Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
Add more filters










Publication year range
1.
Steroids ; 208: 109456, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38889811

ABSTRACT

Occupancy of prostate cancer (PCa) cell androgen receptors (AR) signals proliferation, therefore testosterone biosynthesis inhibitors and AR antagonists are important PCa treatments. Conversely, androgen mimics (e.g., prednisone) used in management of PCa might cause proliferation. The balance between PCa proliferation and inhibition predicts treatment success. We used in silico molecular modelling to explore interactions between ARs, androgens (testosterone, dihydrotestosterone (DHT)) and drugs used to treat (bicalutamide) and manage (dexamethasone, prednisone, hydrocortisone) PCa. We found that hydrogen (H-) bonds between testosterone, DHT and Arg752, Asn705 and Thr877 followed by ligand binding cleft hydrophobic interactions signal proliferation, whereas bicalutamide antagonism is via Phe764 interactions. Hydrocortisone, dexamethasone and prednisone H-bond Asn705 and Thr877, but not Arg752 in the absence of a water molecule. Studies with a bicalutamide agonist AR mutation showed different amino acid interactions, indicating testosterone and DHT would not promote proliferation as effectively as via the native receptor. However, hydrocortisone and bicalutamide form Arg752 and Asn705 H-bonds indicating agonism. Our results suggest that as PCa progresses the resulting mutations will change the proliferative response to androgens and their drug mimics, which have implications for the treatment of prostate cancer.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Male , Receptors, Androgen/metabolism , Humans , Anilides/pharmacology , Anilides/chemistry , Tosyl Compounds/pharmacology , Tosyl Compounds/chemistry , Tosyl Compounds/metabolism , Computer Simulation , Molecular Docking Simulation , Models, Molecular , Nitriles/chemistry , Nitriles/pharmacology , Nitriles/metabolism , Steroids/metabolism , Steroids/chemistry , Testosterone/metabolism , Testosterone/pharmacology , Protein Binding , Dihydrotestosterone/metabolism
2.
Pharm Dev Technol ; 26(1): 48-59, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33121318

ABSTRACT

The pharmaceutical industry has to tackle the explosion of high amounts of poorly soluble APIs. This phenomenon leads to numerous sophisticated solutions. These include the use of multifactorial data analysis identifying correlations between the components and dosage form properties, laboratory and production process parameters with respect to the API liberation Example of such API is bicalutamide. Improved liberation is achieved by particle size reduction. Laboratory batches, with different PSD of API, were filled into gelatinous capsules and consequently granulated for tablet compression. Comparative dissolution profiles with Casodex 150 mg (Astra Zeneca) were performed. The component analysis was used for the statistical evaluation of f1 and f2 factors and D(v,0.9) and D[4,3] parameters of PSD to identify optimal PSD values. Suitable PSD limits for API were statistically confirmed in laboratory and in commercial scale with respect to optimized tablet properties. The tablets were bioequivalent with originator (n = 20; 90% CI for ln AUC0-120: 99.8-111.9%; 90% CI for ln cmax: 101.1-112.9%). In conclusion, the micronisation of the API is still an efficient and inexpensive method improving the bioavailability, although there are more complicated and expensive methods available. Statistical multifactorial methods improved the safety and reproducibility of production.


Subject(s)
Anilides/chemical synthesis , Anilides/metabolism , Chemistry, Pharmaceutical/methods , Nitriles/chemical synthesis , Nitriles/metabolism , Tosyl Compounds/chemical synthesis , Tosyl Compounds/metabolism , Biological Availability , Multivariate Analysis , Tablets , Therapeutic Equivalency
3.
Eur J Med Chem ; 208: 112699, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32927391

ABSTRACT

A library of thirty N-substituted tosyl N'-acryl-hydrazones was prepared with p-toluenesulfonyl hydrazide, methyl propiolate and different aldehydes in a one-pot synthesis via an aza-Michael reaction. The scope of the reaction was studied, including aliphatic, isoprenylic, aromatic and carbocyclic aldehydes. The prepared collection was tested against Mycobacterium tuberculosis H37Rv. Nine analogs of the collection showed Minimum Inhibitory Concentration ≤10 µM, of which the most active members (MIC of 1.25 µM) were exclusively E isomers. In order to validate the mechanism of action of the most active acrylates, we tested their activity on a M. tuberculosis InhA over-expressing strain obtaining MIC that consistently doubled those obtained on the wild type strain. Additionally, the binding mode of those analogs on M. tuberculosis InhA was investigated by docking simulations. The results displayed a hydrogen bond interaction between the sulfonamide and Ile194 and the carbonyl of the methyl ester with Tyr 158 (both critical residues in the interaction with the fatty acyl chain substrate), where the main differences on the binding mode relays on the hydrophobicity of the nitrogen substituent. Additionally, chemoinformatic analysis was performed to evaluate in silico possible cytotoxicity risk and ADME-Tox profile. Based on their simple preparation and interesting antimycobacterial activity profile, the newly prepared aza-acrylates are promising candidates for antitubercular drug development.


