated gastric fluid was the most precise predictor of the difference in the oral RDT lowest observed adverse effect levels of 2 compounds representing the highly and poorly bioaccessible subset of substances. The 2 compounds cobalt dichloride hexahydrate and tricobalt tetraoxide differed by a factor of 440 in their in vitro bioaccessibility and by a factor of 310 in their RDT lowest observed adverse effect level. In summary, this set of studies shows that solubility, specifically in vitro bioelution in simulated gastric fluid, is a good, yet conservative, predictor of in vivo bioavailability and oral systemic toxicity of inorganic cobalt substances. Bioelution data are therefore an invaluable tool for grouping and read across of cobalt substances for hazard and risk assessment.


Subject(s)
Cobalt/toxicity , Oxides/toxicity , Administration, Oral , Animals , Biological Availability , Cobalt/administration & dosage , Cobalt/chemistry , Cobalt/pharmacokinetics , Female , Gastric Juice/chemistry , Injections, Intravenous , Intestinal Secretions/chemistry , Male , Oxides/administration & dosage , Oxides/chemistry , Oxides/pharmacokinetics , Rats, Sprague-Dawley , Risk Assessment , Solubility , Toxicokinetics
12.
J Ethnopharmacol ; 249: 112421, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31759111

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Pyrrolizidine alkaloids (PAs) are a group of phytotoxins widely present in about 3% of flowering plants. Many PA-containing herbal plants can cause liver injury. Our previous studies demonstrated that PA N-oxides are also hepatotoxic, with toxic potency much lower than the corresponding PAs, due to significant differences in their toxicokinetic fates. AIM OF STUDY: This study aimed to investigate the oral absorption of PAs and PA N-oxides for better understanding of their significant differences in toxicokinetics and toxic potency. MATERIALS AND METHODS: The oral absorption of PAs and PA N-oxides in rats and in rat in situ single pass intestine perfusion model was investigated. The intestinal permeability and absorption mechanisms of five pairs of PAs and PA N-oxides were evaluated by using Caco-2 monolayer model. RESULTS: The plasma concentrations of total PAs and PA N-oxides within 0-60 min were significantly lower in rats orally treated with a PA N-oxide-containing herbal alkaloid extract than with a PA-containing herbal alkaloid extract at the same dose, indicating that the absorption of PA N-oxides was lower than that of PAs. Using the rat in situ single pass intestine perfusion model, less cumulative amounts of retrorsine N-oxide in mesenteric blood were observed compared to that of retrorsine. In Caco-2 monolayer model, all five PAs showed absorption with Papp AtoB values [(1.43-16.26) × 10-6 cm/s] higher than those of corresponding N-oxides with Papp AtoB values lower than 1.35 × 10-6 cm/s. A further mechanistic study demonstrated that except for senecionine N-oxide, retrorsine N-oxide, and lycopsamine N-oxide, all PAs and PA N-oxides investigated were absorbed via passive diffusion. While, for these 3 PA N-oxides, in addition to passive diffusion as their primary transportation, efflux transporter-mediated active transportation was also involved but to a less extent with the efflux ratio of 2.31-3.41. Furthermore, a good correlation between lipophilicity and permeability of retronecine-type PAs and their N-oxides with absorption via passive diffusion was observed, demonstrating that PAs have a better oral absorbability than that of the corresponding PA N-oxides. CONCLUSION: We discovered that among many contributors, the lower intestinal absorption of PA N-oxides was the initiating contributor that caused differences in toxicokinetics and toxic potency between PAs and PA N-oxides.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Intestinal Absorption , Oxides/toxicity , Plant Extracts/toxicity , Pyrrolizidine Alkaloids/toxicity , Administration, Oral , Animals , Asteraceae/chemistry , Caco-2 Cells , Chemical and Drug Induced Liver Injury/blood , Disease Models, Animal , Humans , Intestinal Mucosa/metabolism , Male , Oxides/administration & dosage , Oxides/chemistry , Oxides/pharmacokinetics , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/pharmacokinetics , Plant Roots/chemistry , Pyrrolizidine Alkaloids/administration & dosage , Pyrrolizidine Alkaloids/chemistry , Pyrrolizidine Alkaloids/pharmacokinetics , Rats
13.
ACS Appl Mater Interfaces ; 11(44): 41009-41018, 2019 Nov 06.
Article in English | MEDLINE | ID: mdl-31599564

