Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
1.
J Ethnopharmacol ; 280: 114409, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34265378

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The edible plant Opuntia dillenii (Ker Gawl.) Haw. commonly known as Nagphana, belongs to the Cactaceae family. It is traditionally used to treat various ailments including inflammation, gastric ulcers, diabetes, hepatitis, asthma, whooping cough and intestinal spasm. AIM OF THE STUDY: Despite its traditional use in various countries, detailed toxicological studies of O. dillenii cladode are few. Thus in the current study, toxicity of O. dillenii cladode derived methanol extract, fractions and its α-pyrones: opuntiol and opuntioside have been addressed. METHODS: The test agents were assessed using both in vitro and in vivo toxicity assays. MTT on human embryonic kidney cell line (HEK-293), tryphan blue exclusion in rat neutrophils, Cytokinesis-B block micronucleus (CBMN) in human lymphocytes and genomic DNA fragmentation using agarose gel electrophoresis were performed. In acute toxicity test, mice orally received extract (5 g/kg) for 7 days followed by measurements of relative organ weight, biochemical (blood profile, liver and kidney function test) and histological studies (liver and kidney) were carried out. Rat bone marrow micronucleus genotoxicity assay was also conducted. RESULTS: O. dillenii derived test agents were non-cytotoxic and had no effect on the integrity of DNA. Methanol extract (5 g/kg) orally administered in mice did not cause any significant change in relative organ weights, biochemical parameters and liver and kidney histology as compared to vehicle control. In parallel, extract did not stimulate micronuclei formation in rat bone marrow polychromatic erythrocytes. CONCLUSION: These results led to conclude that edible O. dillenii extract is non-toxic via the oral route and appears to be non-cyto-, hepato-, nephro- or genotoxic, thereby supporting its safe traditional use against various ailments. Therefore, opuntiol and opuntioside may serve as lead compounds in designing new drug(s) derived from edible plants.


Subject(s)
Coumaric Acids/toxicity , Monosaccharides/toxicity , Opuntia/chemistry , Plant Extracts/toxicity , Animals , Coumaric Acids/isolation & purification , DNA Fragmentation/drug effects , Female , HEK293 Cells , Humans , Male , Methanol/chemistry , Mice , Micronucleus Tests , Monosaccharides/isolation & purification , Neutrophils/drug effects , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Pyrones/isolation & purification , Pyrones/toxicity , Rats , Rats, Sprague-Dawley , Toxicity Tests, Acute
2.
Pharm Biol ; 59(1): 232-241, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33632062

ABSTRACT

CONTEXT: Andrographolide (Andro) has a neuroprotective effect and a potential for treating Alzheimer's disease (AD), but the mechanism has not been elucidated. OBJECTIVE: The efficacy of Andro on p62-mediated Kelch-like ECH-associated protein 1(Keap1)-Nuclear factor E2 related factor 2 (Nrf2) pathways in the aluminium maltolate (Al(mal)3)-induced neurotoxicity in PC12 cell was explored. MATERIALS AND METHODS: PC12 cells were induced by Al(mal)3 (700 µM) to establish a neurotoxicity model. Following Andro (1.25, 2.5, 5, 10, 20, 40 µM) co-treatment with Al(Mal)3, cell viability was detected with MTT, protein expression levels of ß-amyloid precursor protein (APP), ß-site APP cleaving enzyme 1 (BACE1), Tau, Nrf2, Keap1, p62 and LC3 were measured via western blotting or immunofluorescence analyses. Nrf2, Keap1, p62 and LC3 mRNA, were detected by reverse transcription-quantitative PCR. RESULTS: Compared with the 700 µM Al(mal)3 group, Andro (5, 10 µM) significantly increased Al(mal)3-induced cell viability from 67.4% to 91.9% and 91.2%, respectively, and decreased the expression of APP, BACE1 and Keap1 proteins and the ratio of P-Tau to Tau (from 2.75- fold to 1.94- and 1.70-fold, 2.12-fold to 1.77- and 1.56-fold, 0.68-fold to 0.51- and 0.55-fold, 1.45-fold to 0.82- and 0.91-fold, respectively), increased the protein expression of Nrf2, p62 and the ratio of LC3-II/LC3-I (from 0.67-fold to 0.93- and 0.94-fold, 0.64-fold to 0.88- and 0.87-fold, 0.51-fold to 0.63- and 0.79-fold, respectively), as well as the mRNA expression of Nrf2, p62 and LC3 (from 0.48-fold to 0.92-fold, 0.49-fold to 0.92-fold, 0.25-fold to 0.38-fold). Furthermore, Nrf2 and p62 nuclear translocation were increased and keap1 in the cytoplasm was decreased in the presence of Andro. Silencing p62 or Nrf2 can significantly reduce the protein and mRNA expression of Nrf2 and p62 under co-treatment with Andro and Al(mal)3. DISCUSSION AND CONCLUSIONS: Our results suggested that Andro could be a promising therapeutic lead against Al-induced neurotoxicity by regulating p62-mediated keap1-Nrf2 pathways.