Subject(s)
Antitubercular Agents/pharmacology , Hydrazones/pharmacology , Tosyl Compounds/pharmacology , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/metabolism , Bacterial Proteins/metabolism , Chlorocebus aethiops , Hydrazones/chemical synthesis , Hydrazones/metabolism , Isoniazid/chemistry , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Mycobacterium tuberculosis/drug effects , Oxidoreductases/metabolism , Protein Binding , Structure-Activity Relationship , Tosyl Compounds/chemical synthesis , Tosyl Compounds/metabolism , Vero Cells
4.
ChemMedChem ; 15(1): 50-67, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31670489

ABSTRACT

The nuclear farnesoid X receptor (FXR) and the enzyme soluble epoxide hydrolase (sEH) are validated molecular targets to treat metabolic disorders such as non-alcoholic steatohepatitis (NASH). Their simultaneous modulation in vivo has demonstrated a triad of anti-NASH effects and thus may generate synergistic efficacy. Here we report dual FXR activators/sEH inhibitors derived from the anti-asthma drug Zafirlukast. Systematic structural optimization of the scaffold has produced favorable dual potency on FXR and sEH while depleting the original cysteinyl leukotriene receptor antagonism of the lead drug. The resulting polypharmacological activity profile holds promise in the treatment of liver-related metabolic diseases.


Subject(s)
Epoxide Hydrolases/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/agonists , Tosyl Compounds/chemistry , Binding Sites , Catalytic Domain , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Drug Evaluation, Preclinical , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Indoles , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Molecular Docking Simulation , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Phenylcarbamates , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Sulfonamides , Tosyl Compounds/metabolism , Tosyl Compounds/pharmacology
5.
Med Sci Monit ; 25: 8753-8763, 2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31745068

ABSTRACT

BACKGROUND Zafirlukast is an antagonist of cysteinyl leukotriene receptor 1 (CysLTR1). Advanced glycation end-products (AGEs) are formed by the glycation of lipids and proteins in hyperglycemia, including diabetes mellitus. Zafirlukast has not previously been studied in diabetic nephropathy. This study aimed to investigate the effects of zafirlukast on rat renal mesangial cells cultured with AGEs in vitro. MATERIAL AND METHODS Mesangial cells were cultured in AGEs (0, 20, 50, 100 µg/ml), and with AGEs (100 µg/ml) and zafirlukast (2.5 µm, 5 µm, and 100 µm). An enzyme-linked immunoassay (ELISA) was used to measure the levels of tumor necrosis factor-alpha (TNF-alpha), interleukin-1ß (IL-1ß), IL-6, and monocyte chemoattractant protein-1 (MCP-1). Reactive oxygen species (ROS) were assessed by intracellular fluorescence measurement of 2'-7'-dichlorodihydrofluorescein diacetate (DCFH-DA), and detection kits were used to measure malondialdehyde (MDA), lactate dehydrogenase (LDH), glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD). Cell apoptosis was assessed by flow cytometry, and Western blot was used to measure protein levels. RESULTS In mesangial cells cultured with AGEs, markers of inflammation, oxidative stress, and apoptosis and levels of CysLTR1 increased, and these effects were reduced by zafirlukast in a dose-dependent manner. The effects of zafirlukast as a CysLTR1 antagonist protected mesangial cells from the effects of AGE in vitro. CONCLUSIONS Zafirlukast, a CysLTR1 antagonist, reduced the levels of inflammatory cytokines, markers of oxidative stress, and cell apoptosis induced by AGE in mesangial cells in a dose-dependent way. Future in vivo studies are needed to investigate the potential role for zafirlukast in models of diabetic nephropathy.


Subject(s)
Glycation End Products, Advanced/drug effects , Oxidative Stress/drug effects , Tosyl Compounds/pharmacology , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Glutathione Peroxidase/metabolism , Glycation End Products, Advanced/metabolism , Indoles , Inflammation/pathology , Malondialdehyde/metabolism , Mesangial Cells/drug effects , Phenylcarbamates , Rats , Reactive Oxygen Species/metabolism , Receptors, Leukotriene/metabolism , Sulfonamides , Superoxide Dismutase/metabolism , Tosyl Compounds/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Sci Adv ; 5(10): eaax2518, 2019 10.
Article in English | MEDLINE | ID: mdl-31633023

