Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 234
1.
Exp Eye Res ; 216: 108947, 2022 03.
Article En | MEDLINE | ID: mdl-35074344

Zebrafish possess the ability to completely regenerate the retina following injury, however little is understood about the damage signals that contribute to inducing Müller glia reprogramming and proliferation to regenerate lost neurons. Multiple studies demonstrated that iron contributes to various retinal injuries, however no link has been shown between iron and zebrafish retinal regeneration. Here we demonstrate that Müller glia exhibit transcriptional changes following injury to regulate iron levels within the retina, allowing for increased iron uptake and decreased export. The response of the zebrafish retina to intravitreal iron injection was then characterized, showing that ferrous, and not ferric, iron induces retinal cell death. Additionally, iron chelation resulted in decreased numbers of TUNEL-positive photoreceptors and fewer proliferating Müller glia. Despite the contribution of iron to retinal cell death, inhibition of ferroptosis did not significantly reduce cell death following light treatment. Finally, we demonstrate that both the anti-ferroptotic protein Glutathione peroxidase 4b and the Transferrin receptor 1b are required for Müller glia proliferation following light damage. Together these findings show that iron contributes to cell death in the light-damaged retina and is essential for inducing the Müller glia regeneration response.


Cell Proliferation/drug effects , Ependymoglial Cells/drug effects , Ferrous Compounds/toxicity , Photoreceptor Cells/drug effects , Radiation Injuries, Experimental/etiology , Retinal Degeneration/chemically induced , Animals , Animals, Genetically Modified , Apoptosis , Deferiprone/pharmacology , Ependymoglial Cells/metabolism , In Situ Nick-End Labeling , Intravitreal Injections , Light , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Photoreceptor Cells/radiation effects , Radiation Injuries, Experimental/metabolism , Receptors, Transferrin/metabolism , Retinal Degeneration/metabolism , Zebrafish , Zebrafish Proteins/metabolism
2.
Angew Chem Int Ed Engl ; 60(32): 17570-17578, 2021 08 02.
Article En | MEDLINE | ID: mdl-34041833

The clinical application of chemodynamic therapy is impeded by the insufficient intracellular H2 O2 level in tumor tissues. Herein, we developed a supramolecular nanoparticle via a simple one-step supramolecular polymerization-induced self-assembly process using platinum (IV) complex-modified ß-cyclodextrin-ferrocene conjugates as supramolecular monomers. The supramolecular nanoparticles could dissociate rapidly upon exposure to endogenous H2 O2 in the tumor and release hydroxyl radicals as well as platinum (IV) prodrugs in situ, which is reduced into cisplatin to significantly promote the generation of H2 O2 in the tumor tissue. Thus, the supramolecular nanomedicine overcomes the limitation of conventional chemodynamic therapy via the self-augmented cascade radical generation and drug release. In addition, dissociated supramolecular nanoparticles could be readily excreted from the body via renal clearance to effectively avoid systemic toxicity and ensure long term biocompatibility of the nanomedicine. This work may provide new insights on the design and development of novel supramolecular nanoassemblies for cascade chemo/chemodynamic therapy.


Antineoplastic Agents/therapeutic use , Drug Carriers/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Polymers/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Coordination Complexes/metabolism , Coordination Complexes/therapeutic use , Coordination Complexes/toxicity , Drug Carriers/chemical synthesis , Drug Carriers/metabolism , Drug Carriers/toxicity , Drug Liberation , Female , Ferrous Compounds/chemical synthesis , Ferrous Compounds/metabolism , Ferrous Compounds/therapeutic use , Ferrous Compounds/toxicity , Hydrogen Peroxide/metabolism , Hydroxyl Radical/metabolism , Metallocenes/chemical synthesis , Metallocenes/metabolism , Metallocenes/therapeutic use , Metallocenes/toxicity , Mice, Inbred BALB C , Nanomedicine/methods , Nanoparticles/chemistry , Nanoparticles/metabolism , Nanoparticles/toxicity , Platinum/chemistry , Polymerization , Polymers/chemical synthesis , Polymers/metabolism , Polymers/toxicity , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/therapeutic use , Prodrugs/toxicity , beta-Cyclodextrins/chemical synthesis , beta-Cyclodextrins/metabolism , beta-Cyclodextrins/therapeutic use , beta-Cyclodextrins/toxicity
3.
Amino Acids ; 53(3): 359-380, 2021 Mar.
Article En | MEDLINE | ID: mdl-33586041

The antioxidant and anti-proinflammatory activities of L-leucine were investigated on oxidative testicular injury, ex vivo. In vitro analysis revealed L-leucine to be a potent scavenger of free radicals, while inhibiting acetylcholinesterase activity. Oxidative injury was induced in testicular tissues using FeSO4. Treatment with L-leucine led to depletion of oxidative-induced elevated levels of NO, MDA, and myeloperoxidase activity, with concomitant elevation of reduced glutathione and non-protein thiol levels, SOD and catalase activities. L-leucine caused a significant (p < 0.05) alteration of oxidative-elevated acetylcholinesterase and chymotrypsin activities, while concomitantly elevating the activities of ATPase, ENTPDase and 5'-nucleotidase. L-leucine conferred a protective effect against oxidative induced DNA damage. Molecular docking revealed molecular interactions with COX-2, IL-1 beta and iNOS. Treatment with L-leucine led to restoration of oxidative depleted ascorbic acid-2-sulfate, with concomitant depletion of the oxidative induced metabolites: D-4-Hydroxy-2-oxoglutarate, L-cystine, adenosine triphosphate, maleylacetoacetic acid, cholesteryl ester, and 6-Hydroxy flavin adenine dinucleotide. Treatment with L-leucine reactivated glycolysis while concomitantly deactivating oxidative-induced citrate cycle and increasing the impact-fold of purine metabolism pathway. L-leucine was predicted not to be an inhibitor of CYP1A2, CYP2C19, CYP2C9, CYP2D6, and CYP3A4, with a predicted LD50 value of 5000 mg/Kg and toxicity class of 5. Additionally, L-leucine showed little or no in vitro cytotoxicity in mammalian cells. These results suggest the therapeutic potentials of L-leucine on oxidative testicular injury, as evident by its ability to attenuate oxidative stress and proinflammation, while stalling cholinergic dysfunction and modulating nucleotide hyrolysis; as well as modulate oxidative dysregulated metabolites and their pathways.


Cholinergic Agents/metabolism , Leucine/pharmacology , Metabolic Networks and Pathways/drug effects , Oxidative Stress/drug effects , Purinergic Agents/metabolism , Testis/injuries , Animals , Anti-Inflammatory Agents/metabolism , Antioxidants/metabolism , Cell Line , Cell Survival/drug effects , Cholinergic Agents/chemistry , DNA Damage/drug effects , Ferrous Compounds/toxicity , Humans , Leucine/chemistry , Male , Molecular Docking Simulation , Rats , Testis/metabolism
4.
Nanotoxicology ; 15(2): 257-275, 2021 03.
Article En | MEDLINE | ID: mdl-33503388

Safety on the use of magnetic nanomaterials (MNMs) has become an active topic of research given all the recent applications of these materials in various fields. It is known that the toxicity of MNMs depends on size, shape, and surface functionalization. In this study, we evaluate the biocompatibility with different aquatic organisms of engineered MNMs-CIT with excellent aqueous dispersion and long-term colloidal stability. Primary producers (the alga Pseudokirchneriella subcapitata), primary consumers (the rotifer Lecane papuana), and predators (the fish, Danio rerio) interacted with these materials in acute and sub-chronic toxicity tests. Our results indicate that P. subcaptita was the most sensitive taxon to MNMs-CIT. Inhibition of their population growth (IC50 = 22.84 mg L-1) elicited cell malformations and increased the content of photosynthetic pigments, likely due to inhibition of cell division (as demonstrated in AFM analysis). For L. papuana, the acute exposure to MNMs shows no significant mortality. However, adverse effects such as decreased rate of population and altered swimming patterns arise after chronic interaction with MNMs. For D. rerio organisms on early life stages, their exposure to MNMs results in delayed hatching of eggs, diminished survival of larvae, altered energy resources allocation (measured as the content of total carbohydrates, lipids, and protein), and increased glucose demand. As to our knowledge, this is the first study that includes three different trophic levels to assess the effect of MNMs in aquatic organisms; furthermore, we demonstrated that these MNMs pose hazards on aquatic food webs at low concentrations (few mgL-1).


Aquatic Organisms/drug effects , Chlorides/toxicity , Ferric Compounds/toxicity , Ferrous Compounds/toxicity , Nanostructures/toxicity , Water Pollutants, Chemical/toxicity , Animals , Biomarkers/analysis , Chlorides/chemistry , Ferric Compounds/chemistry , Ferrous Compounds/chemistry , Food Chain , Larva/drug effects , Magnetic Phenomena , Microalgae/drug effects , Nanostructures/chemistry , Particle Size , Rotifera/drug effects , Surface Properties , Water Pollutants, Chemical/chemistry , Zebrafish/growth & development
5.
Int J Pharm ; 582: 119309, 2020 May 30.
Article En | MEDLINE | ID: mdl-32278055

Ferrous sulfate (FeSO4)-directed dual-cross-linked hydrogels were designed for application in single-syringe injections. The use of FeSO4, rather than other iron salts, can modulate the gelation time and make it available for subcutaneous injection with a single syringe. These hydrogels are based on hyaluronic acid-dopamine (HA-dp) that contain donepezil (DPZ)-entrapping poly(lactic-co-glycolic acid) (PLGA) microsphere (MS). Although DPZ has been administered orally, its sustained release formulation via subcutaneous injection may reduce the dosing frequency for patients with Alzheimer's disease. The HA-dp conjugate was synthesized via an amide bond reaction for coordination of dp with a metal ion (Fe2+ or Fe3+) and self-polymerization of dp. The HA-dp/DPZ-loaded PLGA MS (PD MS)/FeSO4 gel system was considerably hardened via both the coordination of the metal ion with HA-dp and covalent bonding of dp. In addition, a quick restoration of the collapsed gel structure and sustained DPZ release from the HA-dp/PD MS/FeSO4 structure were achieved. The pharmacokinetic parameters after its subcutaneous injection in a rat indicate the sustained release and absorption of DPZ from the HA-dp/PD MS/FeSO4 system. The proposed system can be prepared by a simple method and can be efficiently and safely used for the long-term delivery of DPZ after the subcutaneous injection.


