Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
Pharmacol Rep ; 74(5): 890-908, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36129673

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is considered the most common cause of dementia among the elderly. One of the modifiable causes of AD is neuroinflammation. The current study aimed to investigate the influence of new tricyclic 1,2-thiazine derivatives on in vitro model of neuroinflammation and their ability to cross the blood-brain barrier (BBB). METHODS: The potential anti-inflammatory effect of new tricyclic 1,2-thiazine derivatives (TP1, TP4, TP5, TP6, TP7, TP8, TP9, TP10) was assessed in SH-SY5Y cells differentiated to the neuron-like phenotype incubated with bacterial lipopolysaccharide (5 or 50 µg/ml) or THP-1 microglial cell culture supernatant using MTT, DCF-DA, Griess, and fast halo (FHA) assays. Additionally, for cultures preincubated with 50 µg/ml lipopolysaccharide (LPS), a cyclooxygenase (COX) activity assay was performed. Finally, the potential ability of tested compounds to cross the BBB was evaluated by computational studies. Molecular docking was performed with the TLR4/MD-2 complex to assess the possibility of binding the tested compounds in the LPS binding pocket. Prediction of ADMET parameters (absorption, distribution, metabolism, excretion and toxicity) was also conducted. RESULTS: The unfavorable effect of LPS and co-culture with THP-1 cells on neuronal cell viability was counteracted with TP1 and TP4 in all tested concentrations. Tested compounds reduced the oxidative and nitrosative stress induced by both LPS and microglia activation and also reduced DNA damage. Furthermore, new derivatives inhibited total COX activity. Additionally, new compounds would cross the BBB with high probability and reach concentrations in the brain not lower than in the serum. The binding affinity at the TLR4/MD-2 complex binding site of TP4 and TP8 compounds is similar to that of the drug donepezil used in Alzheimer's disease. The ADMET analysis showed that the tested compounds should not be toxic and should show high intestinal absorption. CONCLUSIONS: New tricyclic 1,2-thiazine derivatives exert a neuroregenerative effect in the neuroinflammation model, presumably via their inhibitory influence on COX activity and reduction of oxidative and nitrosative stress.


Subject(s)
Alzheimer Disease , Neuroblastoma , Thiazines , Humans , Lipopolysaccharides/toxicity , Microglia , Toll-Like Receptor 4/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Molecular Docking Simulation , Coculture Techniques , Neuroinflammatory Diseases , Donepezil/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Neuroblastoma/metabolism , Anti-Inflammatory Agents/pharmacology , Thiazines/adverse effects , Thiazines/metabolism
2.
Parasit Vectors ; 14(1): 450, 2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34488852

ABSTRACT

BACKGROUND: Alveolar echinococcosis (AE) is a chronic zoonosis caused by the larval form of Echinococcus multilocularis (E. multilocularis). Current chemotherapy against AE has relied on albendazole and mebendazole, which only exhibit parasitostatic and not parasiticidal efficacy. Therefore, novel compounds for the treatment of this disease are needed. METHODS: Phosphoglucose isomerase (PGI) assays were used for compound screening of seven neonicotinoids. The anti-parasitic effects of thiacloprid were then evaluated on E. multilocularis metacestode vesicles, germinal cells and protoscoleces in vitro. Human foreskin fibroblasts (HFF) and Reuber rat hepatoma (RH) cells were used to assess cytotoxicity. Glucose consumption in E. multilocularis protoscoleces and germinal cells was assessed by measuring uptake of 2-deoxyglucose (2-DG). Molecular docking was used to evaluate the potential binding sites of thiacloprid to acetylcholine receptors. In vivo efficacy of thiacloprid was evaluated in mice by secondary infection with E. multilocularis. In addition, ELISA and flow cytometry were used to evaluate the effects of cytokines and T lymphocyte subsets after thiacloprid treatment. Furthermore, collagen deposition and degradation in the host lesion microenvironment were evaluated. RESULTS: We found that thiacloprid is the most promising compound, with an IC50 of 4.54 ± 1.10 µM and 2.89 ± 0.34 µM, respectively, against in vitro-cultured E. multilocularis metacestodes and germinal cells. Thiacloprid was less toxic for HFF and RH mammalian cell lines than for metacestodes. In addition, thiacloprid inhibited the acetylcholinesterase activity in protoscoleces, metacestodes and germinal cells. Thiacloprid inhibited glucose consumption by protoscoleces and germinal cells. Subsequently, transmission electron microscopy revealed that treatment with thiacloprid damaged the germinal layer. In vivo, metacestode weight was significantly reduced following oral administration of thiacloprid at 15 and 30 mg/kg. The level of CD4+ T lymphocytes in metacestodes and spleen increased after thiacloprid treatment. Anti-echinococcosis-related cytokines (IL-2, IL-4, IL-10) were significantly increased. Furthermore, thiacloprid inhibited the expression of matrix metalloproteinases (MMPs 1, 3, 9, 13) and promoted collagen deposition in the host lesion microenvironment. CONCLUSIONS: The results demonstrated that thiacloprid had parasiticidal activity against E. multilocularis in vitro and in vivo, and could be used as a novel lead compound for the treatment of AE.