ABSTRACT

The G protein-coupled cysteinyl leukotriene receptor CysLT1R mediates inflammatory processes and plays a major role in numerous disorders, including asthma, allergic rhinitis, cardiovascular disease, and cancer. Selective CysLT1R antagonists are widely prescribed as antiasthmatic drugs; however, these drugs demonstrate low effectiveness in some patients and exhibit a variety of side effects. To gain deeper understanding into the functional mechanisms of CysLTRs, we determined the crystal structures of CysLT1R bound to two chemically distinct antagonists, zafirlukast and pranlukast. The structures reveal unique ligand-binding modes and signaling mechanisms, including lateral ligand access to the orthosteric pocket between transmembrane helices TM4 and TM5, an atypical pattern of microswitches, and a distinct four-residue-coordinated sodium site. These results provide important insights and structural templates for rational discovery of safer and more effective drugs.


Subject(s)
Anti-Asthmatic Agents/metabolism , Receptors, Leukotriene/metabolism , Anti-Asthmatic Agents/chemistry , Binding Sites , Chromones/chemistry , Chromones/metabolism , Crystallography, X-Ray , Humans , Indoles , Leukotriene Antagonists/chemistry , Leukotriene Antagonists/metabolism , Ligands , Molecular Docking Simulation , Phenylcarbamates , Protein Structure, Tertiary , Receptors, Leukotriene/chemistry , Receptors, Leukotriene/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sodium/chemistry , Sodium/metabolism , Sulfonamides , Tosyl Compounds/chemistry , Tosyl Compounds/metabolism
7.
J Inorg Biochem ; 196: 110684, 2019 07.
Article in English | MEDLINE | ID: mdl-31054419

ABSTRACT

Targeted delivery of clinically approved anticancer drug to tumor sites is an effective way to achieve enhanced drug efficacy as well as reduced side effects and toxicity. Here bicalutamide is caged by the Ru(II) center through the nitrile group, and three photoactive Ru(II) complexes were designed and synthesized. Docking study showed that the ruthenium(II) fragments can effectively block the binding of complexes 1-3 with AR (androgen receptor) owing to the large steric structures, thus bicalutamide in complexes 1-3 could not interact with AR-LBD (ligand binding domain). Once irradiation with blue light (465nm), complexes 1-3 can release bicalutamide and anticancer Ru(II) fragments, which possesses dual-action of AR binding and DNA interaction simultaneously. In vitro cytotoxicity study on these complexes further confirmed that complexes 1-3 exhibited considerable cytotoxicity upon irradiation with blue light. Significantly, complex 3 could be activated at 660nm, which greatly increases the scope of complex 3 to treat deeper within tissue. Theoretical calculations showed that the lowest singlet excitation energy of complex 3 is lower than those of complexes 1-2, which explains the experimental results well. Moreover, the 3MC (metal centered) states of these complexes are more stable than their 3MLCT (metal to ligand charge transfer) states, indicating that the photoactive processes of these complexes are likely to result in ligand dissociation.


Subject(s)
Anilides/chemistry , Anilides/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Light , Nitriles/chemistry , Nitriles/metabolism , Prostatic Neoplasms/metabolism , Ruthenium/chemistry , Tosyl Compounds/chemistry , Tosyl Compounds/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Coordination Complexes/chemistry , Humans , Magnetic Resonance Spectroscopy , Male , Microscopy, Atomic Force , PC-3 Cells , Photolysis , Prodrugs/chemistry , Prodrugs/pharmacology , Receptors, Androgen/metabolism
8.
Cell Chem Biol ; 26(2): 213-222.e6, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30527998

ABSTRACT

Diabetes is a hyperglycemic condition characterized by pancreatic ß-cell dysfunction and depletion. Whereas methods for monitoring ß-cell function in vivo exist, methods to deliver therapeutics to ß cells are lacking. We leveraged the rare ability of ß cells to concentrate zinc to preferentially trap zinc-binding molecules within ß cells, resulting in ß-cell-targeted compound delivery. We determined that zinc-rich ß cells and islets preferentially accumulated TSQ (6-methoxy-8-p-toluenesulfonamido-quinoline) in a zinc-dependent manner compared with exocrine pancreas. Next, we asked whether appending a zinc-chelating moiety onto a ß-cell replication-inducing compound was sufficient to confer preferential ß-cell accumulation and activity. Indeed, the hybrid compound preferentially accumulated within rodent and human islets in a zinc-dependent manner and increased the selectivity of replication-promoting activity toward ß cells. These data resolve the fundamental question of whether intracellular accumulation of zinc-chelating compounds is influenced by zinc content. Furthermore, application of this principle yielded a proof-of-concept method for ß-cell-targeted drug delivery and bioactivity.


Subject(s)
Chelating Agents/chemistry , Insulin-Secreting Cells/metabolism , Zinc/chemistry , Aminoquinolines/analysis , Aminoquinolines/chemistry , Aminoquinolines/metabolism , Animals , Chelating Agents/metabolism , Chromatography, High Pressure Liquid , Dithizone/chemistry , Dithizone/metabolism , Ethylenediamines/analysis , Ethylenediamines/chemistry , Ethylenediamines/metabolism , Humans , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Tandem Mass Spectrometry , Tosyl Compounds/analysis , Tosyl Compounds/chemistry , Tosyl Compounds/metabolism , Dyrk Kinases
9.
Neuroscience ; 399: 135-145, 2019 02 10.
Article in English | MEDLINE | ID: mdl-30593920

ABSTRACT

Hydrogen peroxide (H2O2) is one of the reactive oxygen species (ROS), endogenously produced during metabolism, which acts as a second messenger. In skeletal muscles, hypoxia- or hyperthermia-induced increase in H2O2 might affect synaptic transmission by targeting the most redox-sensitive presynaptic compartment (Giniatullin et al., 2006). However, the effects of H2O2 as a signal molecule have not previously been studied in different patterns of the synaptic activity. Here, using optical and microelectrode recording of synaptic vesicle exocytosis, we studied the use-dependent action of low concentrations of H2O2 and other oxidants in the mouse neuromuscular junction. We found that: (i) H2O2 at low micromole concentrations inhibited both spontaneous and evoked transmitter releases from the motor nerve terminals in a use-dependent manner, (ii) the antioxidant N-acetylcysteine (NAC) eliminated these depressant effects, (iii) the influence of H2O2 was not associated with lipid oxidation suggesting a pure signaling action, (iv) the intracellular oxidant Chloramine-T or (v) the glutathione depletion produced similar to H2O2 depressant effects. Taken together, our data revealed the effective inhibition of neurotransmitter release by ROS, which was proportional to the intensity of synaptic activity at the neuromuscular junction. The combination of various oxidants suggested an intracellular location for redox-sensitive sites responsible for modulation of the synaptic transmission in the skeletal muscle.


Subject(s)
Hydrogen Peroxide/pharmacology , Neuromuscular Junction/drug effects , Oxidants/pharmacology , Synaptic Transmission/drug effects , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Chloramines/metabolism , Diaphragm/drug effects , Diaphragm/innervation , Diaphragm/metabolism , Dose-Response Relationship, Drug , Exocytosis/drug effects , Exocytosis/physiology , Female , Glutathione/metabolism , Male , Membrane Lipids/metabolism , Mice , Neuromuscular Junction/physiology , Phrenic Nerve/drug effects , Phrenic Nerve/metabolism , Reactive Oxygen Species/metabolism , Synaptic Transmission/physiology , Synaptic Vesicles/drug effects , Synaptic Vesicles/physiology , Tissue Culture Techniques , Tosyl Compounds/metabolism
10.
Int J Biol Macromol ; 120(Pt B): 2285-2292, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30149080

ABSTRACT

Bicalutamide (BCT), a drug used in the treatment of prostate cancer, antagonises the actions of androgens, at the receptor level, thereby inhibiting the growth of prostate tumours. Alpha-2-macroglobulin (α2M), a pan-proteinase inhibitor, inhibits proteinase, regardless of specificity and catalytic mechanism. α2M is deficient in patients of advanced prostate cancer with bone metastases. Our studies explored the interaction of BCT with α2M and analysed the BCT induced structural alteration to the α2M. The result suggests that BCT decreases the antiproteolytic potential and causes structural and functional change in human α2M. UV-visible absorption spectroscopy confirms the formation of α2M-BCT complex. Fluorescence analysis shows significant quenching in fluorescence intensity of α2M upon binding with BCT. Synchronous fluorescence result suggests the interaction of BCT with α2M changed the microenvironment around tyrosine residues. Secondary structure of α2M also undergoes a slight change upon complexation with the drug as evident by shift in negative ellipticity in far UV CD spectroscopy. FTIR results confirm the alteration in secondary structure of α2M upon drug interaction. Molecular docking studies show that BCT bind to a monomer of α2M primarily through hydrophobic force. Thermodynamics parameters were determined by isothermal titration calorimetry found that the binding was exothermic in nature.


Subject(s)
Androgen Antagonists/metabolism , Anilides/metabolism , Molecular Docking Simulation , Nitriles/metabolism , Pregnancy-Associated alpha 2-Macroglobulins/metabolism , Tosyl Compounds/metabolism , alpha-Macroglobulins/metabolism , Humans , Protease Inhibitors/metabolism , Protein Binding , Protein Conformation , alpha-Macroglobulins/chemistry
11.
ACS Appl Mater Interfaces ; 10(37): 31168-31177, 2018 Sep 19.
Article in English | MEDLINE | ID: mdl-30156819

ABSTRACT

Advanced technologies for controlled cell adhesion and detachment in novel biointerface designs profit from stimuli-responsive systems that are able to react to their environment. Here, a multilayer system made of thiolated chitosan and thiolated chondroitin sulfate was constructed, with the potential of switchable inter- and intramolecular thiol/disulfide interactions representing a redox-sensitive nanoplatform. Owing to the formation and cleavage of inherent disulfide bonds by oxidation and reduction, surface properties of the multilayer can be controlled toward protein adsorption/desorption and cell adhesion in a reversible manner. Oxidation of thiols by chloramine-T promotes fibronectin (FN) adsorption and fibroblast cell adhesion, whereas the reduction by tris(2-carboxyethyl)phosphine reverses these effects, leading to low FN adsorption and little cell adhesion and spreading. These effects on the biological systems are related to significant changes of wetting properties, zeta potential, and mechanical properties of these multilayer films. The system presented may be useful for biomedical applications as responsive and obedient surfaces in medical implants and support tissue regeneration.


Subject(s)
Cell Adhesion/drug effects , Chitosan/chemistry , Chondroitin Sulfates/chemistry , Fibroblasts/drug effects , Adsorption , Cells, Cultured , Chloramines/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Humans , Oxidation-Reduction , Sulfhydryl Compounds/chemistry , Surface Properties , Tosyl Compounds/metabolism
12.
J Med Chem ; 61(13): 5758-5764, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29878767

ABSTRACT

Multitarget design offers access to bioactive small molecules with potentially superior efficacy and safety. Particularly multifactorial chronic inflammatory diseases demand multiple pharmacological interventions for stable treatment. By minor structural changes, we have developed a close analogue of the cysteinyl-leukotriene receptor antagonist zafirlukast that simultaneously inhibits soluble epoxide hydrolase and activates peroxisome proliferator-activated receptor γ. The triple modulator exhibits robust anti-inflammatory activity in vivo and highlights the therapeutic potential of designed multitarget agents.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Design , Polypharmacology , Tosyl Compounds/pharmacology , 3T3 Cells , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Catalytic Domain , Epoxide Hydrolases/chemistry , Epoxide Hydrolases/metabolism , Hep G2 Cells , Humans , Indoles , Mice , Molecular Docking Simulation , PPAR gamma/chemistry , PPAR gamma/metabolism , Phenylcarbamates , Sulfonamides , Tosyl Compounds/metabolism
13.
Sci Rep ; 8(1): 2405, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29402925

ABSTRACT

The release of aromatic amines from drugs and other xenobiotics resulting from the hydrolysis of metabolically labile amide bonds presents a safety risk through several mechanisms, including geno-, hepato- and nephrotoxicity. Whilst multiple in vitro systems used for studying metabolic stability display serine hydrolase activity, responsible for the hydrolysis of amide bonds, they vary in their efficiency and selectivity. Using a range of amide-containing probe compounds (0.5-10 µM), we have investigated the hydrolytic activity of several rat, minipig and human-derived in vitro systems - including Supersomes, microsomes, S9 fractions and hepatocytes - with respect to their previously observed human in vivo metabolism. In our hands, human carboxylesterase Supersomes and rat S9 fractions systems showed relatively poor prediction of human in vivo metabolism. Rat S9 fractions, which are commonly utilised in the Ames test to assess mutagenicity, may be limited in the detection of genotoxic metabolites from aromatic amides due to their poor concordance with human in vivo amide hydrolysis. In this study, human liver microsomes and minipig subcellular fractions provided more representative models of human in vivo hydrolytic metabolism of the aromatic amide compounds tested.


Subject(s)
Amides/metabolism , Carboxylesterase/metabolism , Hepatocytes/metabolism , Microsomes, Liver/metabolism , Subcellular Fractions/metabolism , Acetaminophen/metabolism , Acetanilides/metabolism , Anilides/metabolism , Animals , Flutamide/metabolism , Humans , Hydrolysis , Lidocaine/metabolism , Male , Niclosamide/metabolism , Nitriles/metabolism , Prilocaine/metabolism , Primary Cell Culture , Propanil/metabolism , Rats , Rats, Sprague-Dawley , Swine , Swine, Miniature , Tosyl Compounds/metabolism
14.
Metallomics ; 9(4): 391-401, 2017 04 19.
Article in English | MEDLINE | ID: mdl-27918051

ABSTRACT

Nitric oxide (NO) is both an important regulatory molecule in biological systems and a toxic xenobiotic. Its oxidation products react with sulfhydryl groups and either nitrosylate or oxidize them. The aerobic reaction of NO supplied by diethylamine NONOate (DEA-NO) with pig kidney LLC-PK1 cells and Zn-proteins within the isolated proteome was examined with three fluorescent zinc sensors, zinquin (ZQ), TSQ, and FluoZin-3 (FZ-3). Observations of Zn2+ labilization from Zn-proteins depended on the specific sensor used. Upon cellular exposure to DEA-NO, ZQ sequestered about 13% of the proteomic Zn2+ as Zn(ZQ)2 and additional Zn2+ as proteome·Zn-ZQ ternary complexes. TSQ, a sensor structurally related to ZQ with lower affinity for Zn2+, did not form Zn(TSQ)2. Instead, Zn2+ mobilized by DEA-NO was exclusively bound as proteome·Zn-TSQ adducts. Analogous reactions of proteome with ZQ or TSQ in vitro displayed qualitatively similar products. Titration of native proteome with Zn2+ in the presence of ZQ resulted in the sole formation of proteome·Zn-ZQ species. This result suggested that sulfhydryl groups are involved in non-specific proteomic binding of mobile Zn2+ and that the appearance of Zn(ZQ)2 after exposure of cells and proteome to DEA-NO resulted from a reduction in proteomic sulfhydryl ligands, favoring the formation of Zn(ZQ)2 instead of proteome·Zn-ZQ. With the third sensor, FluoZin-3, neither Zn-FZ-3 nor proteome·Zn-FZ-3 was detected during the reaction of proteome with DEA-NO. Instead, it reacted independently with DEA-NO with a modest enhancement of fluorescence.


Subject(s)
Fluorescent Dyes/metabolism , Hydrazines/metabolism , Nitric Oxide Donors/metabolism , Nitric Oxide/metabolism , Proteome/metabolism , Spectrometry, Fluorescence/methods , Zinc/metabolism , Animals , Fluorescent Dyes/analysis , LLC-PK1 Cells , Metalloproteins/analysis , Metalloproteins/metabolism , Polycyclic Compounds/analysis , Polycyclic Compounds/metabolism , Proteome/analysis , Proteomics/methods , Quinolones/analysis , Quinolones/metabolism , Sulfhydryl Compounds/analysis , Sulfhydryl Compounds/metabolism , Swine , Tosyl Compounds/analysis , Tosyl Compounds/metabolism , Zinc/analysis
15.
Drug Test Anal ; 9(7): 1017-1025, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27726308

ABSTRACT

Besides their development for therapeutic purposes, non-steroidal selective androgen receptor modulators (non-steroidal SARMs) are also known to impact growth-associated pathways as ligands of androgenic receptors (AR). They present a potential for abuse in sports and food-producing animals as an interesting alternative to anabolic androgenic steroids (AAS). These compounds are easily available and could therefore be (mis)used in livestock production as growth promoters. To prevent such practices, dedicated analytical strategies should be developed for specific and sensitive detection of these compounds in biological matrices. The present study focused on Bicalutamide, a non-steroidal SARM used in human treatment of non-metastatic prostate cancer because of its anti-androgenic activity exhibiting no anti-anabolic effects. To select the most appropriate matrix to be used for control purposes, different animal matrices (urine and faeces) have been investigated and SARM metabolism studied to highlight relevant metabolites of such treatments and establish associated detection time windows. The aim of this work was thus to compare the urinary and faecal eliminations of bicalutamide in a calf, and investigate phase I and II metabolites. The results in both matrices showed that bicalutamide was very rapidly and mainly excreted under its free form. The concentration levels were observed as higher in faeces (ppm) than urine (ppb); although both matrices were assessed as suitable for residue control. The metabolites found were consistent with hydroxylation (phase I reaction) combined or not with glucuronidation and sulfation (phase II reactions). Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Androgen Antagonists/analysis , Androgen Antagonists/urine , Anilides/analysis , Anilides/urine , Cattle/urine , Feces/chemistry , Nitriles/analysis , Nitriles/urine , Tosyl Compounds/analysis , Tosyl Compounds/urine , Androgen Antagonists/metabolism , Anilides/metabolism , Animals , Cattle/metabolism , Chromatography, High Pressure Liquid/methods , Doping in Sports , Nitriles/metabolism , Receptors, Androgen/metabolism , Substance Abuse Detection/methods , Tandem Mass Spectrometry/methods , Tosyl Compounds/metabolism
16.
Eur J Med Chem ; 118: 230-43, 2016 Aug 08.
Article in English | MEDLINE | ID: mdl-27131065

ABSTRACT

Prostate cancer (PC) is one of the major causes of male death worldwide and the development of new and more potent anti-PC compounds is a constant requirement. Among the current treatments, (R)-bicalutamide and enzalutamide are non-steroidal androgen receptor antagonist drugs approved also in the case of castration-resistant forms. Both these drugs present a moderate antiproliferative activity and their use is limited due to the development of resistant mutants of their biological target. Insertion of fluorinated and perfluorinated groups in biologically active compounds is a current trend in medicinal chemistry, applied to improve their efficacy and stability profiles. As a means to obtain such effects, different modifications with perfluoro groups were rationally designed on the bicalutamide and enzalutamide structures, leading to the synthesis of a series of new antiproliferative compounds. Several new analogues displayed improved in vitro activity towards four different prostate cancer cell lines, while maintaining full AR antagonism and therefore representing promising leads for further development. Furthermore, a series of molecular modelling studies were performed on the AR antagonist conformation, providing useful insights on potential protein-ligand interactions.


Subject(s)
Anilides/chemical synthesis , Anilides/pharmacology , Drug Design , Nitriles/chemical synthesis , Nitriles/pharmacology , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/pathology , Tosyl Compounds/chemical synthesis , Tosyl Compounds/pharmacology , Anilides/chemistry , Anilides/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Benzamides , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Drug Resistance, Neoplasm/drug effects , Humans , Male , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Nitriles/chemistry , Nitriles/metabolism , Permeability , Phenylthiohydantoin/chemical synthesis , Phenylthiohydantoin/chemistry , Phenylthiohydantoin/metabolism , Phenylthiohydantoin/pharmacology , Protein Conformation , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Tosyl Compounds/chemistry , Tosyl Compounds/metabolism
17.
Antonie Van Leeuwenhoek ; 108(6): 1485-1494, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26419902

ABSTRACT

The nematode-trapping fungi possess a unique capability of predating and invading nematodes. As a representative nematode-trapping fungus, Arthrobotrys oligospora has been widely used to study the interactions between nematode-trapping fungi and their hosts. Serine proteinase is one of the important virulence factors during process of invasion of the nematode-trapping fungi into nematodes. In this study, using reverse transcription polymerase chain reaction, we amplified the gene sequence of serine proteinase 186 from A. oligospora, cloned it into pPIC9K vector and expressed it in the yeast Pichia pastoris. The expressed recombinant serine proteinase186 (reP186) was purified via Ni-affinity chromatography. The in vitro nematode-degrading activity of reP186 was analyzed. Sodium dodecyl sulfate polyacrylamide gel electrophoresis and Western blot analysis revealed that reP186 with molecular weight of 33 kDa was successfully obtained. ReP186 was capable of degrading a series of protein substrates including casein, gelatin, bovine serum albumin, denatured collagen and nematode cortical layer. The reP186 exhibited the maximal activity at pH 8.0 and 55 °C and was highly sensitive to the inhibitor, phenylmethanesulfonylfluoride. Treatment of Caenorhabditis elegans and Haemonchus contortus with reP186 for 12, 24 and 36 h, respectively, resulted in 62, 88 and 100 % of killing rates for C. elegans, and 52, 65 and 84 % of killing rates for H. contortus, respectively, indicating a relatively strong nematode-degrading bioactivity of reP186.


Subject(s)
Ascomycota/enzymology , Caenorhabditis elegans/drug effects , Haemonchus/drug effects , Recombinant Proteins/metabolism , Serine Proteases/metabolism , Animals , Blotting, Western , Caenorhabditis elegans/physiology , Chromatography, Affinity , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Gene Expression , Haemonchus/physiology , Hydrogen-Ion Concentration , Molecular Weight , Pichia/genetics , Pichia/metabolism , Polymerase Chain Reaction , Protease Inhibitors/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Serine Proteases/chemistry , Serine Proteases/genetics , Serine Proteases/isolation & purification , Survival Analysis , Temperature , Tosyl Compounds/metabolism
18.
Drug Metab Dispos ; 43(6): 812-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25834030

ABSTRACT

Inhibition of drug metabolizing enzymes is a major mechanism in drug-drug interactions (DDIs). A number of cases of DDIs via inhibition of UDP-glucuronosyltranseferases (UGTs) have been reported, although the changes in pharmacokinetics are relatively small in comparison with drugs that are metabolized by cytochrome P450s. Most of the past studies have investigated hepatic UGTs, although recent studies have revealed a significant contribution of UGTs in the small intestine to drug clearance. To evaluate potential DDIs caused by inhibition of intestinal UGTs, we assessed inhibitory effects of 578 compounds, including drugs, xenobiotics, and endobiotics, on human UGT1A8 and UGT1A10, which are major contributors to intestinal glucuronidation. We identified 29 inhibitors by monitoring raloxifene glucuronidation with recombinant UGTs. All of the inhibitors potently inhibited UGT1A1 activity, as well. We found that zafirlukast is a potent general inhibitor of UGT1As and a moderate inhibitor of UGT2Bs because it monitors 4-methylumbelliferone glucuronidation by recombinant UGTs. However, zafirlukast did not potently inhibit diclofenac glucuronidation, suggesting that the inhibitory effects might be substrate specific. Inhibitory effects of zafirlukast on some UGT substrates were further investigated in human liver and human small intestine microsomes in order to evaluate potential DDIs. The R values (the ratios of intrinsic clearance with and without an inhibitor) revealed that zafirlukast has potential to cause clinical DDIs in the small intestine. Although we could not identify specific UGT1A8 and UGT1A10 inhibitors, zafirlukast was identified as a general inhibitor for UGTs in vitro. The present study suggests that the inhibition of UGT in the small intestine would be an underlying mechanism for DDIs.


Subject(s)
Enzyme Inhibitors/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Intestine, Small/drug effects , Leukotriene Antagonists/pharmacology , Metabolic Detoxication, Phase II , Microsomes/drug effects , Tosyl Compounds/pharmacology , Drug Evaluation, Preclinical , Drug Interactions , Enzyme Inhibitors/adverse effects , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Indoles , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestine, Small/enzymology , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Leukotriene Antagonists/adverse effects , Leukotriene Antagonists/metabolism , Microsomes/enzymology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Phenylcarbamates , Raloxifene Hydrochloride/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Selective Estrogen Receptor Modulators/metabolism , Small Molecule Libraries , Substrate Specificity , Sulfonamides , Tosyl Compounds/adverse effects , Tosyl Compounds/metabolism
19.
J Pharm Biomed Anal ; 111: 324-32, 2015.
Article in English | MEDLINE | ID: mdl-25771488

ABSTRACT

The binding of some novel bicalutamide analogues to human serum albumin (HSA) and rat serum albumin (RSA) was investigated by surface plasmon resonance (SPR) based optical biosensor technique. The serum protein binding of the bicalutamide analogues was determined and compared to that of the parent compound. Furthermore, HSA and RSA were used as target plasma proteins, in order to highlight possible differences among species when performing pharmacokinetic studies. HSA and RSA were covalently immobilized on carboxymethyl dextran matrixes, using an amine coupling procedure. The anchoring method was validated by determining the dissociation constant (KD) of a standard analyte to confirm that the binding properties of the proteins were maintained. The ranking of the bicalutamide analogues for their HSA and RSA bound fractions was used to compare the behaviour of the two albumins. Most of the bicalutamide analogues showed higher binding levels with respect to the lead compound, (R)-bicalutamide. Further, meaningful differences in the binding level to the two serum proteins were obtained. The dissociation constants (KD) of the interaction between the lead compound, (R)-bicalutamide, and the two proteins were calculated. As a result, the KD obtained with HSA was one order of magnitude higher than that obtained with RSA. The observed differences in the HSA and RSA bonding of the bicalutamide analogues increase the knowledge on the possible low reliability in extrapolating the distribution data obtained on animals to humans. This work demonstrates that SPR based optical biosensor technique is well suited for the medium-high throughput screening of compounds' ligand binding to serum albumins.


Subject(s)
Anilides/metabolism , Nitriles/metabolism , Serum Albumin/metabolism , Tosyl Compounds/metabolism , Animals , Biosensing Techniques/methods , Blood Proteins/metabolism , Dextrans/metabolism , Humans , Lead/metabolism , Protein Binding , Rats , Reproducibility of Results , Surface Plasmon Resonance/methods
20.
Methods Mol Biol ; 1266: 243-63, 2015.
Article in English | MEDLINE | ID: mdl-25560080

ABSTRACT

Introducing nongenetically encoded, synthetic probes into specific proteins is now recognized as a key component in chemical biology. In particular, the ability to chemically modify specific "native" proteins in various contexts from in vitro to cellular systems is of fundamental importance to study biological systems. We developed a protein-labeling technique termed ligand-directed tosyl (LDT) chemistry for this purpose. This method is capable of labeling specific native proteins with diverse synthetic probes with high site specificity and target selectivity without compromising protein function. Here we describe the principle of the LDT chemistry and the protocol for selective chemical labeling of native carbonic anhydrase in vitro, in blood cells (ex vivo), and in living mice (in vivo).


Subject(s)
Carbonic Anhydrase II/metabolism , Tosyl Compounds/chemistry , Animals , Carbonic Anhydrase II/chemistry , Cells, Cultured , Ligands , Male , Mice, Inbred ICR , Staining and Labeling , Tosyl Compounds/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...