Cross-Linking Reagents/chemistry , Donepezil/administration & dosage , Drug Carriers , Ferrous Compounds/chemistry , Hyaluronic Acid/chemistry , Animals , Cross-Linking Reagents/toxicity , Delayed-Action Preparations , Donepezil/chemistry , Donepezil/pharmacokinetics , Donepezil/toxicity , Dopamine/chemistry , Drug Compounding , Drug Liberation , Ferrous Compounds/toxicity , Hardness , Hyaluronic Acid/toxicity , Hydrogels , Injections, Subcutaneous , Male , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rats, Sprague-Dawley
6.
Curr Pharm Biotechnol ; 21(7): 587-595, 2020.
Article En | MEDLINE | ID: mdl-31893988

OBJECTIVE: The current study reports a green, rapid and one-pot synthesis of FeSO4 nanoparticles using Hibiscus rosasinensis floral extract as a reducing and capping agent. 0.5M of FeSO4 was stirred with the floral extract of H. rosasinensis for around 20 minutes at 37ºC and pH 7. METHODS: The development of pink color was considered as the endpoint of reduction and the nanoparticles were characterized by UV-Vis spectrum, EDAX, DLS, FTIR, FESEM, and XRD. UV-Vis spectral analysis indicated a peak at 530 nm and EDAX measurement revealed the presence of Fe, S, O and C elements in the nanoparticle sample. The FTIR analysis showed amines, alcohol and alkene groups that act as capping agents for the produced nanoparticles. FESEM and XRD determination presented FeSO4 nanoparticles of 40-60 nm in size. The synthesized nanoparticles were found to have antibacterial activity against 6 pathogenic bacteria with MIC and MBC of 40 mg/mL. RESULTS: To determine the toxicity at the eukaryotic level, brine shrimp toxicity assay was conducted and 100% mortality was found at concentrations >0.06 mg/mL. Gel shift assay suggested the mechanism of toxicity of FeSO4 NPs by binding and degradation of DNA molecules. CONCLUSION: From the results, the authors demonstrate the ease of green synthesis of FeSO4 nanoparticles and its bioactivity that may have potential applications as drugs and drug delivery systems against various diseases.


Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , DNA Damage , Ferrous Compounds/chemistry , Green Chemistry Technology/methods , Nanoparticles/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Artemia/drug effects , Artemia/genetics , Biofilms/drug effects , Biofilms/growth & development , Electrophoretic Mobility Shift Assay , Ferrous Compounds/pharmacology , Ferrous Compounds/toxicity , Flowers/chemistry , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Hibiscus , Microbial Sensitivity Tests , Plant Extracts/chemistry , Spectroscopy, Fourier Transform Infrared , Survival Analysis
7.
Horm Mol Biol Clin Investig ; 42(1): 49-56, 2020 Dec 14.
Article En | MEDLINE | ID: mdl-33781008

OBJECTIVES: To compare the effect of Iberet Folic® and Zincofer® on haemoglobin (Hb) and serum ferritin level; and its adverse effect. METHODS: This randomised controlled trial conducted from January 2018 until December 2018. Pregnant women below 34 weeks of gestation, with Hb concentration less than 11 g/dL and serum ferritin level less than 12 ug/L were randomised to receive either one tablet Zincofer® or one tablet Iberet Folate® daily for four weeks. Both groups were compared in terms of effect on Hb level, serum ferritin level, and other haematological indices adverse effect related to treatment, and treatment cost. RESULTS: Hundred and thirty patients were recruited in this study with 68 patients in Iberet Folic group and 62 patients in Zincofer group. The change in the Hb and serum ferritin level from baseline to day 30 did not differ significantly between treatment groups. The mean (±SD) change from baseline to day 30 was 2.15 (±0.59) g/dL in the Iberet Folic group, and 1.98 (±0.49) in the Zincofer (p value = 0.08). Mean serum ferritin at day 30 was 17.2 (±3.68) ug/L and 16.7 (±4.28) ug/L with 8.44 (±3.41) and 8.55 (±3.50) difference, respectively (p = 0.86). Adverse events were comparable in between groups, with p value >0.05. GI intolerance and constipation were among the common side effects, occurred in 34.6 and 29.2% cases, respectively. CONCLUSIONS: Zincofer® offers equivalent efficacy and side effect profile in comparison with Iberet Folic® for the treatment of iron deficiency anaemia (IDA) during pregnancy, but with lower cost.


Anemia, Iron-Deficiency/drug therapy , Ferrous Compounds/therapeutic use , Pregnancy Complications, Hematologic/drug therapy , Adult , Anemia, Iron-Deficiency/blood , Erythrocyte Indices , Female , Ferritins/blood , Ferrous Compounds/administration & dosage , Ferrous Compounds/toxicity , Gastrointestinal Diseases/chemically induced , Hemoglobins/analysis , Humans , Parity , Pregnancy , Pregnancy Complications, Hematologic/blood , Prospective Studies , Recurrence , Socioeconomic Factors
9.
Ecotoxicol Environ Saf ; 189: 109982, 2020 Feb.
Article En | MEDLINE | ID: mdl-31830603

The increasing application of nanomaterials in various fields such as drug delivery, cosmetics, disease detection, cancer treatment, food preservation etc. has resulted in high levels of engineered nanoparticles in the environment, thus leading to higher possibility of direct or indirect interactions between these particles and biological systems. In this study, the toxic effects of three commercially available nanomaterials; copper oxide nanoparticles, copper-iron oxide nanopowders and carbon nanopowders were determined in the human hepatoma HepG2 cells using various toxicological assays which are indicative of cytotoxicity (MTT and neutral red assays), mutagenicity (cytokinesis-block micronucleus assay), oxidative stress (total reactive oxygen species and superoxide anion production) and mitochondrial impairment (cellular oxygen consumption). There was increased cytotoxicity, mutagenicity, and mitochondrial impairment in the cells treated with higher concentrations of the nanomaterials, especially the copper oxide nanoparticles. The fold production of reactive oxygen species was similar at the concentrations tested in this study but longer exposure duration resulted in production of more superoxide anions. The results of this study showed that copper oxide nanoparticles are highly toxic to the human HepG2 cells, thus implying that the liver is a target organ in human for copper oxide nanoparticles toxicity.


Carbon/toxicity , Copper/toxicity , Environmental Pollutants/toxicity , Ferrous Compounds/toxicity , Nanoparticles/toxicity , Carbon/chemistry , Copper/chemistry , DNA Damage/drug effects , Environmental Pollutants/chemistry , Ferrous Compounds/chemistry , Hep G2 Cells , Humans , Mitochondria/drug effects , Nanoparticles/chemistry , Oxidative Stress/drug effects
10.
Environ Sci Pollut Res Int ; 27(2): 1923-1940, 2020 Jan.
Article En | MEDLINE | ID: mdl-31760622

Environmental concern related to Ag+ release from conventional AgNPs is expected to be prevented once contained into a magnetic core like magnetite or CoFe2O4. Accordingly, we obtained CoFe2O4 NPs by microwave-assisted synthesis, which AgNO3 addition rendered Ag@CoFe2O4 NPs. NPs were characterized, and before exploring potential applications, we carried out 7-day wheat toxicity assays. Seed germination and seedling growth were used as toxicity endpoints and photosynthetic pigments and antioxidant enzymes as oxidative stress biomarkers. Total Fe, Co, and Ag determination was initial indicative of Ag@CoFe2O4 NPs uptake by plants. Then NPs localization in seedling tissues was sought by scanning electron microscopy (SEM) and darkfield hyperspectral imaging (DF-HSI). Not any silver ion (Ag+) was detected into the ferrite structure, but results only confirmed the presence of metallic silver (Ag0) adsorbed on the CoFe2O4 NPs surface. Agglomerates of Ag@CoFe2O4 NPs (~10 nm) were fivefold smaller than CoFe2O4 NPs, and ferrimagnetic properties of the CoFe2O4 NPs were conserved after the formation of the Ag@CoFe2O4 composite NPs. Seed germination was not affected by NPs, but root and shoot lengths of seedlings diminished 50% at 54.89 mg/kg and 168.18 mg/kg NPs, respectively. Nonetheless, hormesis was observed in roots of plants exposed to lower Ag@CoFe2O4 NPs treatments. Photosynthetic pigments and the antioxidant enzymes catalase (CAT), superoxide dismutase (SOD), guaiacol peroxidase (GPX), and ascorbate peroxidase (APX) indicated oxidative damage by reactive oxygen species (ROS) generation. SEM suggested NPs presence in shoots and roots, whereas DF-HSI confirmed some Ag@CoFe2O4 NPs contained in shoots of wheat plants.


Metal Nanoparticles/toxicity , Silver/toxicity , Triticum/drug effects , Cobalt/toxicity , Ferrous Compounds/toxicity , Germination , Oxidative Stress , Photosynthesis , Seedlings/drug effects , Triticum/enzymology
11.
Toxicol In Vitro ; 58: 170-177, 2019 Aug.
Article En | MEDLINE | ID: mdl-30928693

This research reports on the cytotoxicity of materials present in a wound biosensor on human keratinocytes (HaCaT) to evaluate the biocompatibility of the sensor for continuous wound monitoring applications. Individual and collective effects of the sensor materials, gold (Au) and silver (Ag) nanoparticles (NPs), uricase enzyme (UOx), ferrocene carboxylic acid (FCA), multi-walled carbon nanotubes (MWCNTs) and poly vinyl alcohol-based polymer (PVA-SbQ) on HaCaT were studied. The toxicology profiles of these materials were derived from cell viability, mitochondrial activity retention and apoptotic behavior studies. At the concentrations present in the sensor, the cell viability studies showed minimal toxicity for Au and Ag NPs, UOx and FCA (cell viability >75%), while MWCNTs and PVA-SbQ exhibited excellent biocompatibility towards keratinocytes (cell viability >90%). Resazurin assay confirmed minimal impairment of mitochondrial activity at lower concentrations for all the materials (mitochondrial activity >0.7). The caspase-3/7 apoptotic assay showed no pronounced apoptotic behavior caused by the materials. The material mixtures studied were Au/UOx/FCA/PVA-SbQ, Ag/UOx/FCA/PVA-SbQ, and MWCNTs/UOx/FCA/PVA-SbQ. A higher toxicity profile was observed for the heterogeneous material mixtures as a result of the cumulative effect of the individual materials. However, the biosensor itself was seen to exhibit lower toxicity (~5%) compared to the material mixtures, due to the protective PVA-SbQ capping over the biosensor. This work establishes the biocompatibility of the reported wound sensor for human measurements with minimal toxic effects on human keratinocytes.


Ferrous Compounds/toxicity , Gold/toxicity , Keratinocytes/drug effects , Metal Nanoparticles/toxicity , Nanotubes, Carbon/toxicity , Polyvinyl Alcohol/toxicity , Silver/toxicity , Urate Oxidase/toxicity , Biosensing Techniques , Catalysis , Cell Line , Cell Survival/drug effects , Humans , Metallocenes , Wounds and Injuries
12.
Neurotoxicology ; 73: 142-149, 2019 07.
Article En | MEDLINE | ID: mdl-30914277

Iron is an essential element for multiple metabolic reactions, but excessive iron accumulation in the brain can lead to astrocyte swelling and death and cause cerebral edema. Aquaporin-4 (AQP4) is the important water channel expressed in the astrocytes, and maintains the water homeostasis of the brain. Previous study has shown that iron deposition could increase AQP4 expression, however, the mechanism of AQP4 expression upregulation after iron overload is still unclear. In this study, we investigated the effect of ferrous iron overload on AQP4 expression in cultured mouse astrocytes. Primary cultures of astrocytes were exposed to ferrous iron, and the expression of AQP4 as well as the swelling of astrocyte were determined. AQP4 expression was inhibited by small interfering RNA (siRNA). The role of oxidative stress and mitogen-activated protein kinases (MAPKs) signaling pathway in ferrous iron-induced AQP4 expression upregulation were further studied. Ferrous iron exposure induced astrocyte death as well as cell swelling, and increased AQP4 expression. AQP4 gene silencing after siRNA transfection attenuated ferrous iron-induced astrocyte death. After treatment with antioxidants, the increased AQP4 expression was diminished. MAPKs were activated after ferrous iron treatment, and inhibitors of ERK and p38-MAPK relieved AQP4 expression upregulation as well as astrocyte death. These results suggest that ferrous iron has distinctive toxic effects on cultured astrocytes and induces AQP4 expression upregulation. MAPKs activation may play important roles in ferrous iron-induced astrocyte death through upregulation of AQP4 expression.


Aquaporin 4/metabolism , Astrocytes/drug effects , Ferrous Compounds/toxicity , Mitogen-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Aquaporin 4/genetics , Astrocytes/metabolism , Astrocytes/pathology , Cell Death/drug effects , Cell Size/drug effects , Cells, Cultured , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice, Inbred C57BL , Oxidative Stress/drug effects , Phosphorylation , Signal Transduction , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Gen Physiol Biophys ; 38(1): 27-38, 2019 Jan.
Article En | MEDLINE | ID: mdl-30657458

Iron is a vitally important element for the maintenance of health in living organisms. But, iron overload can be toxic. This study investigated the protective efficacy of quercetin against ferrous sulfate-induced oxidative stress, hepato- and nephrotoxicity in rats. There were five experimental groups (n = 7): Sham (distilled water, 1 ml/day for 14 days, i.p.), Quer (quercetin, 50 mg/kg/day for 14 days, i.p.), DMSO (dimethyl sulfoxide 1%, 1 ml i.p.), Fe (ferrous sulfate, 30 mg/kg/day for 14 days, i.p.), Fe+Quer (ferrous sulfate, 30 mg/kg/day for 14 days; quercetin, 50 mg/kg/day for 11 days from fourth day of ferrous sulfate injection). Blood, 24-h urine and tissue samples were collected at the end of experiment. Quercetin prevented ferrous sulfate-induced hepatotoxicity and nephrotoxicity as indicated by decreased activities of serum hepatic marker enzymes and decreased serum bilirubin concentration, higher levels of serum triglyceride, cholesterol, glucose, albumin and total protein, as well as higher creatinine clearance and lower fractional excretion of sodium. Besides, quercetin decreased malondialdehyde levels and histological damages in the liver and kidney of Fe group as compared with sham, DMSO and Quer groups. The protective effect of quercetin relies, at least partially, on its antioxidative effect which leads to decreased lipid peroxidation as well as iron-chelating property.