Subject(s)
Diterpenes/pharmacology , Neuroprotective Agents/pharmacology , Organometallic Compounds/toxicity , Pyrones/toxicity , Animals , Diterpenes/administration & dosage , Dose-Response Relationship, Drug , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/administration & dosage , PC12 Cells , RNA, Messenger/metabolism , Rats , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism
3.
Ecotoxicol Environ Saf ; 208: 111613, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33396133

ABSTRACT

The environmental effects of additives have attracted increasing attention. Sodium dehydroacetate (DHA-S), as an approved preservative, is widely added in processed foods, cosmetics and personal care products. However, DHA-S has been recently reported to induce hemorrhage and coagulation aberration in rats. Yet little is known about the ecotoxicological effect and underlying mechanisms of DHA-S. Here, we utilized the advantage of zebrafish model to evaluate such effects. DHA-S induced cerebral hemorrhage, mandibular dysplasia and pericardial edema in zebrafish after 24 h exposure (48-72 hpf) at 50 mg/L. We also observed the defective heart looping and apoptosis in DHA-S-treated zebrafish through o-dianisidine and acridine orange staining. Meanwhile, DHA-S induced the deficiency of Ca2+ and vitamin D3 in zebrafish. We further demonstrated that DHA-S stimulated Ca2+ influx resulting in Ca2+-dependent mitochondrial damage in cardiomyocytes. Additionally, DHA-S inhibited glucose uptake and repressed the biosynthesis of amino acids. Finally, we identified that sodium bicarbonate could rescue zebrafish from DHA-S induced cardiovascular toxicity. Altogether, our results suggest that DHA-S is a potential risk for cardiovascular system.


Subject(s)
Calcium/metabolism , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Heart/drug effects , Pyrones/toxicity , Zebrafish , Animals , Apoptosis/drug effects , Cardiotoxicity , Cell Line , Cerebral Hemorrhage/chemically induced , Dose-Response Relationship, Drug , Edema, Cardiac/chemically induced , Heart/embryology , Myocardium/metabolism , Myocardium/pathology , Pericardium/drug effects , Pericardium/pathology , Rats , Zebrafish/growth & development
4.
Nat Prod Res ; 35(2): 318-326, 2021 Jan.
Article in English | MEDLINE | ID: mdl-31204847

ABSTRACT

Two new 2-pyrone derivatives sydowiones A-B (1, 2), one new cyclopentenone derivative sydowione C (3), and one new mycotoxin 6-methoxyl austocystin A (4) along with two known analogues paecilpyrone A (5) and austocystin A (6), were isolated from the marine-derived fungus Aspergillus sydowii SCSIO 00305. The structures of 1-4 were elucidated by extensive spectroscopic analysis. The absolute configuration of C-8 in 1 was established by Mosher method, and further confirmed by calculation of the electronic circular dichroism (ECD) spectra. The absolute configuration of C-11 in 3 was also determined by calculation of ECD spectra. The absolute configuration of 6 was determined by a single-crystal X-ray diffraction experiment for the first time. Compounds 1-4 showed moderate toxicity towards brine shrine naupalii with LC50 values of 19.5, 14.3, 8.3 and 2.9 µM, respectively. And 1 and 2 also showed antioxidant activity against 2,2-diphenyl-picrylhydrazyl (DPPH) radicals with IC50 values of 46.0 and 46.6 µM, respectively.[Formula: see text].


Subject(s)
Antioxidants/chemistry , Aspergillus/chemistry , Cyclopentanes/chemistry , Pyrones/chemistry , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Aquatic Organisms/chemistry , Artemia/drug effects , Circular Dichroism , Crystallography, X-Ray , Cyclopentanes/pharmacology , Cyclopentanes/toxicity , Lethal Dose 50 , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Structure , Pyrones/pharmacology , Pyrones/toxicity
5.
Toxicol Mech Methods ; 31(1): 33-42, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32900247

ABSTRACT

Aluminum (Al), a neurotoxic element, can induce Alzheimer's disease (AD) via triggering neuronal death. Ferroptosis is a new type of programmed cell death related to neurological diseases. Unfortunately, its role in aluminum-induced neuronal death remains completely unclear. This study aimed to investigate whether ferroptosis is involved in neuronal death in response to aluminum exposure as well as its underlying mechanism. In this study, rat adrenal pheochromocytoma (PC12) cells were treated with 200 µM aluminum maltolate (Al(mal)3) for 24 h, and related biochemical indicators were assessed to determine whether ferroptosis was induced by aluminum in neurons. Then, the potential mechanism was explored by detecting of these genes and proteins associated with ferroptosis after adding ferroptosis-specific agonist Erastin (5 µM) and antagonist Ferrostatin-1 (Fer-1) (5 µM). The experimental results demonstrated that aluminum exposure significantly increased the death of PC12 cells and caused specific mitochondrial pathological changes of ferroptosis in PC12 cells. Further research confirmed that ferroptosis was triggered by aluminum in PC12 cells by means of activating the oxidative damage signaling pathway, which was displayed as inhibition of the cysteine/glutamate antiporter system (system Xc-), causing the depletion of cellular glutathione (GSH) and inactivation of glutathione peroxidase (GSH-PX) eventually lead to accumulation of reactive oxygen species (ROS). Taken together, ferroptosis was a means of neuronal death induced by aluminum and oxidative damage may be its underlying mechanism, which also provided some new clues to potential target for the intervention and therapy of AD.


Subject(s)
Ferroptosis/drug effects , Mitochondria/drug effects , Neurons/drug effects , Organometallic Compounds/toxicity , Oxidative Stress/drug effects , Pyrones/toxicity , Animals , Mitochondria/metabolism , Mitochondria/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , PC12 Cells , Rats , Reactive Oxygen Species/metabolism , Signal Transduction
6.
Toxicol Appl Pharmacol ; 410: 115354, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33271249

ABSTRACT

Ethyl maltol (EM) is a flavoring agent commonly used in foods that falls under the generally recognized as safe category. It is added to many commercial e-cigarette vaping fluids and has been detected in the aerosols. Considering that EM facilitates heavy metal transport across plasma membranes, and that heavy metals have been detected in aerosols generated from e-cigarettes, this study examines whether EM enhances heavy metal mediated toxicity. A decrease in viability was observed in the Calu-6 and A549 lung epithelial cell lines co-exposed to EM and copper (Cu) but no decrease was observed after co-exposure to EM with iron (Fe). Interestingly, co-exposure to EM and Fe decreased viability of the HEK293 and IMR-90 fibroblast cell lines but co-exposure to EM and Cu did not. Increases in the apoptotic markers Annexin V staining and fragmented nuclei were observed in Calu-6 cells co-exposed to EM and Cu. Co-exposure to EM and Cu in Calu-6 cells resulted in DNA damage as indicated by activation of ATM and expression of γH2A.x foci. Finally, co-exposure to EM and Cu caused oxidative stress as indicated by increases in the generation of reactive oxygen species and the expression of ferritin light chain mRNA and hemeoxygenase-1 mRNA and protein. These data show that co-exposure to EM and Cu, at concentrations that are not toxic for either chemical individually, induce apoptosis and evoke oxidative stress and DNA damage in lung epithelial cells. We suggest that there is a greater risk of lung damage in users of c-cigarette who vape with vaping fluid containing EM.


Subject(s)
Copper/toxicity , Cytotoxins/toxicity , Lung/drug effects , Pyrones/toxicity , Respiratory Mucosa/drug effects , A549 Cells , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Synergism , HEK293 Cells , Humans , Lung/pathology , Respiratory Mucosa/pathology
7.
Antivir Chem Chemother ; 28: 2040206620983780, 2020.
Article in English | MEDLINE | ID: mdl-33353394

ABSTRACT

BACKGROUND: Gallium has demonstrated strong anti-inflammatory activity in numerous animal studies, and has also demonstrated direct antiviral activity against the influenza A H1N1 virus and the human immunodeficiency virus (HIV). Gallium maltolate (GaM), a small metal-organic coordination complex, has been tested in several Phase 1 clinical trials, in which no dose-limiting or other serious toxicity was reported, even at high daily oral doses for several months at a time. For these reasons, GaM may be considered a potential candidate to treat coronavirus disease 2019 (COVID-19), which is caused by the SARS-CoV-2 virus and can result in severe, sometimes lethal, inflammatory reactions. In this study, we assessed the ability of GaM to inhibit the replication of SARS-CoV-2 in a culture of Vero E6 cells. METHODS: The efficacy of GaM in inhibiting the replication of SARS-CoV-2 was determined in a screening assay using cultured Vero E6 cells. The cytotoxicity of GaM in uninfected cells was determined using the Cell Counting Kit-8 (CCK-8) colorimetric assay. RESULTS: The results showed that GaM inhibits viral replication in a dose-dependent manner, with the concentration that inhibits replication by 50% (EC50) being about 14 µM. No cytotoxicity was observed at concentrations up to at least 200 µM. CONCLUSION: The in vitro activity of GaM against SARS-CoV-2, together with GaM's known anti-inflammatory activity, provide justification for testing GaM in COVID-19 patients.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Organometallic Compounds/pharmacology , Pyrones/pharmacology , SARS-CoV-2/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/toxicity , Antiviral Agents/therapeutic use , Antiviral Agents/toxicity , Chlorocebus aethiops , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Iron/metabolism , Organometallic Compounds/therapeutic use , Organometallic Compounds/toxicity , Pyrones/therapeutic use , Pyrones/toxicity , SARS-CoV-2/physiology , Vero Cells , Virus Replication/drug effects
8.
Eur J Med Chem ; 201: 112480, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32652434

ABSTRACT

As part of our effort to develop potential tyrosinase inhibitors, we have conjugated the well-known tyrosinase inhibitor kojic acid (KA) with several phenolic natural products such as umbelliferone, sesamol, thymol, carvacrol, eugenol, isoeugenol, vanillin, isovanillin, and apocynin that some reports have shown their activity on tyrosinase enzyme. The designed compounds were synthesized using click reaction and 1,2,3-triazole formation. All compound showed potent anti-tyrosinase activity significantly higher than KA. The best activities were observed with apocynin and 4-coumarinol analogs (10c and 16c) displaying IC50 values of 0.03 and 0.02 µM, respectively. The potency of 16c was >460-times more than that of KA. Cell-based assays against B16F10 and HFF cells revealed that the representative compounds can efficiently suppress the melanogenesis without significant toxicity on cells.


Subject(s)
Biological Products/pharmacology , Enzyme Inhibitors/pharmacology , Monophenol Monooxygenase/antagonists & inhibitors , Pyrones/pharmacology , Agaricales/enzymology , Animals , Biological Products/chemical synthesis , Biological Products/metabolism , Biological Products/toxicity , Catalytic Domain , Cell Line, Tumor , Copper/chemistry , Enzyme Assays , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/toxicity , Humans , Kinetics , Melanins/metabolism , Mice , Molecular Docking Simulation , Molecular Structure , Monophenol Monooxygenase/chemistry , Monophenol Monooxygenase/metabolism , Protein Binding , Pyrones/chemical synthesis , Pyrones/metabolism , Pyrones/toxicity , Structure-Activity Relationship
9.
Environ Toxicol Pharmacol ; 78: 103406, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32438325

ABSTRACT

The main symptoms of Alzheimer's disease (AD) is the loss of learning and memory ability, of which biological basis is synaptic plasticity. Aluminium has been found to cause changes in synaptic plasticity, but its molecular mechanism was unclear. In this study, Sprague-Dawley rats were injected with aluminium maltol (Al(mal)3) through the lateral ventricle to establish an AD-like model. Y-maze, electrophysiological measurements, Golgi staining, scanning electron microscopy, quantitative real-time polymerase chain reaction, and western blot techniques were used to investigate regulation of the metabolic glutamate receptor 1 (mGluR1) in synaptic plasticity impairment induced by Al(mal)3. The results showed that Al(mal)3 inhibited the induction and maintenance of long-term potentiation in the hippocampal CA1 region. During this process, the expression of mGluR1 was up-regulated and it inhibited the expression and phosphorylation of the N-methyl-D-aspartic acid receptors (NMDARs). This mainly affected NMDAR1 and NMDAR2B but did not affect protein kinase C expression.


Subject(s)
Hippocampus/drug effects , Neuronal Plasticity/drug effects , Organometallic Compounds/toxicity , Pyrones/toxicity , Receptors, Metabotropic Glutamate/physiology , Animals , Hippocampus/physiology , Hippocampus/ultrastructure , Male , Microscopy, Electron, Transmission , Neurons/drug effects , Neurons/ultrastructure , Protein Kinase C/metabolism , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism
10.
Nat Prod Res ; 34(6): 889-892, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30445856

ABSTRACT

A search for bioactive secondary metabolites from the endophytic fungus Fusarium chlamydosporum, isolated from the root of Suaeda glauca, led to the isolation of three indole derivatives (1-3), three cyclohexadepsipeptides (4-6), and four pyrones (7-10). The structures of new (1) and known compounds (2-10) were elucidated on the basis of extensive spectroscopic analysis. All these compounds were evaluated for phytotoxic, antimicrobial activities, and brine shrimp lethality. Compound 1 showed significant phytotoxic activity against the radicle growth of Echinochloa crusgalli, even better than the positive control of 2,4-D. Cyclohexadepsipeptides (4-6) and pyrones (7-10) exhibited brine shrimp lethality, especially 4 and 7 with the LD50 values of 2.78 and 7.40 µg mL-1, respectively, better than the positive control.


Subject(s)
Echinochloa/microbiology , Fusarium/metabolism , Secondary Metabolism , Animals , Artemia/drug effects , Depsipeptides/isolation & purification , Depsipeptides/metabolism , Depsipeptides/toxicity , Echinochloa/drug effects , Endophytes , Indoles/isolation & purification , Indoles/metabolism , Indoles/toxicity , Pyrones/isolation & purification , Pyrones/metabolism , Pyrones/toxicity
11.
Environ Toxicol Chem ; 39(2): 381-395, 2020 02.
Article in English | MEDLINE | ID: mdl-31721268

ABSTRACT

Melanophores are pigmented cells that change the distribution of melanosomes, enabling animals to appear lighter or darker for camouflage, thermoregulation, and protection from ultraviolet radiation. A complex series of hormonal and neural mechanisms regulates melanophore pigment distribution, making these dynamic cells a valuable tool to screen toxicants as they rapidly respond to changes in the environment. We found that maltol, a naturally occurring flavor enhancer and fragrance agent, induces melanophore pigment aggregation in a dose-dependent manner in Xenopus laevis tadpoles. To determine if maltol affects camouflage adaptation, we placed tadpoles into maltol baths situated over either a white or a black background. Maltol induced pigment aggregation in a similar dose-dependent pattern regardless of background color. We also tested how maltol treatment compares to melatonin treatment and found that the degree of pigment aggregation induced by maltol is similar to treatment with melatonin but that maltol induces over a much longer time course. Last, maltol had no effect on mRNA expression in the brain of genes that regulate camouflage-related pigment aggregation. The present results suggest that maltol does not exert its effects via the camouflage adaptation mechanism or via melatonin-related mechanisms. These results are the first to identify a putative toxicological effect of maltol exposure in vivo and rule out several mechanisms by which maltol may exert its effects on pigment aggregation. Environ Toxicol Chem 2020;39:381-395. © 2019 SETAC.


Subject(s)
Dura Mater/drug effects , Flavoring Agents/toxicity , Larva/drug effects , Melanophores/drug effects , Pigments, Biological/metabolism , Pyrones/toxicity , Skin/drug effects , Animals , Dose-Response Relationship, Drug , Dura Mater/cytology , Dura Mater/metabolism , Flavoring Agents/metabolism , Gene Expression/drug effects , Larva/genetics , Larva/metabolism , Larva/radiation effects , Melanophores/metabolism , Melatonin/metabolism , Melatonin/pharmacology , Pigmentation/drug effects , Pyrones/metabolism , Skin/cytology , Skin/metabolism , Toxicity Tests , Ultraviolet Rays , Xenopus laevis
12.
J Med Chem ; 63(1): 283-294, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31793781

ABSTRACT

Autoimmune diseases are chronic inflammatory diseases associated with high morbidity and mortality. Treatment options for autoimmune diseases have increased over the past several decades, but they are, in general, limited in their clinical efficacy due to high toxicity and lack of selectivity. Thus, efforts must be made to identify new immunomodulatory agents that are effective through a novel mechanism to circumvent existing side effects. To define the structural requirements of subglutinols for immunomodulatory activity and to provide guiding principles on future therapeutic development, we prepared and evaluated several subglutinol analogs for their immunomodulatory activities. Our efforts identified a subglutinol analog with reduced structural complexity as a potential lead compound for future autoimmune drug development. Our study will provide an important framework for the design of potent and nontoxic immunomodulating agents derived from subglutinols.


Subject(s)
Diterpenes/therapeutic use , Immunosuppressive Agents/therapeutic use , Pyrones/therapeutic use , Animals , CD8-Positive T-Lymphocytes/drug effects , Cell Survival/drug effects , Diterpenes/chemical synthesis , Diterpenes/toxicity , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/toxicity , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Pyrones/chemical synthesis , Pyrones/toxicity , Th1 Cells/drug effects , Th17 Cells/drug effects
13.
Food Chem Toxicol ; 135: 110900, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31654710

ABSTRACT

We aimed to study the effect of vanadium(V) exposure on cell viability, nuclear DNA (nDNA) and mitochondrial DNA (mtDNA) and to elucidate if these effects can be reverted by co-exposure to V and manganese (Mn). HepG2 cells were incubated with various concentrations of bis(maltolato)oxovanadium(IV) or MnCl2 for 32 h for viability study. The higher concentrations (59   µM V, 54 nM Mn and 59   µM V+54 nM Mn) were used to study DNA damage and uptake of V and Mn. Comet assay was used for the study of nDNA damage; mtDNA damage was studied by determining deletions and number of copies of the ND1/ND4 mtDNA region. Cellular content of V and Mn was determined using ICPMS. Cellular exposure to 59   µM V decreased viability (14%) and damaged nDNA and mtDNA. This effect was partially prevented by the co-exposure to V + Mn. Exposure to V increased the cellular content of V and Mn (812.3% and 153.5%, respectively). Exposure to Mn decreased the content of V and Mn (62% and 56%, respectively). Exposure to V + Mn increased V (261%) and decreased Mn (56%) content. The positive effects on cell viability and DNA damage when incubated with V + Mn could be due to the Mn-mediated inhibition of V uptake.


Subject(s)
Cell Nucleus/drug effects , Chlorides/pharmacology , DNA Damage/drug effects , Manganese Compounds/pharmacology , Mitochondria/drug effects , Protective Agents/pharmacology , Pyrones/toxicity , Vanadates/toxicity , Cell Survival/drug effects , DNA, Mitochondrial/metabolism , Hep G2 Cells , Humans
14.
Environ Sci Pollut Res Int ; 26(12): 12071-12079, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30827024

ABSTRACT

Highly bioavailable plant phospholipid complex that can reverse aluminum maltolate (AlM)-induced toxicity is not yet reported. Hence, the present study was planned to investigate the impact of oxidative stress and apoptotic changes provoked by Al and ameliorative role of Bacopa phospholipid complex (BPC) in albino rats. The levels of antioxidant enzymes such as superoxide dismutase (SOD), catalase activity (CAT), glutathione peroxidase (GPx), and thiobarbituric acid-reactive substance (TBA-RS) were measured and immunohistochemistry analysis of apoptotic markers, Bax and Bcl-2, was done from the four brain regions such as the hippocampus, cerebral cortex, cerebellum, and medulla oblongata. The levels of antioxidant enzymes and apoptotic markers that were decreased on AlM induction showed a significant increase in their levels, almost as observed in the control, when treated with BPC and Bm. Our results indicate that both BPC and Bm showed a therapeutic effect against AlM toxicity; however, it was found that the therapeutic potential of BPC was more pronounced than Bm against AlM-induced neurotoxicity.


Subject(s)
Antioxidants/pharmacology , Brain/physiology , Organometallic Compounds/toxicity , Plant Extracts/pharmacology , Pyrones/toxicity , Animals , Bacopa/chemistry , Brain/drug effects , Catalase/metabolism , Cerebellum/drug effects , Glutathione Peroxidase/metabolism , Hippocampus/drug effects , Male , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phospholipids , Rats , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances
15.
Sci Rep ; 9(1): 2468, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30792477

ABSTRACT

We characterized the flavor chemicals in a broad sample of commercially available electronic cigarette (EC) refill fluids that were purchased in four different countries. Flavor chemicals in 277 refill fluids were identified and quantified by gas chromatography-mass spectrometry, and two commonly used flavor chemicals were tested for cytotoxicity with the MTT assay using human lung fibroblasts and epithelial cells. About 85% of the refill fluids had total flavor concentrations >1 mg/ml, and 37% were >10 mg/ml (1% by weight). Of the 155 flavor chemicals identified in the 277 refill fluids, 50 were present at ≥1 mg/ml in at least one sample and 11 were ≥10 mg/ml in 54 of the refill fluids. Sixty-one% (170 out of 277) of the samples contained nicotine, and of these, 56% had a total flavor chemical/nicotine ratio >2. Four chemicals were present in 50% (menthol, triacetin, and cinnamaldehyde) to 80% (ethyl maltol) of the samples. Some products had concentrations of menthol ("Menthol Arctic") and ethyl maltol ("No. 64") that were 30 times (menthol) and 100 times (ethyl maltol) their cytotoxic concentration. One refill fluid contained cinnamaldehyde at ~34% (343 mg/ml), more than 100,000 times its cytotoxic level. High concentrations of some flavor chemicals in EC refill fluids are potentially harmful to users, and continued absence of any regulations regarding flavor chemicals in EC fluids will likely be detrimental to human health.


Subject(s)
Flavoring Agents/analysis , Lung/cytology , Menthol/toxicity , Pyrones/toxicity , Acrolein/analogs & derivatives , Acrolein/toxicity , Cells, Cultured , Electronic Nicotine Delivery Systems , Epithelial Cells/cytology , Epithelial Cells/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Flavoring Agents/toxicity , Gas Chromatography-Mass Spectrometry , Humans , Lung/drug effects , Nicotine/toxicity
16.
Neurotox Res ; 35(4): 931-944, 2019 May.
Article in English | MEDLINE | ID: mdl-30649678

ABSTRACT

To investigate the effect of aluminum-maltolate [Al(mal)3] on the expression of ApoER2, VLDLRs, and LRP1 in PC12-ApoE4 cells. The lentiviral vector carrying human ApoE4 gene was transfected into PC12 cells; after screening with puromycin, PC12 cells carrying ApoE4 gene (PC12-ApoE4 cells) were established. After 24-h treatment with Al(mal)3, the cell survival rate was measured by CCK-8 assay. The expression of Aß40 and Aß42 was detected by ELISA assay; the expression of the APP, ApoER2, LRP1, and VLDLRs genes was detected by RT-PCR, and Western blot assay was used to detect the expression of the APP, ApoER2, LRP1, and VLDLRs proteins. Factorial experiment design was performed to analyze interaction between cell type and Al dose. Al(mal)3 treatment induced dose-dependent decreases of survival rate in the two cell groups and dose-dependent increases of Aß42 content(P < 0.05). The expressions of ApoER2, LRP1, and VLDLR proteins and their mRNA transcription decreased gradually with the increase of Al(mal)3 doses (P < 0.05), while the expression of APP protein and mRNA transcription gradually increased with the increase of Al(mal)3 doses (P < 0.05). As regard to the interaction of cell type and Al dose, the decrease of cell survival rate and the increase of the Aß42 were both statistically significant (P < 0.05). And the decrease of ApoER2 and LRP1 proteins was both statistically significant too (P < 0.05). The effect of Al(mal)3 and ApoE4 gene on the survival rate and the increase of Aß content in PC12 cells. That is to say, there is interaction between ApoE4 gene and aluminum on the Aß content, especially the change of the Aß42 content, which may be related to the down-regulation of the expression of ApoER2 and LRP1 proteins.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Organometallic Compounds/toxicity , Pyrones/toxicity , Receptors, LDL/metabolism , Animals , Apolipoproteins E/genetics , Cell Survival/drug effects , Genetic Vectors , Humans , PC12 Cells , Rats
17.
Biol Trace Elem Res ; 187(1): 243-257, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29797207

ABSTRACT

In our previous study, chromium malate is beneficial for type 2 diabetic rats in control glycometabolism and lipid metabolism. The present study was designed to observe the chronic toxicity, lipid metabolism, learning and memory ability, and related enzymes of chromium malate in rats during the year. The results showed that pathological, toxic, feces, and urine of chromium malate (at daily doses of 10.0, 15.0, and 20.0 µg Cr/kg bm) did not change measurably. Chromium malate (at daily doses of 15.0 and 20.0 µg Cr/kg bm) could significantly reduce the levels of total cholesterol (TC), LDL, and triglyceride (TG), and increase the level of HDL in male rats compared to control group and chromium picolinate group. Significant escalating trends of the escape latency and swimming speed (Morris water maze test), and the original platform quadrant stops, residence time, and swimming speed (Space exploration test) in male rats of chromium malate groups were obtained. The SOD, GSH-Px, and TChE activities of chromium malate (at daily doses of 15.0 and 20.0 µg Cr/kg bm) were enhanced significantly in male rats compared with those of the normal control group and chromium picolinate group. Glycometabolism and related enzymes had no significant changes compared to normal control group and chromium picolinate group. These results indicated that long-term chromium malate supplementation did not cause measurable toxicity at daily doses of 10.0, 15.0, and 20.0 µg Cr/kg bm and could improve dyslipidemia and learning and memory deficits.


Subject(s)
Acetylcholinesterase/metabolism , Glutathione Peroxidase/metabolism , Lipid Metabolism/drug effects , Maze Learning/drug effects , Memory/drug effects , Organometallic Compounds/administration & dosage , Organometallic Compounds/toxicity , Pyrones/administration & dosage , Pyrones/toxicity , Animals , Dietary Supplements , Female , Male , Rats , Rats, Sprague-Dawley
18.
Chem Biol Interact ; 299: 15-26, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30481499

ABSTRACT

Many reports demonstrated that aluminum maltolate (Almal) has potential toxicity to human and animal. Our study has demonstrated that Almal can induce oxidative damage and apoptosis in PC12 cells and SH-SY5Y Cells, two in vitro models of neuronal cells. Hyperforin (HF) is a well-known antioxidant, anti-inflammatory, anti-amyloid and anti-depressant compound extracted from Hypericum perforatum extract. Here, we investigated the neuroprotective effect of HF against Almal-induced neurotoxicity in cultured PC12 cells and SH-SY5Y cells, mainly caused by oxidative stress. In the present study, HF significantly inhibited the formation of reactive oxygen species (ROS), decreased the level of lipid peroxide and enhanced the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) compared with Almal group in PC12 cells and SH-SY5Y cells. Additionally, HF suppressed the reduction of the mitochondrial membrane potential (MMP), cytochrome c (Cyt-c) release, activation of caspase-3, and the down-regulation of Bcl-2 expression and up-regulation of Bax expression induced by Almal in PC12 cells and SH-SY5Y cells. In summary, HF protects PC12 cells and SH-SY5Y cells from damage induced by Almal through reducing oxidative stress and preventing of mitochondrial-mediated apoptosis.


Subject(s)
Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Phloroglucinol/analogs & derivatives , Terpenes/pharmacology , Animals , Caspase 3/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Glutathione Peroxidase/metabolism , Humans , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Organometallic Compounds/toxicity , PC12 Cells , Phloroglucinol/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrones/toxicity , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
19.
Neurotox Res ; 35(2): 318-330, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30242626

ABSTRACT

The neuroprotective role of tannoid principles of Emblica officinalis (EoT), an Indian and Chinese traditional medicinal plant against memory loss in aluminum chloride-induced in vivo model of Alzheimer's disease through attenuating AChE activity, oxidative stress, amyloid and tau toxicity, and apoptosis, was recently reported in our lab. However, to further elucidate the mechanism of neuroprotective effect of EoT, the current study was designed to evaluate endoplasmic reticulum stress-suppressing and anti-inflammatory role of EoT in PC 12 and SH-SY 5Y cells. These cells were divided into four groups: control (aluminum maltolate (Al(mal)3), EoT + Al(mal)3, and EoT alone based on 3-(4, 5-dimethyl 2-yl)-2, and 5-diphenyltetrazolium bromide (MTT) assay. EoT significantly reduced Al(mal)3-induced cell death and attenuated ROS, mitochondrial membrane dysfunction, and apoptosis (protein expressions of Bax; Bcl-2; cleaved caspases 3, 6, 9, 12; and cytochrome c) by regulating endoplasmic reticulum stress (PKR-like ER kinase (PERK), α subunit of eukaryotic initiation factor 2 (EIF2-α), C/EBP-homologous protein (CHOP), and high-mobility group box 1 protein (HMGB1)). Moreover, inflammatory response (NF-κB, IL-1ß, IL-6, and TNF-α) and Aß toxicity (Aß1-42) triggered by Al(mal)3 was significantly normalized by EoT. Our results suggested that EoT could be a possible/promising and novel therapeutic lead against Al-induced neurotoxicity. However, further extensive research is needed to prove its efficacy in clinical studies.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Neurons/drug effects , Organometallic Compounds/toxicity , Phyllanthus emblica , Plant Extracts/pharmacology , Pyrones/toxicity , Aluminum/toxicity , Animals , Apoptosis/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/physiology , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Neurons/metabolism , PC12 Cells , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Rats
20.
Bioorg Chem ; 82: 414-422, 2019 02.
Article in English | MEDLINE | ID: mdl-30428420

ABSTRACT

A series of kojic acid-derived compounds 6a-p bearing aryloxymethyl-1H-1,2,3-triazol-1-yl moiety were designed by modifying primary alcoholic group of kojic acid as tyrosinase inhibitors. The target compounds 6a-p were synthesized via click reaction. All compounds showed very potent anti-tyrosinase activity (IC50s = 0.06-6.80 µM), being superior to reference drug, kojic acid. In particular, the naphthyloxy analogs 6o and 6p were found to be 31-155 times more potent than kojic acid. The metal-binding study of selected compound 6o revealed that the prototype compound possesses metal-chelating ability, particularly with Cu2+ ions. The promising compounds 6o and 6p had acceptable safety profile as demonstrated by cytotoxicity assay against melanoma (B16) cell line and Human Foreskin Fibroblast (HFF) cells.


Subject(s)
Enzyme Inhibitors/pharmacology , Monophenol Monooxygenase/antagonists & inhibitors , Pyrones/pharmacology , Triazoles/pharmacology , Animals , Catalytic Domain , Cell Line, Tumor , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Chelating Agents/pharmacology , Chelating Agents/toxicity , Click Chemistry , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Humans , Metals/chemistry , Mice , Molecular Docking Simulation , Molecular Structure , Monophenol Monooxygenase/chemistry , Pyrones/chemical synthesis , Pyrones/chemistry , Pyrones/toxicity , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL