Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 865
Filter
Add more filters

Publication year range
1.
Environ Res ; 252(Pt 1): 118396, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38331143

ABSTRACT

The recombination of photogenerated carrier leads to inefficient Fe2+ regeneration, which limits the extensive application of heterogeneous photo-Fenton. Here, a novel Fe@Fe2O3/BiOBr catalyst with Z-scheme heterojunction structure is designed, and the establishment of the Z-scheme heterojunction facilitates the separation and transfer of photogenerated carrier and maintains the superior redox capability of the system. As-prepared Fe@Fe2O3/BiOBr catalyst exhibits outstanding catalytic performance and stability, especially for the optimum composite FFB-3, its degradation efficiency of tetracycline (TC) achieves 98.22% and the mineralization degree reaches 59.48% within 90 min under natural pH. The preeminent catalytic efficiency benefited from the synergistic of heterogeneous photo-Fenton and Z-scheme carriers transfer mechanism, where Fe2+ regeneration was achieved by photogenerated electrons, and increased hydroxyl radicals were produced with the participation of H2O2 in-situ generated. The results of free-radical scavenging experiment and ESR illustrated that •OH, •O2-, 1O2 and h+ were active species participating in TC degradation. Furthermore, the TC degradation paths were proposed according to LC-MS, and the toxicity evaluation result showed that the toxicity of TC solutions was markedly decreased after degradation. This study provides an innovative strategy for heterogeneous photo-Fenton degradation of antibiotic contaminations by constructing Z-scheme heterojunctions.


Subject(s)
Bismuth , Hydrogen Peroxide , Tetracycline , Tetracycline/chemistry , Tetracycline/toxicity , Hydrogen Peroxide/chemistry , Bismuth/chemistry , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/toxicity , Iron/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Ferric Compounds/chemistry , Ferric Compounds/toxicity , Animals , Catalysis
2.
Blood ; 137(18): 2532-2543, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33512448

ABSTRACT

Mechanisms thought to regulate activated factor VIII (FVIIIa) cofactor function include A2-domain dissociation and activated protein C (APC) cleavage. Unlike A2-domain dissociation, there is no known phenotype associated with altered APC cleavage of FVIII, and biochemical studies have suggested APC plays a marginal role in FVIIIa regulation. However, the in vivo contribution of FVIIIa inactivation by APC is unexplored. Here we compared wild-type B-domainless FVIII (FVIII-WT) recombinant protein with an APC-resistant FVIII variant (FVIII-R336Q/R562Q; FVIII-QQ). FVIII-QQ demonstrated expected APC resistance without other changes in procoagulant function or A2-domain dissociation. In plasma-based studies, FVIII-WT/FVIIIa-WT demonstrated dose-dependent sensitivity to APC with or without protein S, whereas FVIII-QQ/FVIIIa-QQ did not. Importantly, FVIII-QQ demonstrated approximately fivefold increased procoagulant function relative to FVIII-WT in the tail clip and ferric chloride injury models in hemophilia A (HA) mice. To minimize the contribution of FV inactivation by APC in vivo, a tail clip assay was performed in homozygous HA/FV Leiden (FVL) mice infused with FVIII-QQ or FVIII-WT in the presence or absence of monoclonal antibody 1609, an antibody that blocks murine PC/APC hemostatic function. FVIII-QQ again demonstrated enhanced hemostatic function in HA/FVL mice; however, FVIII-QQ and FVIII-WT performed analogously in the presence of the PC/APC inhibitory antibody, indicating the increased hemostatic effect of FVIII-QQ was APC specific. Our data demonstrate APC contributes to the in vivo regulation of FVIIIa, which has the potential to be exploited to develop novel HA therapeutics.


Subject(s)
Factor VIII/metabolism , Hemophilia A/pathology , Hemostasis , Protein C/metabolism , Recombinant Proteins/metabolism , Animals , Chlorides/toxicity , Factor VIII/genetics , Female , Ferric Compounds/toxicity , Hemophilia A/chemically induced , Hemophilia A/metabolism , Male , Mice , Mice, Inbred C57BL , Protein C/genetics , Recombinant Proteins/genetics
3.
Blood ; 137(5): 678-689, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33538796