Ferrous Compounds/toxicity , Kidney/drug effects , Liver/drug effects , Quercetin/pharmacology , Animals , Antioxidants/pharmacology , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Rats
14.
Curr Org Synth ; 16(1): 160-164, 2019.
Article En | MEDLINE | ID: mdl-31965930

AIM AND OBJECTIVE: Some ferrocenyl derivatives are active in vitro and in vivo against cancer. Generally, ferrocenyl derivatives for cancer research have three key components: a ferrocene moiety, a conjugated linker that lowers the oxidation potential and some derivative (peptide, nucleobase and others) that can interact with biomolecules. Since the pyrimidine fragment can easily pass through the membrane into the cells and become involved in metabolism; it appears to be promising. Furthermore, this fragment is an electron-acceptor group, so a spacer can be excluded. Therefore, the synthesis of 6-ferrocenylpyrimidin-4(3H)-one derivatives and the study of their anticancer activity have scientific and practical interest. METHODS: The syntheses of 6-ferrocenylpyrimidin-4(3H)-one derivatives were performed by the condensation of ethyl 3-ferrocenyl-3-oxopropionate with thiourea or acetamidine or guanidine. The cytotoxicity of four 6- ferrocenylpyrimidin-4(3H)-one derivatives was evaluated by using the MTT assay in vitro against Human breast adenocarcinoma MCF-7 and normal human skin fibroblast HSF cells. The tested derivatives induced a concentration-dependent cytotoxic response in cell lines. RESULTS: A study of the cytotoxic activity of 6-ferrocenylpyrimidin-4(3H)-one derivatives by the MTT test has found that all compounds have a dose-dependent toxic effect on the lines of breast cancer cells (MCF-7) and normal human fibroblast cells (HSF). The most pronounced cytotoxic effect is exhibited by 2-methyl-6-ferrocenylpyrimidin- 4(3H)-one (MCF-7, IC50 17 ± 1 µM). CONCLUSION: The experimental results confirm the importance of investigation and design of ferrocenylpyrimidin- 4(3H)-one derivatives as anticancer agents. Compounds where the pyrimidine derivatives are directly linked to the ferrocene unit rather than via a spacer group also may be of interest for antiproliferative drug design.


Antineoplastic Agents/pharmacology , Ferrous Compounds/pharmacology , Pyrimidinones/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Ferrous Compounds/chemical synthesis , Ferrous Compounds/toxicity , Fibroblasts/drug effects , Humans , MCF-7 Cells , Pyrimidinones/chemical synthesis , Pyrimidinones/toxicity
15.
Chem Asian J ; 13(18): 2730-2738, 2018 Sep 17.
Article En | MEDLINE | ID: mdl-29963768

A promising cancer-targeting agent for the induction of apoptosis in tumor necrosis factor (TNF) proteins, the TNF-related apoptosis-inducing ligand (TRAIL) ligand, has found limited applications in the treatment of cancer cells, owing to its resistance by cancer cell lines. Therefore, the rational design of anticancer agents that could sensitize cancer cells towards TRAIL is of great significance. Herein, we report that synthetic iron(II)-polypyridyl complexes are capable of inhibiting the proliferation of glioblastoma cancer cells and efficiently enhancing TRAIL-induced cell apoptosis. Mechanistic studies demonstrated that the synthesized complexes induced cancer-cell apoptosis through triggering the activation of p38 and p53 and inhibiting the activation of ERK. Moreover, uPA and MMP-2/MMP-9, among the most important metastatic regulatory proteins, were also found to be significantly alerted after the treatment. Furthermore, we also found that tumor growth in nude mice was significantly inhibited by iron complex Fe2 through the induction of apoptosis without clear systematic toxicity, as indicated by histological analysis. Taken together, this study provides evidence for the further development of metal-based anticancer agents and chemosensitizers of TRAIL for the treatment of human glioblastoma cancer cells.


Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Coordination Complexes/therapeutic use , Ferrous Compounds/therapeutic use , Glioblastoma/drug therapy , Phenanthrolines/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Biological Transport , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Coordination Complexes/toxicity , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Ferrous Compounds/chemical synthesis , Ferrous Compounds/pharmacology , Ferrous Compounds/toxicity , Humans , Ligands , Male , Matrix Metalloproteinase 9/genetics , Mice, Nude , Phenanthrolines/chemical synthesis , Phenanthrolines/pharmacology , Phenanthrolines/toxicity , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/metabolism , Urokinase-Type Plasminogen Activator/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Neurotox Res ; 34(3): 640-648, 2018 Oct.
Article En | MEDLINE | ID: mdl-29949107