ABSTRACT

Inflammatory macrophage (Mφ)-mediated atherosclerosis is a leading cause of mortality and morbidity worldwide. Photothermal therapy (PTT) has been demonstrated as an efficient strategy in killing target cells, and its application in the treatment of inflammation in atherosclerosis is developing. However, the choice of nanomaterials, mechanisms, and side effects are seldom considered. In this study, semiconductor nanomaterials, that is, MoO2 nanoclusters, were synthesized and used for the first time in PTT for inflammatory Mφ-mediated atherosclerosis. Based on cell differential phagocytosis, the optimum amount of MoO2 and treatment time were selected to exert the maximum ablation effect on Mφ and minimal damage on endothelial cells without requiring additional target or selective groups. Moreover, MoO2-based PTT shows an excellent therapeutic effect on atherosclerosis by eliminating Mφ in animal models, with no significant side effects observed. This study explores a new method of nanotechnology and pharmaceutical development by using and optimizing cost-effective metal oxide nanostructures in the treatment of atherosclerosis and motivates further research on minimizing the side effects of related materials.


Subject(s)
Atherosclerosis/therapy , Infrared Rays , Macrophages/radiation effects , Phagocytosis/radiation effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Atherosclerosis/immunology , Atherosclerosis/pathology , Bone Marrow Cells/cytology , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Arteries/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Macrophages/cytology , Macrophages/immunology , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Metal Nanoparticles/toxicity , Mice , Mice, Knockout , Molybdenum/chemistry , Molybdenum/pharmacokinetics , Oxides/chemistry , Oxides/pharmacokinetics , Phototherapy , Semiconductors , Tissue Distribution
14.
Chemosphere ; 230: 24-28, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31102868

ABSTRACT

Lead (Pb) is an important pollutant and is released into the environment in many forms. Different lead compounds have a variety of solubilities and so may impact on lead bioavailability and toxicity when added to soil. In this experimental study, we investigated the bioavailability of Pb in soil spiked with 300, 900 and 1500 mg/kg of Pb-acetate, PbCl2 and PbO using lettuce and wallaby grass. The concentration of Pb in the shoots of both species from control soils (2-3 mg/kg) was similar to previously reported concentrations in plants grown on uncontaminated soils. The Pb concentrations in the plant shoots increased with Pb concentrations in soil for lettuce (R2 = 0.526, P < 0.001) and wallaby grass (R2 = 0.776, P < 0.001). This study demonstrated that Pb bioavailability in soil was not affected by the type of Pb compound added to the soil for both plant species up to 1500 mg/kg Pb concentrations. Instead, the Pb concentration in the plant was best predicted by the total concentration of lead in the soil, irrespective of the original lead compound added to the soil. This research suggests that the original Pb compounds that contaminated the soil are unlikely to be an important factor in assessing Pb bioavailability, and hence risk, in soils.


Subject(s)
Lactuca/chemistry , Lead/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Oxides/pharmacokinetics , Poaceae/chemistry , Soil Pollutants/pharmacokinetics , Biological Availability , Lead/analysis , Organometallic Compounds/analysis , Oxides/analysis , Soil/chemistry , Soil Pollutants/analysis
15.
J Biosci Bioeng ; 128(3): 344-354, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31014562

ABSTRACT

Benefiting from lower operational costs and energy requirements than do hydrometallurgical and pyrometallurgical processes in metal recovery, the bioleaching of LiCoO2 through the use of sulfur-oxidizing and iron-oxidizing bacteria has drawn increasing attention. However, the bioleaching mechanism of LiCoO2 has not been clearly elaborated. In the present study, the effects of the energy source of bacteria, such as Fe2+, pyrite and S0, and the products of bacterial oxidation, such as Fe3+ and sulfuric acid, on the chemical leaching of LiCoO2 were studied. The results indicated that lithium was dissolved by acid, and cobalt was released by the reduction of Fe2+ and acid dissolution. The recovery of Li+ and Co2+ could be significantly improved by pH adjustment. Finally, optimal recoveries of Li+ and Co2+ were observed in the pyrite group, reaching 91.4% and 94.2%, respectively. By using pyrite as the energy source, the role of bacteria in bioleaching of LiCoO2 was investigated. The results showed that bacteria could produce sulfuric acid by oxidizing pyrite to promote the mobilization of Li+ and Co2+. The recovery of lithium and cobalt could be increased to 100.0% and 99.3% by bacteria. Moreover, extracellular polymeric substances secreted by bacteria were found to be a factor for the improvement of Li+ and Co2+ recovery.


Subject(s)
Bacteria/metabolism , Cobalt/pharmacokinetics , Iron/metabolism , Metallurgy , Oxides/pharmacokinetics , Sulfur/metabolism , Acidithiobacillus/metabolism , Acidithiobacillus thiooxidans/metabolism , Bacillus/metabolism , Biodegradation, Environmental , Cobalt/chemistry , Electric Power Supplies , Equipment Reuse , Hydrogen-Ion Concentration , Lithium/pharmacokinetics , Metallurgy/methods , Oxidation-Reduction , Oxides/chemistry , Sulfides/metabolism , Sulfur/chemistry , Sulfuric Acids/metabolism , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/pharmacokinetics
16.
Ultrasound Med Biol ; 45(6): 1455-1465, 2019 06.
Article in English | MEDLINE | ID: mdl-30857759

ABSTRACT

Arginine-glycine-aspartate (RGD)-carrying microbubbles (MBs) have been utilized as a specific contrast agent for glycoprotein IIb/IIIa (αIIbß3 integrin)-expressing activated platelets in ultrasound molecular imaging. Recently, we found that surface modification with lactadherin provides the RGD motif on the surface of phosphatidylserine-containing clinically available MBs, Sonazoid. Here, we examined the potential of lactadherin-bearing Sonazoid MBs to be targeted MBs for glycoprotein IIb/IIIa using the custom-designed in vitro settings with recombinant αIIbß3 integrin, activated platelets or erythrocyte-rich human clots. By modification of the surface with lactadherin, a large number of Sonazoid MBs were attached to the αIIbß3 integrin-coated and platelet-immobilized plate. Additionally, the video intensity of clots after incubation with lactadherin-bearing Sonazoid MBs was significantly higher than that with unmodified Sonazoid MBs, implying the number of attached Sonazoid MBs was increased by the modification with lactadherin. Our results suggest that the lactadherin-bearing Sonazoid MBs have the potential to be thrombus-targeted MBs.


Subject(s)
Antigens, Surface/pharmacology , Contrast Media/pharmacokinetics , Ferric Compounds/pharmacokinetics , Image Enhancement/methods , Iron/pharmacokinetics , Microbubbles , Milk Proteins/pharmacology , Oxides/pharmacokinetics , Ultrasonography/methods , Female , Humans , Male , Molecular Imaging/methods , Reference Values
17.
J Biomed Mater Res B Appl Biomater ; 107(6): 2195-2201, 2019 08.
Article in English | MEDLINE | ID: mdl-30637978

ABSTRACT

To evaluate the properties of experimental mineral trioxide aggregate (MTA) resin-modified materials for root-end filling procedures, varying their compositions regarding the addition of hydroxiapatite (HA) or dicalcium phosphate dihydrate, with or without chlorhexidine digluconate. White MTA (Angelus, Londrina, Brazil) was used as a reference material. Degree of conversion (DC) was evaluated by Fourier transformed infrared (FTIr) spectroscopy (n = 5). Flowability (n = 3) and radiopacity (n = 3) were evaluated following ISO 6876:2001 methods. For splitting tensile strength analysis, cylindrical samples (n = 10) were subjected to compressive load using a universal testing machine (Instron Corporation, Norwood, MA). Water sorption and solubility tests were performed according to ISO 4049:2009 methods. Calcium ion release and pH analysis (n = 10) were evaluated using a pH meter (Orion, Watsonville, CA). Cytotoxicity (n = 8) of materials extracts was evaluated as cell viability percentage. Statistical analysis was performed using Kolmogorov-Smirnov for normal distribution and data was subjected to one-way ANOVA and Tukey test (α = 0.05). Addition of chlorhexidine digluconate reduced DC mean values for experimental materials (<50%). White MTA demonstrated lower flowability (5.3 mm) and higher radiopacity (9.8 mm Al), splitting tensile strength (9.1 MPa), solubility (8.2 µg/mm3 ), calcium ion release (~26.5 ppm), cytotoxicity (55.2%), and pH mean values (10.8), when compared to experimental materials. All groups demonstrated a decrease in calcium release (<85%) and pH (<13%). Formulation containing HA demonstrated similar pH values after 28 days when compared to white MTA. Evaluated experimental resin-modified MTA based materials without chlorhexidine digluconate showed satisfactory results for all physico-chemical properties tested and cytotoxicity. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 2195-2201, 2019.


Subject(s)
Aluminum Compounds , Calcium Compounds , Chlorhexidine/analogs & derivatives , Fibroblasts/metabolism , Materials Testing , Oxides , Root Canal Filling Materials , Silicates , Aluminum Compounds/chemistry , Aluminum Compounds/pharmacokinetics , Aluminum Compounds/pharmacology , Animals , Calcium Compounds/chemistry , Calcium Compounds/pharmacokinetics , Calcium Compounds/pharmacology , Cell Line , Chlorhexidine/chemistry , Chlorhexidine/pharmacokinetics , Chlorhexidine/pharmacology , Drug Combinations , Mice , Oxides/chemistry , Oxides/pharmacokinetics , Oxides/pharmacology , Root Canal Filling Materials/chemistry , Root Canal Filling Materials/pharmacokinetics , Root Canal Filling Materials/pharmacology , Silicates/chemistry , Silicates/pharmacokinetics , Silicates/pharmacology
18.
Environ Sci Pollut Res Int ; 26(20): 20092-20106, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30264340

ABSTRACT

The rapid industrialization and urbanization of intra- and peri-urban areas at the world scale are responsible for the degradation of the quality of edible crops, because of their contamination with airborne pollutants. Their consumption could lead to serious health risks. In this work, we aim to investigate the phytotoxicity induced by foliar transfer of atmospheric particles of industrial/urban origin. Leaves of cabbage plants (Brassica oleracea var. Prover) were contaminated with metal-rich particles (PbSO4 CuO and CdO) of micrometer size. A trichloroacetic acid (TCA) treatment was used to inhibit the synthesis of the epicuticular waxes in order to investigate their protective role against metallic particles toxicity. Besides the location of the particles on/in the leaves by microscopic techniques, photosynthetic activity measurements, genotoxicity assessment, and quantification of the gene expression have been studied for several durations of exposure (5, 10, and 15 days). The results show that the depletion of epicuticular waxes has a limited effect on the particle penetration in the leaf tissues. The stomatal openings appear to be the main pathway of particles entry inside the leaf tissues, as demonstrated by the overexpression of the BolC.CHLI1 gene. The effects of particles on the photosynthetic activity are limited, considering only the photosynthetic Fv/Fm parameter. The genotoxic effects were significant for the contaminated TCA-treated plants, especially after 10 days of exposure. Still, the cabbage plants are able to implement repair mechanisms quickly, and to thwart the physiological effects induced by the particles. Finally, the foliar contamination by metallic particles induces no serious damage to DNA, as observed by monitoring the BolC.OGG1 gene.


Subject(s)
Brassica/drug effects , Metals/pharmacokinetics , Metals/toxicity , Plant Leaves/metabolism , Waxes/metabolism , Brassica/physiology , Cadmium Compounds/pharmacokinetics , Cadmium Compounds/toxicity , Copper/pharmacokinetics , Copper/toxicity , Crops, Agricultural , Gene Expression Regulation, Plant/drug effects , Lead/pharmacokinetics , Lead/toxicity , Mutagenicity Tests/methods , Oxides/pharmacokinetics , Oxides/toxicity , Particulate Matter/toxicity , Photosynthesis/drug effects , Plant Leaves/drug effects , Time Factors , Trichloroacetic Acid/pharmacology
19.
ACS Appl Mater Interfaces ; 10(50): 43429-43438, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30480995

ABSTRACT

A hybrid nanosystem with impeccable cellular imaging and antioxidant functionality is demonstrated. The microwave irradiation-derived molybdenum trioxide nanoparticles (MoO3 NPs) were surface-functionalized with the cationic dye molecule, methylene blue (MB), which enables superior UV-visible absorbance and fluorescence emission wavelengths potential for bioimaging. The radical scavenging property of the pristine MoO3 NPs and MoO3-MB NPs were studied in vivo using Caenorhabditis elegans as the model system. Heat shock-induced oxidative stress in C. elegans was significantly resolved by the MoO3-MB NPs, in agreement with the in vitro radical scavenging study by electron paramagnetic resonance spectroscopy. Hybrid nanostructures of MoO3-MB demonstrate synergistic benefits in intracellular imaging with intrinsic biocompatibility and antioxidant behavior, which can facilitate application as advanced healthcare materials toward bioimaging and clinical therapeutics.


Subject(s)
Caenorhabditis elegans/metabolism , Methylene Blue , Molybdenum , Nanoparticles/chemistry , Oxides , Animals , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacokinetics , Free Radical Scavengers/pharmacology , Heat-Shock Response/drug effects , Methylene Blue/chemistry , Methylene Blue/pharmacokinetics , Methylene Blue/pharmacology , Molybdenum/chemistry , Molybdenum/pharmacokinetics , Molybdenum/pharmacology , Oxidative Stress/drug effects , Oxides/chemistry , Oxides/pharmacokinetics , Oxides/pharmacology
20.
Int J Nanomedicine ; 13: 5937-5952, 2018.
Article in English | MEDLINE | ID: mdl-30323584

ABSTRACT

BACKGROUND: The Traditional Chinese Medicine, arsenic trioxide (ATO, As2O3) could inhibit growth and induce apoptosis in a variety of solid tumor cells, but it is severely limited in the treatment of glioma due to its poor BBB penetration and nonspecifcity distribution in vivo. PURPOSE: The objective of this study was encapsulating ATO in the modified PAMAM den-drimers to solve the problem that the poor antitumor effect of ATO to glioma, which provide a novel angle for the study of glioma treatment. METHODS: The targeting drug carrier (RGDyC-mPEG-PAMAM) was synthesized based on Arg-Gly-Asp (RGDyC) and αvß3 integrin targeting ligand, and conjugated to PEGylated fifth generation polyamidoamine dendrimer (mPEG-PAMAM). It was characterized by nuclear magnetic resonance, fourier transform infrared spectra, Nano-particle size-zeta potential analyzer,etc. The in vitro release characteristics were studied by dialysis bag method. MTT assay was used to investigate the cytotoxicity of carriers and the antitumor effect of ATO formulation. In vitro blood-brain barrier (BBB) and C6 cell co-culture models were established to investigate the inhibitory effect of different ATO formulation after transporting across BBB. Pharmacokinetic and antitumor efficacy studies were investigated in an orthotopic murine model of C6 glioma. RESULTS: The prepared RGDyC-mPEG-PAMAM was characterized for spherical dendrites, comparable size (21.60±6.81 nm), and zeta potential (5.36±0.22 mV). In vitro release showed that more ATO was released from RGDyC-mPEG-PAMAM/ATO (79.5%) at pH 5.5 than that of pH 7.4, during 48 hours. The cytotoxicity of PEG-modified carriers was lower than that of the naked PAMAM on both human brain microvascular endothelial cells and C6 cells. In in vitro BBB model, modification of RGDyC heightened the cytotoxicity of ATO loaded on PAMAM, due to an increased uptake by C6 cells. The results of cell cycle and apoptosis analysis revealed that RGDyC-mPEG-PAMAM/ATO arrested the cell cycle in G2-M and exhibited threefold increase in percentage of apoptosis to that in the PEG-PAMAM/ATO group. Compared with ATO-sol group, both RGDyC-mPEG-PAMAM/ATO and mPEG-PAMAM/ATO groups prolonged the half-life time, increased area under the curve, and improved antitumor effect, significantly. While the tumor volume inhibitory of RGDyC-mPEG-PAMAM/ATO was 61.46±12.26%, it was approximately fourfold higher than the ATO-sol group, and twofold to the mPEG-PAMAM/ATO group. CONCLUSION: In this report, RGDyC-mPEG-PAMAM could enhance the antitumor of ATO to glioma, it provides a desirable strategy for targeted therapy of glioma.


Subject(s)
Arsenicals/therapeutic use , Dendrimers/chemistry , Drug Delivery Systems , Glioma/drug therapy , Oligopeptides/chemistry , Oxides/therapeutic use , Polyethylene Glycols/chemistry , Animals , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/administration & dosage , Arsenicals/pharmacokinetics , Arsenicals/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Drug Carriers/chemistry , Drug Liberation , Endocytosis/drug effects , Glioma/pathology , Humans , Male , Mice , Oxides/administration & dosage , Oxides/pharmacokinetics , Oxides/pharmacology , Rabbits , Rats , Static Electricity , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...