Subject(s)
Anthelmintics/pharmacology , Anthelmintics/therapeutic use , Echinococcus multilocularis/drug effects , Neonicotinoids/pharmacology , Neonicotinoids/therapeutic use , Thiazines/pharmacology , Thiazines/therapeutic use , Animals , Anthelmintics/metabolism , Echinococcosis/drug therapy , Female , Fibroblasts/drug effects , Fibroblasts/parasitology , Foreskin/cytology , Humans , Inhibitory Concentration 50 , Male , Mice, Inbred BALB C , Molecular Docking Simulation , Neonicotinoids/metabolism , Receptors, Cholinergic/metabolism , Specific Pathogen-Free Organisms , Thiazines/metabolism
3.
Molecules ; 26(16)2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34443360

ABSTRACT

Plasma proteins play a fundamental role in living organisms. They participate in the transport of endogenous and exogenous substances, especially drugs. 5-alkyl-12(H)-quino[3,4-b][1,4]benzothiazinium salts, have been synthesized as potential anticancer substances used for cancer treatment. Most anticancer substances generate a toxic effect on the human body. In order to check the toxicity and therapeutic dosage of these chemicals, the study of ligand binding to plasma proteins is very relevant. The present work presents the first comparative analysis of the binding of one of the 5-alkyl-12(H)-quino[3,4-b][1,4]benzothiazinium derivatives (Salt1) with human serum albumin (HSA), α-1-acid glycoprotein (AGP) and human gamma globulin (HGG), assessed using fluorescence, UV-Vis and CD spectroscopy. In order to mimic in vivo ligand-protein binding, control normal serum (CNS) was used. Based on the obtained data, the Salt1 binding sites in the tertiary structure of all plasma proteins and control normal serum were identified. Both the association constants (Ka) and the number of binding site classes (n) were calculated using the Klotz method. The strongest complex formed was Salt1-AGPcomplex (Ka = 7.35·104 and 7.86·104 mol·L-1 at excitation wavelengths λex of 275 and 295 nm, respectively). Lower values were obtained for Salt1-HSAcomplex (Ka = 2.45·104 and 2.71·104 mol·L-1) and Salt1-HGGcomplex (Ka = 1.41·104 and 1.33·104 mol·L-1) at excitation wavelengths λex of 275 and 295 nm, respectively, which is a positive phenomenon and contributes to the prolonged action of the drug. Salt1 probably binds to the HSA molecule in Sudlow sites I and II; for the remaining plasma proteins studied, only one binding site was observed. Moreover, using circular dichroism (CD), fluorescence and UV-Vis spectroscopy, no effect on the secondary and tertiary structures of proteins in the absence or presence of Salt1 has been demonstrated. Despite the fact that the conducted studies are basic, from the scientific point of view they are novel and encourage further in vitro and in vivo investigations. As a next part of the study (Part 2), the second new synthetized quinobenzothiazine derivative (Salt2) will be analyzed and published.


Subject(s)
Blood Proteins/metabolism , Spectrum Analysis , Thiazines/chemistry , Thiazines/metabolism , Blood Proteins/chemistry , Humans , Molecular Docking Simulation , Protein Binding , Protein Conformation , Thermodynamics
4.
Toxicol Lett ; 338: 10-20, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33253783

ABSTRACT

Meloxicam is a thiazole-containing NSAID that was approved for marketing with favorable clinical outcomes despite being structurally similar to the hepatotoxic sudoxicam. Introduction of a single methyl group on the thiazole results in an overall lower toxic risk, yet the group's impact on P450 isozyme bioactivation is unclear. Through analytical methods, we used inhibitor phenotyping and recombinant P450s to identify contributing P450s, and then measured steady-state kinetics for bioactivation of sudoxicam and meloxicam by the recombinant P450s to determine relative efficiencies. Experiments showed that CYP2C8, 2C19, and 3A4 catalyze sudoxicam bioactivation, and CYP1A2 catalyzes meloxicam bioactivation, indicating that the methyl group not only impacts enzyme affinity for the drugs, but also alters which isozymes catalyze the metabolic pathways. Scaling of relative P450 efficiencies based on average liver concentration revealed that CYP2C8 dominates the sudoxicam bioactivation pathway and CYP2C9 dominates meloxicam detoxification. Dominant P450s were applied for an informatics assessment of electronic health records to identify potential correlations between meloxicam drug-drug interactions and drug-induced liver injury. Overall, our findings provide a cautionary tale on assumed impacts of even simple structural modifications on drug bioactivation while also revealing specific targets for clinical investigations of predictive factors that determine meloxicam-induced idiosyncratic liver injury.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP2C9/metabolism , Meloxicam/metabolism , Microsomes, Liver/enzymology , Thiazines/metabolism , Activation, Metabolic , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Chemical and Drug Induced Liver Injury/etiology , Data Mining , Deep Learning , Drug Interactions , Electronic Health Records , Female , Humans , Inactivation, Metabolic , Kinetics , Male , Meloxicam/toxicity , Middle Aged , Substrate Specificity , Thiazines/toxicity
5.
J Enzyme Inhib Med Chem ; 35(1): 1891-1905, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33003975

ABSTRACT

A large library of saccharin and acesulfame derivatives has been synthesised and evaluated against four isoforms of human carbonic anhydrase, the two off-targets hCA I/II and the tumour related isoforms hCA IX/XII. Different strategies of scaffold modification have been attempted on both saccharin as well as acesulfame core leading to the obtainment of 60 compounds. Some of them exhibited inhibitory activity in the nanomolar range, albeit some of the performed changes led to either micromolar activity or to its absence, against hCA IX/XII. Molecular modelling studies focused the attention on the binding mode of these compounds to the enzyme. The proposed inhibition mechanism is the anchoring to zinc-bound water molecule. Docking studies along with molecular dynamics also underlined the importance of the compounds flexibility (e.g. achieved through the insertion of methylene group) which favoured potent and selective hCA inhibition.


Subject(s)
Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrases/metabolism , Saccharin/chemical synthesis , Sweetening Agents/chemical synthesis , Thiazines/chemical synthesis , Carbonic Anhydrase Inhibitors/metabolism , Carbonic Anhydrase Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Saccharin/metabolism , Saccharin/pharmacology , Structure-Activity Relationship , Sweetening Agents/metabolism , Sweetening Agents/pharmacology , Thiazines/metabolism , Thiazines/pharmacology , Triazoles/chemistry , Zinc/chemistry
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 237: 118410, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32361316

ABSTRACT

In this work, the binding interaction of an artificial sweetener, acesulfame (ACS) with human serum albumin (HSA) are investigated at the molecular level by using spectral methods and molecular modeling. ACS has the ability to induce static quenching of the intrinsic fluorescence of HSA by a complex formed between HSA and ACS through weak multi-noncovalent forces including hydrophobic, hydrogen bond and van der Waals forces. ACS enters subdomain IIA of HSA to induce the tertiary structure changes of HSA and decreased the hydrophobicity of protein. In addition, ACS binding does not obviously alter the secondary structure of HSA. This study is hoped to provide some crucial information for further investigations of the biosafety of sweetener.


Subject(s)
Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Thiazines/chemistry , Thiazines/metabolism , Binding Sites , Circular Dichroism , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Sweetening Agents/chemistry , Sweetening Agents/metabolism
7.
Toxicology ; 440: 152478, 2020 07.
Article in English | MEDLINE | ID: mdl-32437779

ABSTRACT

Thiazoles are biologically active aromatic heterocyclic rings occurring frequently in natural products and drugs. These molecules undergo typically harmless elimination; however, a hepatotoxic response can occur due to multistep bioactivation of the thiazole to generate a reactive thioamide. A basis for those differences in outcomes remains unknown. A textbook example is the high hepatotoxicity observed for sudoxicam in contrast to the relative safe use and marketability of meloxicam, which differs in structure from sudoxicam by the addition of a single methyl group. Both drugs undergo bioactivation, but meloxicam exhibits an additional detoxification pathway due to hydroxylation of the methyl group. We hypothesized that thiazole bioactivation efficiency is similar between sudoxicam and meloxicam due to the methyl group being a weak electron donator, and thus, the relevance of bioactivation depends on the competing detoxification pathway. For a rapid analysis, we modeled epoxidation of sudoxicam derivatives to investigate the impact of substituents on thiazole bioactivation. As expected, electron donating groups increased the likelihood for epoxidation with a minimal effect for the methyl group, but model predictions did not extrapolate well among all types of substituents. Through analytical methods, we measured steady-state kinetics for metabolic bioactivation of sudoxicam and meloxicam by human liver microsomes. Sudoxicam bioactivation was 6-fold more efficient than that for meloxicam, yet meloxicam showed a 6-fold higher efficiency of detoxification than bioactivation. Overall, sudoxicam bioactivation was 15-fold more likely than meloxicam considering all metabolic clearance pathways. Kinetic differences likely arise from different enzymes catalyzing respective metabolic pathways based on phenotyping studies. Rather than simply providing an alternative detoxification pathway, the meloxicam methyl group suppressed the bioactivation reaction. These findings indicate the impact of thiazole substituents on bioactivation is more complex than previously thought and likely contributes to the unpredictability of their toxic potential.


Subject(s)
Meloxicam/metabolism , Thiazines/metabolism , Activation, Metabolic , Biotransformation , Chemical and Drug Induced Liver Injury/metabolism , Electrons , Epoxy Compounds/metabolism , Humans , Hydroxylation , In Vitro Techniques , Kinetics , Metabolic Networks and Pathways/drug effects , Microsomes, Liver/metabolism , Thiazoles/metabolism
8.
Nature ; 580(7804): 511-516, 2020 04.
Article in English | MEDLINE | ID: mdl-32322067

ABSTRACT

The taste of sugar is one of the most basic sensory percepts for humans and other animals. Animals can develop a strong preference for sugar even if they lack sweet taste receptors, indicating a mechanism independent of taste1-3. Here we examined the neural basis for sugar preference and demonstrate that a population of neurons in the vagal ganglia and brainstem are activated via the gut-brain axis to create preference for sugar. These neurons are stimulated in response to sugar but not artificial sweeteners, and are activated by direct delivery of sugar to the gut. Using functional imaging we monitored activity of the gut-brain axis, and identified the vagal neurons activated by intestinal delivery of glucose. Next, we engineered mice in which synaptic activity in this gut-to-brain circuit was genetically silenced, and prevented the development of behavioural preference for sugar. Moreover, we show that co-opting this circuit by chemogenetic activation can create preferences to otherwise less-preferred stimuli. Together, these findings reveal a gut-to-brain post-ingestive sugar-sensing pathway critical for the development of sugar preference. In addition, they explain the neural basis for differences in the behavioural effects of sweeteners versus sugar, and uncover an essential circuit underlying the highly appetitive effects of sugar.


Subject(s)
Brain/physiology , Choice Behavior/physiology , Dietary Sugars/metabolism , Food Preferences/physiology , Glucose/metabolism , Intestines/physiology , Animals , Brain/cytology , Dietary Sugars/chemistry , Glucose/analogs & derivatives , Glucose/chemistry , Male , Methylglucosides/chemistry , Methylglucosides/metabolism , Mice , Mice, Inbred C57BL , Neurons/physiology , Taste/physiology , Thiazines/metabolism , Water/metabolism
9.
J Med Chem ; 62(20): 9331-9337, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31549838

ABSTRACT

Genetic evidence points to deposition of amyloid-ß (Aß) as a causal factor for Alzheimer's disease. Aß generation is initiated when ß-secretase (BACE1) cleaves the amyloid precursor protein. Starting with an oxazine lead 1, we describe the discovery of a thiazine-based BACE1 inhibitor 5 with robust Aß reduction in vivo at low concentrations, leading to a low projected human dose of 14 mg/day where 5 achieved sustained Aß reduction of 80% at trough level.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Protease Inhibitors/chemistry , Thiazines/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cytochrome P-450 CYP2C9/chemistry , Cytochrome P-450 CYP2C9/metabolism , Dogs , Drug Evaluation, Preclinical , Female , Half-Life , Haplorhini , Heart/drug effects , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Thiazines/metabolism , Thiazines/pharmacology
10.
J Obstet Gynaecol Res ; 45(11): 2228-2236, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31502345

ABSTRACT

AIM: To identify potential biomarkers for tumor progression and patient outcomes in cervical squamous cell carcinoma. METHODS: We examined the expressions of CK7 and CK17 as potential markers of the squamo-columnar junction, and podoplanin as a basal cell marker using surgical and biopsy samples of patients in grade 3 cervical intraepithelial neoplasia (n = 30), operable invasive carcinoma (OP group, n = 53) and inoperable invasive carcinoma before radiotherapy and/or chemotherapy (RC group, n = 76). RESULTS: The positive rates of CK7 and podoplanin in invasive carcinoma were significantly lower than those in grade 3 cervical intraepithelial neoplasia (P = 0.001, P < 0.0001). The positive rates of CK7 and podoplanin in the RC group were significantly lower than those in the OP group (P < 0.0001, P = 0.04), while CK17 expression showed significantly higher positivity in the RC group than in the OP group (P < 0.0001). Negative CK7 expression showed a potential impact on overall survival in early-stage patients. In the RC group, the prevalence of cases with post-therapeutic residual carcinoma cells was higher in the CK7-negative group than in the positive group (P = 0.003). We found that decreased expression of CK7 could be a prognostic factor in early-stage cervical cancer patients. CONCLUSION: This result may provide strategies and suggestions for new treatment options and follow-up practices in managing patients with cervical cancer.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Keratin-7/metabolism , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Disease Progression , Female , Humans , Imines/metabolism , Immunohistochemistry , Membrane Glycoproteins/metabolism , Middle Aged , Neoplasm Staging , Prognosis , Thiazines/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Dysplasia/pathology
11.
J Biochem Mol Toxicol ; 33(10): e22379, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31364238

ABSTRACT

The interactions of six neonicotinoid pesticides and one neonicotinoid metabolite with drug transporters have been characterized in vitro. Acetamiprid, clothianidin, imidacloprid, nitenpyram, thiacloprid and its metabolite thiacloprid amide, and thiamethoxam, each used at 100 µM, did not impair activity of the efflux pumps P-glycoprotein, multidrug resistance-associated proteins, and breast cancer resistance protein. They also did not inhibit that of the uptake transporters OATP1B1, OATP1B3, OAT4, and MATE1, whereas that of OATP2B1, OAT1, and MATE2-K was affected by only one of the seven neonicotinoids. Activity of OCT1 was moderately stimulated (up to 1.5-fold) by several neonicotinoids. By contrast, that of OAT3 and OCT2 was inhibited by most (OAT3), if not all (OCT2), neonicotinoids, with IC50 values in the 20 to 60 µM range for thiacloprid, likely not relevant to environmental exposure. Thiacloprid was moreover not transported by OAT3 and OCT2. Overall, these data suggest that neonicotinoid pesticides rather poorly interact with drug transporter activities.


Subject(s)
Insecticides/pharmacology , Neonicotinoids/pharmacology , Receptors, Cell Surface/drug effects , ATP-Binding Cassette Transporters/metabolism , Cell Line, Tumor , Drug Interactions , Humans , Insecticides/pharmacokinetics , Neonicotinoids/metabolism , Neonicotinoids/pharmacokinetics , Thiazines/metabolism
12.
Insect Biochem Mol Biol ; 111: 103171, 2019 08.
Article in English | MEDLINE | ID: mdl-31136794

ABSTRACT

Recent work has shown that two bumblebee (Bombus terrestris) cytochrome P450s of the CYP9Q subfamily, CYP9Q4 and CYP9Q5, are important biochemical determinants of sensitivity to neonicotinoid insecticides. Here, we report the characterisation of a third P450 gene CYP9Q6, previously mis-annotated in the genome of B. terrestris, encoding an enzyme that metabolises the N-cyanoamidine neonicotinoids thiacloprid and acetamiprid with high efficiency. The genomic location and complete ORF of CYP9Q6 was corroborated by PCR and its metabolic activity characterised in vitro by expression in an insect cell line. CYP9Q6 metabolises both thiacloprid and acetamiprid more rapidly than the previously reported CYP9Q4 and CYP9Q5. We further demonstrate a direct, in vivo correlation between the expression of the CYP9Q6 enzyme in transgenic Drosophila melanogaster and an increased tolerance to thiacloprid and acetamiprid. We conclude that CYP9Q6 is an efficient metaboliser of N-cyanoamidine neonicotinoids and likely plays a key role in the high tolerance of B. terrestris to these insecticides.


Subject(s)
Bees/enzymology , Cytochrome P-450 Enzyme System/metabolism , Neonicotinoids/metabolism , Thiazines/metabolism , Animals , Animals, Genetically Modified , Bees/genetics , Bees/metabolism , Cell Line , Cytochrome P-450 Enzyme System/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Insecticide Resistance/genetics , Moths
13.
Chemosphere ; 226: 645-650, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30959449

ABSTRACT

Encapsulating fungicides and/or insecticides in film-coatings applied to agronomic seeds has become a widely accepted method for enhancing seed germination and overall seedling health by protecting against many diseases and early-season insect pests. Despite advancements in seed film-coating technologies, abrasion of the seed coating can occur during handling and mechanical planting operations, resulting in variable amounts of detached fragments entering the soil. The present study investigated the degradation in soil of these plastic-like, small-sized fragments, referred to here as microplastic coating fragments. Degradation of microplastic coating fragments in soil was found to be highly variable. The lowest degradation rate (≤48 days) was observed in fragments detached from seeds coated with a commercial polymer mixture, while fragments from a biodegradable plastic formulation degraded completely within 32 days. When spores of the plant growth-promoting bacterium, Bacillus subtilis, were incorporated into the bioplastic, degradation was even more rapid (≤24 days). The fragment degradation rate was unaffected by incorporating two commonly used neonicotinoid insecticides, imidacloprid or thiacloprid, into either coating formulations, but insecticide dissipation rates in soil were more rapid when added associated with seed coating fragments than when spiked in directly. Half-lives of these two insecticides were reduced by up to 27% in fragments from bioplastic-coated seeds. These results are consistent with variable and not easily predicted soil degradation rates for seed coating fragments, with enhanced dissipation of coating-entrapped pesticides and with a higher degradation rate for biodegradable seed coating incorporating selected microbial strains.


Subject(s)
Fungicides, Industrial/metabolism , Insecticides/metabolism , Pesticides/metabolism , Plastics/metabolism , Seeds/chemistry , Soil Pollutants/metabolism , Soil/chemistry , Bacillus subtilis/metabolism , Insecticides/analysis , Neonicotinoids/metabolism , Nitro Compounds/metabolism , Seedlings/metabolism , Soil Pollutants/analysis , Thiazines/metabolism
14.
Int J Food Sci Nutr ; 70(7): 894-900, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30892106

ABSTRACT

Substituting sugar-sweetened for artificially sweetened beverages may reduce energy intakes. This study aims to ascertain the acute glycaemic effects of the NNS aspartame and acesulfame-K in UK diet-cola (DC). Ten healthy participants attended the laboratory fasted on three occasions. Individuals drank (1) 25 g glucose in 125 mL water + 236 mL water, (2) 25 g glucose in 125 mL water with 236 mL DC and (3) 236 mL sucrose-sweetened cola with 125 mL water. Blood (glucose) was measured pre-test and every 15 minutes over a 120-minute period using portable glucometers. The glucose-control and glucose + DC elicited similar blood glucose rises above pre-prandial levels. Sucrose-sweetened cola showed a non-significant lower rise in postprandial glycaemia, exhibiting the lowest glycaemic index (GI) (77.0 ± 9.1). GI of glucose (100.0 ± 15.2) and glucose + DC (104.3 ± 8.5) was similar and a one-way repeated-measures ANOVA showed no significant differences in glycaemic response between test drinks (F(2,29) = 1.68, p > .05). Results demonstrate the glycaemic inactivity of non-nutritive sweeteners.


Subject(s)
Aspartame/pharmacology , Blood Glucose/drug effects , Cola , Diet , Non-Nutritive Sweeteners/pharmacology , Thiazines/metabolism , Adult , Cross-Over Studies , Energy Intake/drug effects , Female , Humans , Male , Postprandial Period , Sucrose , Sweetening Agents/pharmacology , United Kingdom , Young Adult
15.
Biochim Biophys Acta Gen Subj ; 1863(3): 577-585, 2019 03.
Article in English | MEDLINE | ID: mdl-30611847

ABSTRACT

BACKGROUND: HDAC8 is an established target for T-cell lymphoma and childhood neuroblastoma. Benzothiazine-imines are promising HDAC8 inhibitors with unknown binding mechanism lacking a usual zinc binding group. METHODS: In this study high-resolution and quantitative HPLC-coupled ESI-MS/MS techniques are combined with crystal structure determination and a variety of biochemical and computational methods to elucidate the reaction mechanism between benzothiazine-imine 1 and HDAC8. RESULTS: 1) 1 is a covalent inhibitor of HDAC8; 2) inhibition is reversible in the presence of reducing agents; 3) C153 in the active site and C102 are involved in the inhibition mechanism; 4) 1 modifies various cysteines in HDAC8 forming either thiocyanates or mixed disulfides with 3; 5) 1 and 5 dock in close proximity to C153 within the active site. This is supposed to accelerate covalent inactivation particularly in HDAC8 and suggested as major determinant for the observed nanomolar potency and selectivity of 1. CONCLUSIONS: 1 and its analogs are interesting model compounds but unsuitable for therapeutic treatment due to their high unselective reactivity towards thiol groups. However, the postulated preceding non-covalent binding mode of 1 opens a door to optimized next generation compounds that combine potent and selective non-covalent recognition with low reactivity towards C153 at the active site of HDAC8. GENERAL SIGNIFICANCE: 1 represents a completely new class of inhibitors for HDAC8. Initial non-covalent interaction at the bottom of the active site is suggested to be the key for its selectivity. Further optimization of non-covalent interaction and thiol-reactivity provides opportunities to develop therapeutic useful covalent HDAC8 inhibitors.


Subject(s)
Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Pyrimidines/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Binding Sites/genetics , Catalytic Domain/genetics , Drug Design , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Humans , Imines/chemistry , Imines/metabolism , Imines/pharmacology , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Pyrimidines/chemistry , Pyrimidines/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Structure-Activity Relationship , Thiazines/chemistry , Thiazines/metabolism , Thiazines/pharmacology
16.
Sci Total Environ ; 647: 772-784, 2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30096667

ABSTRACT

Acesulfame potassium (ACS) is a widely-used sweetener worldwide. Its presence has been demonstrated in diverse bodies of water. However, the deleterious effects this causes in aquatic organisms has not yet been identified, which generates controversy concerning the risks that ACS represents after its disposal into the bodies of water. Thus, the objective of this work was to evaluate if the exposure of ACS in environmentally-relevant concentrations was capable of producing oxidative stress in blood, liver, gill, brain and muscle of common carp (Cyprinus carpio). With this finality, the carp were exposed to two environmentally-relevant concentrations (0.05 and 149 µg L-1) at different exposure times (12, 24, 48, 72 and 96 h), having controls in the same conditions for each exposure time. Posteriorly, the following biomarkers of damage were evaluated: hydroperoxide content (HPC), level of lipoperoxidation (LPX) and protein carbonyl content (PCC), as well as the activity of antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT). The results showed that ACS produces significant increase in damage biomarkers evaluated in all organs, mainly in gill, brain and muscle, as well as significant changes in the activity of antioxidant enzymes in the same organs. Thus, it is concluded that ACS is capable of producing oxidative stress in common carp (Cyprinus carpio).


Subject(s)
Carps/metabolism , Oxidative Stress/physiology , Thiazines/toxicity , Water Pollutants, Chemical/toxicity , Animals , Antioxidants/metabolism , Biomarkers/metabolism , Catalase/metabolism , Lipid Peroxidation , Liver , Protein Carbonylation , Superoxide Dismutase/metabolism , Thiazines/metabolism
17.
J Nucl Med ; 60(7): 992-997, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30530832

ABSTRACT

ß-secretase 1 (BACE1) is a key enzyme in the generation of ß-amyloid, which is accumulated in the brain of Alzheimer disease patients. PF-06684511 was identified as a candidate PET ligand for imaging BACE1 in the brain and showed high specific binding in an initial assessment in a nonhuman primate (NHP) PET study using 18F-PF-06684511. In this effort, we aimed to quantitatively evaluate the regional brain distribution of 18F-PF-06684511 in NHPs under baseline and blocking conditions and to assess the target occupancy of BACE1 inhibitors. In addition, NHP whole-body PET measurements were performed to estimate the effective radiation dose. Methods: Initial brain PET measurements were performed at baseline and after oral administration of 5 mg/kg of LY2886721, a BACE1 inhibitor, in 2 cynomolgus monkeys. Kinetic analysis was performed with the radiometabolite-corrected plasma input function. In addition, a wide dose range of another BACE1 inhibitor, PF-06663195, was examined to investigate the relationship between the brain target occupancy and plasma concentration of the drug. Finally, the effective radiation dose of 18F-PF-06684511 was estimated on the basis of the whole-body PET measurements in NHPs. Results: Radiolabeling was accomplished successfully with an incorporation radiochemical yield of 4%-12% (decay-corrected) from 18F ion. The radiochemical purity was greater than 99%. The whole-brain uptake of 18F-PF-06684511 peaked (∼220% SUV) at approximately 20 min and decreased thereafter (∼100% SUV at 180 min). A 2-tissue-compartment model described the time-activity curves well. Pretreatment with LY2886721 reduced the total distribution volume of 18F-PF-06684511 by 48%-80% depending on the brain region, confirming its in vivo specificity. BACE1 occupancy of PF-06663195, estimated using the Lassen occupancy plot, showed a dose-dependent increase. The effective dose of 18F-PF-06684511 was 0.043 mSv/MBq for humans. Conclusion: 18F-PF-06684511 is the first successful PET radioligand for BACE1 brain imaging that demonstrates favorable in vivo binding and brain kinetics in NHPs.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Brain/diagnostic imaging , Brain/metabolism , Fluorine Radioisotopes/metabolism , Positron-Emission Tomography , Pyrazines/metabolism , Thiazines/metabolism , Animals , Female , Kinetics , Ligands , Macaca fascicularis , Male , Models, Biological , Radiochemistry , Whole Body Imaging
18.
Acta Biochim Pol ; 65(2): 185-191, 2018.
Article in English | MEDLINE | ID: mdl-29796443

ABSTRACT

The purpose of the present work was to assess the ability of five new oxicam analogues to interact with the lipid bilayers. To characterize the interaction of newly synthesized NSAIDs (non-steroidal anti-inflammatory drugs) analogues with DPPC lipid bilayers the two following techniques were applied - differential scanning calorimetry (DSC) and fluorescence spectroscopy. The results obtained by these experimental approaches show that new oxicams analogues interact with the lipid model membranes under consideration. As demonstrated both in calorimetric and spectroscopic studies, the greatest influence on the thermotropic properties of the lipid membrane and on the quenching of fluorescence of Laurdan and Prodan was exerted by a derivative named PR47 containing in its structure a two-carbon aliphatic linker with a carbonyl group, as well as bromine and trifluoromethyl substituents.


Subject(s)
Lipid Bilayers/metabolism , Thiazines/metabolism , 1,2-Dipalmitoylphosphatidylcholine/metabolism , 2-Naphthylamine/analogs & derivatives , 2-Naphthylamine/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Calorimetry/methods , Laurates/chemistry , Lipid Bilayers/chemistry , Molecular Structure , Piroxicam , Spectrometry, Fluorescence/methods , Thiazines/chemical synthesis , Thiazines/chemistry
19.
J Agric Food Chem ; 66(19): 4842-4852, 2018 May 16.
Article in English | MEDLINE | ID: mdl-29665689

ABSTRACT

The noncaloric sweeteners (NCSs) cyclamate (Cycl) and acesulfame K (AceK) are widely added to foods and beverages. Little is known about their impact on gastric acid secretion (GAS), which is stimulated by dietary protein and bitter-tasting compounds. Since Cycl and AceK have a bitter off taste in addition to their sweet taste, we hypothesized they modulate mechanisms of GAS in human gastric parietal cells (HGT-1). HGT-1 cells were exposed to sweet tastants (50 mM of glucose, d-threonine, Cycl, or AceK) and analyzed for their intracellular pH index (IPX), as an indicator of proton secretion by means of a pH-sensitive dye, and for mRNA levels of GAS-associated genes by RT-qPCR. Since the NCSs act via the sweet taste-sensing receptor T1R2/T1R3, mRNA expression of the corresponding genes was analyzed in addition to immunocytochemical localization of the T1R2 and T1R3 receptor proteins. Exposure of HGT-1 cells to AceK or d-threonine increased the IPX to 0.60 ± 0.05 and 0.80 ± 0.04 ( P ≤ 0.05), respectively, thereby indicating a reduced secretion of protons, whereas Cycl demonstrated the opposite effect with IPX values of -0.69 ± 0.08 ( P ≤ 0.05) compared to controls (IPX = 0). Cotreatment with the T1R3-inhibitor lactisole as well as a TAS1R3 siRNA knock-down approach reduced the impact of Cycl, AceK, and d-thr on proton release ( P ≤ 0.05), whereas cotreatment with 10 mM glucose enhanced the NCS-induced effect ( P ≤ 0.05). Overall, we demonstrated Cycl and AceK as modulators of proton secretion in HGT-1 cells and identified T1R3 as a key element in this response.


Subject(s)
Cyclamates/metabolism , Gastric Acid/metabolism , Receptors, G-Protein-Coupled/metabolism , Sweetening Agents/metabolism , Thiazines/metabolism , Cell Line, Tumor , Humans , Receptors, G-Protein-Coupled/genetics
20.
Chemosphere ; 200: 603-611, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29510368

ABSTRACT

The accumulation of pesticides in vegetables may have serious effects on human health and ecosystems via food chains; therefore, it is of great importance to investigate the uptake and accumulation behaviours of pesticides in vegetable tissues. In the present study, the uptake, translocation and accumulation of five neonicotinoids, thiamethoxam (THIM), clothianidin (CLO), thiacloprid (THID), acetamiprid (ACE) and dinotefuran (DIN), in komatsuna (Brassica rapa var. perviridis, a vegetable) were investigated. The concentrations of neonicotinoids in vegetable tissues ranged from 0.068 ±â€¯0.002 to 29.6 ±â€¯2.5 mg/kg. During the cultivation (except for the first day), the concentration of each neonicotinoid in shoots was the highest, followed by roots and the soil. The concentrating of neonicotinoids from the soil to roots followed the order of THIM > CLO > THID > DIN > ACE, while the order of the ability of translocation neonicotinoids from roots to shoots was the just opposite. The difference in uptake and translocation behaviours of the test neonicotinoids seems to be not correlated with the octanol/water partition coefficient (logKow), water solubility or dissociation constant (pKa), but significantly correlated with molecular weight. In addition, a greater concentration of the THIM-metabolite clothianidin (M-CLO) was detected in vegetable shoots than in roots and the soil.


Subject(s)
Brassica rapa/chemistry , Brassica rapa/metabolism , Neonicotinoids/analysis , Neonicotinoids/metabolism , Pesticides/analysis , Pesticides/metabolism , Soil Pollutants/metabolism , Biological Transport , Environmental Pollution , Guanidines/analysis , Guanidines/metabolism , Humans , Nitro Compounds/analysis , Nitro Compounds/metabolism , Oxazines/analysis , Oxazines/metabolism , Plant Roots/chemistry , Plant Roots/metabolism , Pyridines/analysis , Pyridines/metabolism , Soil Pollutants/analysis , Thiamethoxam , Thiazines/analysis , Thiazines/metabolism , Thiazoles/analysis , Thiazoles/metabolism , Vegetables/chemistry , Vegetables/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...