Molecules exhibiting antioxidant, neuroprotective, and regulatory properties inherent to natural products consumed by humans are gaining attention in biomedical research. Ferulic acid (FA) is a phenolic compound possessing antioxidant and cytoprotective properties. It is found in several vegetables, including sugarcane, where it serves as the main antioxidant component. Here, we compared the antioxidant and cytoprotective effects of FA with those of the total sugarcane aqueous extract (SCAE). Specifically, we assessed biochemical markers of cell dysfunction in rat cortical brain slices and markers of physiological stress in Caenorhabditis elegans upon exposure to toxins evoking different mechanisms of neurotoxicity, including direct oxidative stress and/or excitotoxicity. In rat cortical slices, FA (250 and 500 µM), but not SCAE (~ 270 µM of total polyphenols), prevented the loss of reductive capacity induced by the excitotoxin quinolinic acid (QUIN, 100 µM), the pro-oxidant agent ferrous sulfate (FeSO4, 25 µM), and the dopaminergic pro-oxidant 6-hydroxydopamine (6-OHDA, 100 µM). In wild-type (N2) C. elegans, FA (38 mM) exerted protective effects on decreased survival induced by FeSO4 (15 mM) and 6-OHDA (25 mM), and the motor alterations induced by QUIN (100 mM), FeSO4, and 6-OHDA. In contrast, SCAE (~ 13.5 mM of total polyphenols) evoked protective effects on the decreased survival induced by the three toxic agents, the motor alterations induced by FeSO4, and the reproductive deficit induced by FeSO4. In addition, FA was unable to reverse the decreased survival induced by all these toxins in the skn-1-/- strain (VC1772), which lacks the homolog of mammalian Nrf2, a master antioxidant gene. Altogether, our results suggest that (1) both FA and SCAE afford protection against toxic conditions, (2) not all the effects inherent to SCAE are due to FA, and (3) FA requires the skn-1 pathway to exert its protective effects in C. elegans.


Coumaric Acids/therapeutic use , Neurotoxicity Syndromes/drug therapy , Plant Extracts/therapeutic use , Saccharum/chemistry , Analysis of Variance , Animals , Animals, Genetically Modified , Birth Rate , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Ferrous Compounds/toxicity , In Vitro Techniques , Iron/metabolism , Locomotion/drug effects , Male , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/physiopathology , Oxidopamine/toxicity , Plant Extracts/chemistry , Quinolinic Acid/toxicity , Rats , Rats, Wistar
17.
Sci Rep ; 8(1): 8083, 2018 05 24.
Article En | MEDLINE | ID: mdl-29795396

Increasing utilization of stabilized iron sulfides (FeS) nanoparticles implies an elevated release of the materials into the environment. To understand potential impacts and underlying mechanisms of nanoparticle-induced stress, we used the transcriptome sequencing (RNA-seq) technique to characterize the transcriptomes from adult zebrafish exposed to 10 mg/L carboxymethyl cellulose (CMC) stabilized FeS nanoparticles for 96 h, demonstrating striking differences in the gene expression profiles in liver. The exposure caused significant expression alterations in genes related to immune and inflammatory responses, detoxification, oxidative stress and DNA damage/repair. The complement and coagulation cascades Kyoto encyclopedia of genes and genomes (KEGG) pathway was found significantly up-regulated under nanoparticle exposure. The quantitative real-time polymerase chain reaction using twelve genes confirmed the RNA-seq results. We identified several candidate genes commonly regulated in liver, which may serve as gene indicators when exposed to the nanoparticles. Hepatic inflammation was further confirmed by histological observation of pyknotic nuclei, and vacuole formation upon exposure. Tissue accumulation tests showed a 2.2 times higher iron concentration in the fish tissue upon exposure. This study provides preliminary mechanistic insights into potential toxic effects of organic matter stabilized FeS nanoparticles, which will improve our understanding of the genotoxicity caused by stabilized nanoparticles.


Carboxymethylcellulose Sodium/toxicity , Environmental Exposure/analysis , Ferrous Compounds/toxicity , Nanoparticles/toxicity , Transcriptome/drug effects , Zebrafish , Animals , Environmental Monitoring/methods , Female , Ferrous Compounds/chemistry , Gene Expression Profiling/methods , Male , Nanoparticles/chemistry , Sequence Analysis, RNA/methods , Toxicity Tests , Exome Sequencing , Zebrafish/genetics , Zebrafish/growth & development
18.
J Dev Orig Health Dis ; 9(2): 232-240, 2018 04.
Article En | MEDLINE | ID: mdl-28870272

Mitochondrial dysfunction and resulting changes in adiposity have been observed in the offspring of animals fed a high fat (HF) diet. As iron is an important component of the mitochondria, we have studied the offspring of female rats fed complete (Con) or iron-deficient (FeD) rations for the duration of gestation to test for similar effects. The FeD offspring were ~12% smaller at weaning and remained so because of a persistent reduction in lean tissue mass. The offspring were fed a complete (stock) diet until 52 weeks of age after which some animals from each litter were fed a HF diet for a further 12 weeks. The HF diet increased body fat when compared with animals fed the stock diet, however, prenatal iron deficiency did not change the ratio of fat:lean in either the stock or HF diet groups. The HF diet caused triglyceride to accumulate in the liver, however, there was no effect of prenatal iron deficiency. The activity of the mitochondrial electron transport complexes was similar in all groups including those challenged with a HF diet. HF feeding increased the number of copies of mitochondrial DNA and the prevalence of the D-loop mutation, however, neither parameter was affected by prenatal iron deficiency. This study shows that the effects of prenatal iron deficiency differ from other models in that there is no persistent effect on hepatic mitochondria in aged animals exposed to an increased metabolic load.


Adipose Tissue/metabolism , Aging/metabolism , Anemia, Iron-Deficiency/metabolism , Diet, High-Fat/adverse effects , Liver/metabolism , Mitochondria, Liver/metabolism , Adipose Tissue/drug effects , Adipose Tissue/pathology , Aging/drug effects , Aging/pathology , Anemia, Iron-Deficiency/chemically induced , Anemia, Iron-Deficiency/pathology , Animals , Female , Ferrous Compounds/administration & dosage , Ferrous Compounds/toxicity , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Liver/drug effects , Liver/pathology , Male , Mitochondria, Liver/drug effects , Mitochondria, Liver/pathology , Pregnancy , Rats
19.
Biomed Pharmacother ; 96: 1065-1074, 2017 Dec.
Article En | MEDLINE | ID: mdl-29223552

The antioxidative and antidiabetic effects and toxicity of caffeine-rich infusion of Cola nitida were investigated using in vitro, ex vivo and in silico models. C. nitida was infused in boiling water and allowed to cool before concentrating at <50°C. HPLC analysis of the infusion revealed a caffeine content of 80.08%. The infusion showed potent in vitro antioxidant activity by significantly (p<0.05) scavenging 2,2'-diphenyl-1-picrylhydrazyl (DPPH). It significantly (p<0.05) inhibited α-glucosidase and α-amylase activities. Treatment of Fe2+ induced oxidative hepatic tissues with the infusion led to increase Superoxide Dismutase (SOD) and catalase activities, and glutathione (GSH) level as well as decreased malondialdehyde (MDA) level. FTIR spectroscopy of hepatic metabolite revealed restoration of oxidative-induced depleted functional groups by the infusion. LC-MS analysis of the metabolite also revealed restoration of most depleted metabolites with concomitant generation of 4-O-Methylgallic, (-)-Epicatechin sulfate, L-Arginine, L-tyrosine, Citric acid and Decanoic acid in infusion-treated tissues. Pathway analysis of the identified metabolites revealed the presence of 21 metabolic pathways involved in normal hepatic tissues, 12 in oxidative injured tissues and 17 in the treated tissues. Treatment with the infusion restored 4 metabolic pathways common to the normal tissue and further activated 4 additional pathways. Prediction of oral toxicity of caffeine showed it to belong to class 3, with a LD50 of 127mg/kg. Its toxicity target was predicted as Adenosine Receptor A2a. It was also predicted to be an inhibitor of CYP1A2. These results suggest the antioxidative and antidiabetic properties of C. nitida infusion, with caffeine as the major constituent.


Caffeine/administration & dosage , Carbohydrate Metabolism/physiology , Cola , Ferrous Compounds/toxicity , Liver/metabolism , Plant Extracts/administration & dosage , Animals , Antioxidants/administration & dosage , Antioxidants/isolation & purification , Caffeine/isolation & purification , Carbohydrate Metabolism/drug effects , Liver/drug effects , Male , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Plant Extracts/isolation & purification , Rats , Seeds
20.
Cell Physiol Biochem ; 44(3): 870-883, 2017.
Article En | MEDLINE | ID: mdl-29176318

BACKGROUND/AIMS: Iron overload (IO) is accompanied by hepatic inflammation. The chemokine (C-C motif) ligand 2 (CCL2) mediates inflammation, and its overexpression is associated with IO. However, whether IO results in CCL2 overexpression in the liver and the underlying mechanisms are unclear. METHODS: We subjected mice to IO by administering intraperitoneal injections of dextran-iron or by feeding mice a 3% dextran-iron diet to observe the effects of IO on miR-122/CCL2 expression through real-time qPCR and Western blot analysis. We also used indicators, including the expression of the inflammatory cytokine, the inflammation score based on H&E staining and the serum content of ALT and AST to evaluate the effects of IO on hepatic inflammation. Meanwhile, we observed the effects of vitamin E on IO-induced hepatic inflammation. In cells, we used 100 µΜ FeSO4 or 30 µΜ Holo-Tf to produce IO and observed the roles of miR-122 in regulating CCL2 expression by using miR-122 mimics or inhibitors to overexpress or inhibit miR-122. Then, we used a dual-luciferase reporter assay to prove that miR-122 regulates CCL2 expression through direct binding to its complementary sequence in the CCL2 mRNA 3'UTR. RESULTS: IO induces the downregulation of miR-122 and the upregulation of CCL2, as well as inflammatory responses both in vitro and in vivo. Although IO-induced oxidative stress is eliminated by the antioxidant vitamin E, IO-induced hepatic inflammation still exists, which probably can be explained by the fact that vitamin E has no effects on the miR-122/CCL2 pathway. In in vitro experiments, the overexpression and inhibition of miR-122 significantly reduced and increased CCL2 expression, respectively. The dual-luciferase reporter assay indicates that miR-122 binds CCL2 mRNA 3'UTR. CONCLUSION: We propose the roles of miR-122/CCL2 in IO-induced hepatic inflammation. Our studies should provide a new clue for developing clinical strategies for patients with IO.


Chemokine CCL2/metabolism , Iron-Dextran Complex/toxicity , Liver/pathology , MicroRNAs/metabolism , Up-Regulation/drug effects , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Base Sequence , Cell Line , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/genetics , Ferrous Compounds/toxicity , Humans , Inflammation , Interleukin-6/blood , Iron/analysis , Iron/blood , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oxidative Stress/drug effects , Sequence Alignment , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transferrin/pharmacology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vitamin E/pharmacology
...