ABSTRACT

Thrombospondin-1 (TSP-1) is released by platelets upon activation and can increase platelet activation, but its role in hemostasis in vivo is unclear. We show that TSP-1 is a critical mediator of hemostasis that promotes platelet activation by modulating inhibitory cyclic adenosine monophosphate (cAMP) signaling. Genetic deletion of TSP-1 did not affect platelet activation in vitro, but in vivo models of hemostasis and thrombosis showed that TSP-1-deficient mice had prolonged bleeding, defective thrombosis, and increased sensitivity to the prostacyclin mimetic iloprost. Adoptive transfer of wild-type (WT) but not TSP-1-/- platelets ameliorated the thrombotic phenotype, suggesting a key role for platelet-derived TSP-1. In functional assays, TSP-1-deficient platelets showed an increased sensitivity to cAMP signaling, inhibition of platelet aggregation, and arrest under flow by prostacyclin (PGI2). Plasma swap experiments showed that plasma TSP-1 did not correct PGI2 hypersensitivity in TSP-1-/- platelets. By contrast, incubation of TSP-1-/- platelets with releasates from WT platelets or purified TSP-1, but not releasates from TSP-1-/- platelets, reduced the inhibitory effects of PGI2. Activation of WT platelets resulted in diminished cAMP accumulation and downstream signaling, which was associated with increased activity of the cAMP hydrolyzing enzyme phosphodiesterase 3A (PDE3A). PDE3A activity and cAMP accumulation were unaffected in platelets from TSP-1-/- mice. Platelets deficient in CD36, a TSP-1 receptor, showed increased sensitivity to PGI2/cAMP signaling and diminished PDE3A activity, which was unaffected by platelet-derived or purified TSP-1. This scenario suggests that the release of TSP-1 regulates hemostasis in vivo through modulation of platelet cAMP signaling at sites of vascular injury.


Subject(s)
Blood Platelets/physiology , Cyclic AMP/physiology , Hemorrhagic Disorders/genetics , Hemostasis/physiology , Thrombospondin 1/physiology , Animals , Bleeding Time , Blood Platelets/drug effects , CD36 Antigens/deficiency , CD36 Antigens/physiology , Cells, Cultured , Chlorides/toxicity , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cytoplasmic Granules/metabolism , Epoprostenol/physiology , Ferric Compounds/toxicity , Humans , Iloprost/pharmacology , Mice , Mice, Inbred C57BL , Platelet Transfusion , Second Messenger Systems/physiology , Thrombosis/chemically induced , Thrombosis/prevention & control , Thrombospondin 1/deficiency , Thrombospondin 1/pharmacology
4.
Ecotoxicol Environ Saf ; 249: 114364, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36508806

ABSTRACT

Ferric citrate (FC) has been used as an iron fortifier and nutritional supplement, which is reported to induce colitis in rats, however the underlying mechanism remains to be elucidated. We performed a 16-week study of FC in male healthy C57BL/6 mice (nine-month-old) with oral administration of Ctr (0.9 % NaCl), 1.25 % FC (71 mg/kg/bw), 2.5 % FC (143 mg/kg/bw) and 5 % FC (286 mg/kg/bw). FC-exposure resulted in colon iron accumulation, histological alteration and reduce antioxidant enzyme activities, such as glutathione (GSH), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) and total antioxidant capacity (T-AOC), together with enhanced lipid peroxidation level, including malondialdehyde (MDA) level and 4-Hydroxynonenal (4-HNE) protein expression. Exposure to FC was associated with upregulated levels of the interleukin (IL)- 6, IL-1ß, IL-18, IL-8 and tumor necrosis factor α (TNF-α), while down-regulated levels of IL-4 and IL-10. Exposure to FC was positively associated with the mRNA and protein expressions of cysteine-aspartic proteases (Caspase)- 9, Caspase-3, Bcl-2-associated X protein (Bax), while negatively associated with B-cell lymphoma 2 (Bcl2) in mitochondrial apoptosis signaling pathway. FC-exposure changed the diversity and composition of gut microbes. Additionally, the serum lipopolysaccharide (LPS) contents increased in FC-exposed groups when compared with the control group, while the expression of colonic tight junction proteins (TJPs), such as Claudin-1 and Occludin were decreased. These findings indicate that the colonic mucosal injury induced by FC-exposure are associated with oxidative stress generation, inflammation response and cell apoptosis, as well as the changes in gut microbes diversity and composition.


Subject(s)
Apoptosis , Colon , Ferric Compounds , Food, Fortified , Gastrointestinal Microbiome , Inflammation , Oxidative Stress , Animals , Male , Mice , Rats , Apoptosis/drug effects , Colon/drug effects , Colon/metabolism , Ferric Compounds/toxicity , Food, Fortified/toxicity , Gastrointestinal Microbiome/drug effects , Glutathione/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Intestinal Mucosa/drug effects , Iron/metabolism , Mice, Inbred C57BL , Superoxide Dismutase/metabolism
5.
Ann Neurol ; 89(3): 444-458, 2021 03.
Article in English | MEDLINE | ID: mdl-33219556

ABSTRACT

OBJECTIVE: It is unclear if stopping treatment with dabigatran, a new oral anticoagulant (NOAC), induces a paradoxical rebound prothrombotic state. We investigated if short-term (1-3 days) dabigatran cessation is associated with a higher thrombus volume than expected from a simple reversal of the anticoagulant effect. METHODS: Ten-week-old C57Bl/6 mice (n = 338) received one of the following oral treatments: phosphate-buffered saline (PBS), dabigatran for 7 days with or without 1 to 4 day cessation, and aspirin in either a single dose or daily for 7 days. Some of the animals that ceased dabigatran for 1 to 3 days received single-dose aspirin. Thereafter, we induced FeCl3 -mediated carotid thrombosis in 130 mice, after which we performed micro computed tomography thrombus imaging. The other 208 mice underwent coagulation assays or platelet function tests. As an explorative pilot study, we reviewed the medical records of 18 consecutive patients with NOAC cessation-related cerebral infarction in a large acute stroke cohort. RESULTS: We observed a ~ 40% higher volume of carotid thrombus after dabigatran cessation at 1 to 3 days than after vehicle treatment and showed that this effect could be prevented by single-dose aspirin pretreatment. Dabigatran cessation unduly increased platelet aggregability for 2 days after drug cessation, an effect mediated through thrombin or arachidonic acid, which effect was significantly attenuated by single-dose aspirin pretreatment. In patients, short-term (≤ 3 days) cessation of NOAC therapy, compared with longer-term (≥ 5 days) cessation, tended to be associated with relatively high stroke severity. INTERPRETATION: We provide the first preclinical evidence that a rebound prothrombotic state follows short-term cessation of dabigatran therapy. ANN NEUROL 2021;89:444-458.


Subject(s)
Antithrombins/adverse effects , Carotid Artery Thrombosis/diagnostic imaging , Dabigatran/adverse effects , Deprescriptions , Platelet Aggregation/drug effects , Substance Withdrawal Syndrome/blood , Thrombophilia/blood , Aged , Aged, 80 and over , Animals , Antithrombins/pharmacology , Arachidonic Acid/blood , Aspirin/pharmacology , Carotid Artery Thrombosis/chemically induced , Carotid Artery Thrombosis/prevention & control , Cerebral Infarction/diagnostic imaging , Cerebral Infarction/etiology , Cerebral Infarction/physiopathology , Cerebral Infarction/prevention & control , Chlorides/toxicity , Computed Tomography Angiography , Dabigatran/pharmacology , Factor Xa Inhibitors/adverse effects , Female , Ferric Compounds/toxicity , Humans , Ischemic Stroke/diagnostic imaging , Ischemic Stroke/etiology , Ischemic Stroke/physiopathology , Ischemic Stroke/prevention & control , Magnetic Resonance Angiography , Male , Mean Platelet Volume , Mice , Noxae/toxicity , Pilot Projects , Platelet Aggregation Inhibitors/pharmacology , Platelet Count , Pyrazoles/adverse effects , Pyridones/adverse effects , Rivaroxaban/adverse effects , Severity of Illness Index , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/prevention & control , Thrombin/metabolism , Thrombophilia/etiology , Thrombophilia/prevention & control , X-Ray Microtomography
6.
J Nanobiotechnology ; 20(1): 98, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35236363

ABSTRACT

Iron oxide nanoparticles (IONPs)-based contrast agents are widely used for T2-weighted magnetic resonance imaging (MRI) in clinical diagnosis, highlighting the necessity and importance to evaluate their potential systematic toxicities. Although a few previous studies have documented the toxicity concerns of IONPs to major organs, limited data are available on the potential reproductive toxicity caused by IONPs, especially when administrated via intravenous injection to mimic clinical use of MRI contrast agents. Our study aimed to determine whether exposure to IONPs would affect male reproductive system and cause other related health concerns in ICR mice. The mice were intravenously injected with different concentrations IONPs once followed by routine toxicity tests of major organs and a series of reproductive function-related analyses at different timepoints. As a result, most of the contrast agents were captured by reticuloendothelial system (RES) organs such as liver and spleen, while IONPs have not presented adverse effects on the normal function of these major organs. In contrast, although IONPs were not able to enter testis through the blood testicular barrier (BTB), and they have not obviously impaired the overall testicular function or altered the serum sex hormones levels, IONPs exposure could damage Sertoli cells in BTB especially at a relative high concentration. Moreover, IONPs administration led to a short-term reduction in the quantity and quality of sperms in a dose-dependent manner, which might be attributed to the increase of oxidative stress and apoptotic activity in epididymis. However, the semen parameters have gradually returned to the normal range within 14 days after the initial injection of IONPs. Collectively, these results demonstrated that IONPs could cause reversible damage to the reproductive system of male mice without affecting the main organs, providing new guidance for the clinical application of IONPs as T2-MRI contrast agents.


Subject(s)
Contrast Media , Ferric Compounds , Animals , Contrast Media/toxicity , Ferric Compounds/toxicity , Genitalia , Magnetic Iron Oxide Nanoparticles , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred ICR
7.
Adv Exp Med Biol ; 1357: 303-350, 2022.
Article in English | MEDLINE | ID: mdl-35583650

ABSTRACT

Iron oxide nanoparticles (ION), with unique magnetic properties, have attracted huge scientific attention for a wide variety of uses, mostly in the biomedical field, due to their high biocompatibility, ability to cross biological membranes, appropriate surface architecture and easy conjugation with targeting ligands. Their current applications include diagnostic imaging, cell labelling, site-directed drug delivery and anticancer hyperthermia therapy. The ION surface may be modified by coating with different materials, aiming to stabilize the nanoparticles in different environments, to allow biomolecule binding favouring surface attachments with several molecules, and to prolong the recognition time by the immune system. Although the potential benefits of ION are considerable, and more and more ION are being manufactured to meet the demands of the rapidly proliferating field of nanomedicine, there is an urgent need to define their toxicological profile in order to avoid any potential health risks associated with their exposure and to reach optimal benefits of their use. The purpose of this chapter is to de-scribe the current knowledge on the ION toxicological features, addressing their structure and physicochemical characteristics, main exposure pathways and toxicokinetic aspects, interaction with cells, and their toxic effects, with special attention to those at the cellular and molecular level.


Subject(s)
Nanomedicine , Nanoparticles , Drug Delivery Systems/adverse effects , Ferric Compounds/chemistry , Ferric Compounds/therapeutic use , Ferric Compounds/toxicity , Magnetic Iron Oxide Nanoparticles , Magnetics , Nanoparticles/chemistry , Nanoparticles/toxicity
8.
J Mater Sci Mater Med ; 33(1): 10, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35022850

ABSTRACT

Herein we report synthesis of hematite (α-Fe2O3) nanorods by calcinating hydrothermally synthesized goethite nanorods at 5000C. The structural, optical and MRI imaging guided cancer therapeutic properties of fabricated nanorods have been discussed in this manscript. FESEM and TEM imaging techniques were used to confirm the nanorod like morphology of as prepared materials. As we know that Fe2O3 nanorods with size in the range of 25-30 nm exhibit super magnetism. After coating with the PEG, the as prepared nanorods can be used as T2 MR imaging contrast agents. An excellent T2 MRI contrast of 38.763 mM-1s-1 achieved which is highest reported so far for α-Fe2O3. Besides the as prepared nanorods display an excellent photothermal conversion efficiency of 39.5% thus acts as an excellent photothermal therapeutic agent. Thus, we envision the idea of testing our nanorods for photothermal therapy and MR imaging application both in vitro and in vivo, achieving an excellent T2 MRI contrast and photothermal therapy effect with as prepared PEGylated nanorods.


Subject(s)
Ferric Compounds/chemistry , Nanotubes/chemistry , Animals , Biocompatible Materials/chemistry , Cell Line , Cell Survival , Female , Ferric Compounds/toxicity , HeLa Cells , Humans , In Vitro Techniques , Magnetic Resonance Imaging , Materials Testing , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Microscopy, Electron, Scanning , Nanotubes/toxicity , Nanotubes/ultrastructure , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Phototherapy/methods , Polyethylene Glycols/chemistry , Spectrum Analysis, Raman , X-Ray Diffraction
9.
Pak J Pharm Sci ; 35(1(Supplementary)): 297-303, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35228191

ABSTRACT

Thrombosis, the formation of blood clots due to platelet aggregation, vascular injury or hypercoagulability, leads to cardiovascular pathologies including myocardial or cerebral infarction. Antiplatelet and thrombolytic agents have promising effects in ameliorating thromboembolism and dissolving blood clots. However, the associated limitations generate the need to explore agents from natural origin. The aim of the study was to explore the potential of aqueous methanolic extract (Sc.Cr) of an indigenous plant, Sida cordifolia L., traditionally used for cardiovascular complaints. Sc.Cr was evaluated by clot lysis assay, acute pulmonary embolism, carrageenan-induced tail vein thrombosis and ferric chloride-induced carotid arterial thrombosis models. Hemostasis parameters were increased in a dose-dependent manner. Histological studies showed restoration with clear alveolar spaces and less red blood cell congestion. Significant reduction in infarcted length of thrombus, escalation in coagulation parameters with a profound decrease in platelet count (PC) were observed. Arterial occlusion time was increased with a reduction in weight of thrombus dose-dependently with significant augmentation in PT and APTT. Sc.Cr was also analyzed for phytochemical constituents and antioxidant potential. The results demonstrated the antithrombotic and thrombolytic potential of Sc.Cr using in vitro and in vivo experimental models.


Subject(s)
Anticoagulants/pharmacology , Plant Extracts/pharmacology , Sida Plant/chemistry , Thrombosis/drug therapy , Animals , Anticoagulants/chemistry , Carrageenan/toxicity , Chlorides/toxicity , Collagen/toxicity , Epinephrine/toxicity , Female , Ferric Compounds/toxicity , Lung/drug effects , Lung/pathology , Male , Mice , Phytochemicals/chemistry , Phytochemicals/pharmacology , Plant Extracts/chemistry , Rats , Rats, Wistar , Thrombosis/chemically induced
10.
Neurochem Res ; 46(8): 1981-1994, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33942214

ABSTRACT

Isorhynchophylline (IRN), a component of traditional Chinese herb Uncaria rhynchophylla, possesses strong antioxidant activity. Ferroptosis induced by iron overload causes cell oxidative stress after intracerebral hemorrhage (ICH). Therefore, this study aims to explore the effects of IRN on the ferroptosis following ICH. In this study, mouse hippocampal HT-22 cells were treated with ferric ammonium citrate (FAC) alone or together with IRN, and we found IRN reduced the FAC-induced cell damage. Then, cells were treated with IRN following treatment with FAC after transfection with miR-122-5p inhibitor, and the results showed IRN reduced the FAC-induced decrease of miR-122-5p levels and relieved the ferroptosis by detecting ferroptotic marker proteins, iron ion concentration and oxidative stress level; after transfection with miR-122-5p inhibitor, the protective effects of IRN against FAC-induced ferroptosis in these cells were weakened. TP53 (also known as p53) was verified as a target of miR-122-5p by using dual luciferase reporter assay, and restoration of TP53 attenuated the effects of miR-122-5p on ferroptotic marker proteins expression, iron ion concentration and lipid ROS levels, as well as solute carrier family seven member 11 (SLC7A11) mRNA expression. SLC7A11 siRNA reversed the inhibitory effects of IRN on FAC-induced ferroptosis and oxidative stress levels. Subsequently, IRN increased the mNSS score, and decreased brain water content and EB content in ICH model. Moreover, IRN decreased ferroptosis and lipid ROS level, upregulated the expression of miR-122-5p and SLC7A11 mRNA, and inhibited TP53 expression. Our findings reveal that IRN protects neurocyte from ICH-induced ferroptosis via miR-122-5p/TP53/SLC7A11 pathway, which may provide a potential therapeutic mechanism for ICH.


Subject(s)
Cerebral Hemorrhage/drug therapy , Ferroptosis/drug effects , Neuroprotective Agents/therapeutic use , Oxindoles/therapeutic use , Amino Acid Transport System y+/metabolism , Animals , Cell Line , Cerebral Hemorrhage/metabolism , Ferric Compounds/toxicity , Male , Mice , MicroRNAs/metabolism , Neuroprotective Agents/pharmacology , Oxindoles/pharmacology , Quaternary Ammonium Compounds/toxicity , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
11.
Biotechnol Lett ; 43(8): 1659-1673, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33934256

ABSTRACT

OBJECTIVE: An attractive cell source for stem cell-based therapy are WJ-MSCs. Hence, tracking WJ-MSCs using non-invasive imaging procedures (such as MRI) and contrast agents (Zn0.5Ni0.5Fe2O4, NFNPs) are required to evaluate cell distribution, migration, and differentiation. RESULTS: Results showed that the bare and dextrin-coated NFNPs were internalized inside the WJ-MSCs and had no effect on the cell viability, proliferation, apoptosis, karyotyping, and morphology of WJ-MSCs up to 125 µg/mL. Besides, treated WJ-MSCs were differentiated into osteo/adipocyte-like cells. The expression of RUNX 2, SPP 1 (P < 0.05), and OCN (P > 0.05) genes in the WJ-MSCs treated with dextrin-coated NFNPs was higher than the untreated WJ-MSCs; and the expression of CFD, LPL, and PPAR-γ genes was reduced in WJ-MSCs treated with both NFNPs in comparison with the untreated WJ-MSCs (P > 0.05). CONCLUSION: Overall, results showed that dextrin-coated NFNPs had no adverse effect on the cellular characteristics, proliferation, and differentiation of WJ-MSCs, and suggesting their potential clinical efficacy.


Subject(s)
Adipogenesis/drug effects , Ferric Compounds/toxicity , Magnetic Iron Oxide Nanoparticles/toxicity , Mesenchymal Stem Cells/drug effects , Nickel/toxicity , Osteogenesis/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Humans , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/metabolism
12.
Toxicol Ind Health ; 37(12): 763-775, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34797179

ABSTRACT

Nanoparticles of iron oxide, with diameters beteween 1 to 100 nm, have notable implications for human health and well being. In the current study, we have investigated the effects of iron oxide nanoparticles (IONP) exposure on general physiology and health of adult Wistar rats. IONP used in the study had spherical shape and average size in the range of 15-20 nm. A total of eight groups of rats were repeatedly injected with 0 (control), 20, 40, and 80 mg IONP per kg body weight intraperitoneally under two different exposure schemes (sub-acute and sub-chronic). IONP exposure caused significant changes in lungs, liver, and kidney indices in both exposure schemes. Sub-acute exposure did not affect body weight gain in treated rats, but longer duration exposure was responsible for significant reduction in body weight. Mesenteries, visceral fatty tissues, and visceral peritoneal membranes demonstrated apparent accumulations of IONP in a dose and time-dependent manner. Hematological analysis showed that total RBC count, hemoglobin content, hematocrit, mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC) and mean platelet volume (MPV) were not affected by IONP exposure. Total lymphocyte count, however, was elevated in low- and mid-dose treated rats, but not in high-dose group. Serum lactate dehydrogenase (LDH) increased significantly in rats treated with mid and high doses as compared to control. Serum creatinine and blood urea nitrogen levels were also significantly altered in treated rats. Histological study found significant hepatic damage and mild spleen toxicity. Our report suggests that IONP exhibit significant toxicity in rats.


Subject(s)
Ferric Compounds/toxicity , Nanoparticles/toxicity , Animals , Blood Urea Nitrogen , Creatinine/blood , Dose-Response Relationship, Drug , L-Lactate Dehydrogenase/blood , Liver/pathology , Male , Rats, Wistar , Spleen/pathology
13.
Toxicol Ind Health ; 37(5): 289-302, 2021 May.
Article in English | MEDLINE | ID: mdl-34078188

ABSTRACT

The study on the health effects of combined exposure to various contaminants has been recommended by many authors. The objective of the present study was to examine the effects of the co-exposure to hematite and amorphous silicon dioxide (A-SiO2) nanoparticles on the human lung A549 cell line. The A549 cell line was exposed to 10, 50, 100, and 250 µg/ml concentrations of hematite and A-SiO2 nanoparticles both independently and in combination. Their toxicity in both circumstances was investigated by MTT, intracellular reactive oxygen species, cell glutathione content, and mitochondrial membrane potential tests, and the type of interaction was investigated by statistical analysis using Statistical Package for Social Sciences (SPSS, v. 21). Results showed that the independent exposure to either hematite or A-SiO2 compared with the control group produced more toxic effects on the A549 cell line. The toxicity of combined exposure of the nanoparticles was lower compared with independent exposure, and antagonistic interactive effects were detected. The findings of this study could be useful in clarifying the present debate on the health effects of combined exposure of hematite and A-SiO2 nanoparticles. Because of the complexities of combined exposures, further studies of this kind are recommended.


Subject(s)
Cell Line/drug effects , Environmental Exposure/adverse effects , Ferric Compounds/toxicity , Lung Injury/chemically induced , Lung Injury/physiopathology , Nanoparticles/toxicity , Silicon Dioxide/toxicity , Dose-Response Relationship, Drug , Humans
14.
Toxicol Ind Health ; 37(2): 77-89, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33308053

ABSTRACT

Increased production and use of different types of nanoparticles (NPs) in the last decades has led to increased environmental release of these NPs with potential detrimental effects on both the environment and public health. Information is scarce in the literature on the cytotoxic effect of co-exposure to many NPs as this concern is relatively recent. Thus, in this study, we hypothesized scenarios of cell's co-exposure to two kinds of NPs, solid lipid nanoparticles (SLNs) and superparamagnetic iron oxide nanoparticles (SPIONs), to assess the potential cytotoxicity of exposure to NPs combination. Cytotoxicity of SPIONs, SLNs, and their 1:1 mixture (MIX) in six tumor and six non-tumor cell lines was investigated. The mechanisms underlining the induced cytotoxicity were studied through cell cycle analysis, detection of reactive oxygen species (ROS), and alterations in mitochondrial membrane potential (ΔΨM). Double staining with acridine orange and ethidium bromide was also used to confirm cell morphology alterations. The results showed that SPIONs induced low cytotoxicity compared to SLNs. However, the mixture of SPIONs and SLNs showed synergistic, antagonistic, and additive effects based on distinct tests such as viability assay, ROS generation, ΔΨM, and DNA damage, depending on the cell line. Apoptosis triggered by ROS and disturbances in ΔΨM are the most probable related mechanisms of action. As was postulated, there is possible cytotoxic interaction between the two kinds of NPs.


Subject(s)
Cell Survival/drug effects , Lipids/toxicity , Magnetic Iron Oxide Nanoparticles/toxicity , Animals , Cell Line, Tumor , DNA Damage/drug effects , Ferric Compounds/toxicity , Humans , Mice , Nanoparticles/toxicity
15.
Cancer Invest ; 38(1): 61-84, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31791151

ABSTRACT

Cancer treatment by magnetic hyperthermia offers numerous advantages, but for practical applications many variables still need to be adjusted before developing a controlled and reproducible cancer treatment that is bio-compatible (non-damaging) to healthy cells. In this work, Fe3O4 and CoFe2O4 were synthesized and systematically studied for the development of efficient therapeutic agents for applications in hyperthermia. The biocompatibility of the materials was further evaluated using HepG2 cells as biological model. Colorimetric and microscopic techniques were used to evaluate the interaction of magnetic nano-materials (MNMs) and HepG2 cells. Finally, the behavior of MNMs was evaluated under the influence of an alternating magnetic field (AMF), observing a more efficient temperature increment for CoFe2O4, a desirable behavior for biomedical applications since lower doses and shorter expositions to alternating magnetic field might be required.


Subject(s)
Hyperthermia, Induced/methods , Magnetite Nanoparticles/administration & dosage , Nanomedicine/methods , Neoplasms/therapy , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Biocompatible Materials/toxicity , Cobalt/administration & dosage , Cobalt/chemistry , Cobalt/toxicity , Colorimetry , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Ferric Compounds/toxicity , Ferrosoferric Oxide/administration & dosage , Ferrosoferric Oxide/chemistry , Ferrosoferric Oxide/toxicity , Hep G2 Cells , Humans , Hyperthermia, Induced/adverse effects , Liver/radiation effects , Magnetic Field Therapy/adverse effects , Magnetic Field Therapy/methods , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity , Male , Materials Testing/methods , Rats , Time Factors , Toxicity Tests/methods
16.
Arterioscler Thromb Vasc Biol ; 39(3): 311-318, 2019 03.
Article in English | MEDLINE | ID: mdl-30786739

ABSTRACT

Murine models are widely used valuable tools to study deep vein thrombosis. Leading experts in venous thrombosis research came together through the American Venous Forum to develop a consensus on maximizing the utility and application of available mouse models of venous thrombosis. In this work, we provide an algorithm for model selection, with discussion of the advantages, disadvantages, and applications of the main mouse models of venous thrombosis. Additionally, we provide a detailed surgical description of the models with guidelines to validate surgical technique.


Subject(s)
Disease Models, Animal , Mice , Venous Thrombosis , Algorithms , Animals , Chlorides/toxicity , Electrolysis , Endothelial Cells/drug effects , Endothelium, Vascular/pathology , Ferric Compounds/toxicity , Free Radicals , Hemorheology , Ligation , Recurrence , Research Design , Veins/surgery , Venous Thrombosis/chemically induced , Venous Thrombosis/etiology , Venous Thrombosis/physiopathology , Venules
17.
Arterioscler Thromb Vasc Biol ; 39(3): 402-412, 2019 03.
Article in English | MEDLINE | ID: mdl-30602303

ABSTRACT

Objective- Mutations in Krüppel like factor-11 ( KLF11), a gene also known as maturity-onset diabetes mellitus of the young type 7, contribute to the development of diabetes mellitus. KLF11 has anti-inflammatory effects in endothelial cells and beneficial effects on stroke. However, the function of KLF11 in the cardiovascular system is not fully unraveled. In this study, we investigated the role of KLF11 in vascular smooth muscle cell biology and arterial thrombosis. Approach and Results- Using a ferric chloride-induced thrombosis model, we found that the occlusion time was significantly reduced in conventional Klf11 knockout mice, whereas bone marrow transplantation could not rescue this phenotype, suggesting that vascular KLF11 is critical for inhibition of arterial thrombosis. We further demonstrated that vascular smooth muscle cell-specific Klf11 knockout mice also exhibited significantly reduced occlusion time. The expression of tissue factor (encoded by the F3 gene), a main initiator of the coagulation cascade, was increased in the artery of Klf11 knockout mice, as determined by real-time quantitative polymerase chain reaction and immunofluorescence. Furthermore, vascular smooth muscle cells isolated from Klf11 knockout mouse aortas showed increased tissue factor expression, which was rescued by KLF11 overexpression. In human aortic smooth muscle cells, small interfering RNA-mediated knockdown of KLF11 increased tissue factor expression. Consistent results were observed on adenovirus-mediated overexpression of KLF11. Mechanistically, KLF11 downregulates F3 at the transcriptional level as determined by reporter and chromatin immunoprecipitation assays. Conclusions- Our data demonstrate that KLF11 is a novel transcriptional suppressor of F3 in vascular smooth muscle cells, constituting a potential molecular target for inhibition of arterial thrombosis.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Repressor Proteins/physiology , Thromboplastin/biosynthesis , Thrombosis/prevention & control , Animals , Antithrombin III/analysis , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Bone Marrow Transplantation , Cells, Cultured , Chlorides/toxicity , Chromatin Immunoprecipitation , Down-Regulation , Female , Ferric Compounds/toxicity , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Hydrolases/analysis , Platelet Aggregation , RNA Interference , Recombinant Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/deficiency , Repressor Proteins/genetics , Thromboplastin/genetics , Thrombosis/chemically induced , Transcription, Genetic
18.
Platelets ; 31(4): 432-438, 2020 May 18.
Article in English | MEDLINE | ID: mdl-31608756

ABSTRACT

The ferric chloride models of arterial thrombosis are useful tools with which to investigate the cellular and molecular mechanisms that contribute to arterial thrombosis. Recent insights have, however, revealed the complex and multifaceted mechanism by which ferric chloride induces thrombus formation. Here, we discuss the strengths and weaknesses of the ferric chloride models of arterial thrombosis. Particular focus is given to the phenotypes of different knockout mice in the ferric chloride models and how these compare to other models with independent modes of initiation. Further, we discuss the relevance of the ferric chloride models to the human pathology of atherothrombotic disease.


Subject(s)
Carotid Artery Thrombosis/metabolism , Chlorides/toxicity , Disease Models, Animal , Erythrocytes/metabolism , Ferric Compounds/toxicity , Animals , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Blood Platelets/pathology , Carotid Artery Thrombosis/chemically induced , Chlorides/metabolism , Ferric Compounds/metabolism , Humans , Mice , Mice, Knockout
19.
Platelets ; 31(4): 439-446, 2020 May 18.
Article in English | MEDLINE | ID: mdl-31957516

ABSTRACT

In spite of current treatment strategies, myocardial infarction and stroke are still major causes of death worldwide. These events are triggered by damage of an atherosclerotic plaque, resulting in occlusive thrombus formation. Mouse studies have significantly contributed to our understanding of the mechanisms of atherogenesis and of thrombosis following plaque injury, but the extent to which the mouse serves as an accurate model of human disease is open to discussion. In this review, we provide a detailed overview and comparison of the described mouse models for atherothrombosis including their (dis)advantages. Herein guidance is provided on how to select a suitable atherothrombosis model for research questions primarily relevant to the field of thrombosis.


Subject(s)
Carotid Artery Thrombosis/etiology , Disease Models, Animal , Mice , Plaque, Atherosclerotic , Animals , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Blood Coagulation , Blood Platelets/metabolism , Blood Platelets/physiology , Carotid Artery Thrombosis/chemically induced , Carotid Artery Thrombosis/metabolism , Chlorides/toxicity , Diet, High-Fat , Ferric Compounds/toxicity , Humans , Ligation , Mice, Knockout, ApoE , Plaque, Atherosclerotic/chemically induced , Plaque, Atherosclerotic/pathology , Ultrasonic Waves
20.
Platelets ; 31(4): 447-454, 2020 May 18.
Article in English | MEDLINE | ID: mdl-31625437

ABSTRACT

Deep vein thrombosis (DVT) is a disease with high prevalence and morbidity. It can lead to pulmonary embolism with severe respiratory insufficiency and risk of death. Mechanisms behind all stages of DVT, such as thrombosis commencement, propagation, and resolution, remain incompletely understood. Animal models represent an invaluable tool to explore these problems and identify new targets for DVT prevention and treatment. In this review, we discuss existing models of venous thrombosis, their advantages and disadvantages, and applicability to studying different aspects of DVT pathophysiology. We also speculate about requirements for an "ideal model" that would best recapitulate features of human DVT and discuss readouts of various models.


Subject(s)
Disease Models, Animal , Mice , Pulmonary Embolism/etiology , Venous Thrombosis/etiology , Venous Thrombosis/physiopathology , Animals , Chlorides/toxicity , Constriction, Pathologic/physiopathology , Constriction, Pathologic/surgery , Femoral Vein/injuries , Femoral Vein/pathology , Femoral Vein/surgery , Ferric Compounds/toxicity , Ligation , Pulmonary Embolism/chemically induced , Pulmonary Embolism/complications , Pulmonary Embolism/physiopathology , Vena Cava, Inferior/injuries , Vena Cava, Inferior/pathology , Vena Cava, Inferior/surgery , Venous Thrombosis/diagnostic imaging , Venous Thrombosis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL