Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(12): 9816-9841, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38857426

ABSTRACT

Clinical guidelines for COPD and asthma recommend inhaled ß-adrenergic agonists, muscarinic antagonists, and, for frequent exacerbators, inhaled corticosteroids, with the challenge of combining them into a single device. The MABA (muscarinic antagonist and ß2 agonist) concept has the potential to simplify this complexity while increasing the efficacy of both pharmacologies. In this article, we report the outcome of our solid-state driven back-up program that led to the discovery of the MABA compound CHF-6550. A soft drug approach was applied, aiming at high plasma protein binding and high hepatic clearance, concurrently with an early stage assessment of crystallinity through a dedicated experimental workflow. A new chemotype was identified, the diphenyl hydroxyacetic esters, able to generate crystalline material. Among this class, CHF-6550 demonstrated in vivo efficacy, suitability for dry powder inhaler development, favorable pharmacokinetics, and safety in preclinical settings and was selected as a back-up candidate, fulfilling the desired pharmacological and solid-state profile.


Subject(s)
Adrenergic beta-2 Receptor Agonists , Muscarinic Antagonists , Muscarinic Antagonists/pharmacokinetics , Muscarinic Antagonists/pharmacology , Muscarinic Antagonists/chemistry , Muscarinic Antagonists/chemical synthesis , Muscarinic Antagonists/therapeutic use , Muscarinic Antagonists/administration & dosage , Animals , Humans , Adrenergic beta-2 Receptor Agonists/pharmacokinetics , Adrenergic beta-2 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Agonists/chemistry , Adrenergic beta-2 Receptor Agonists/administration & dosage , Administration, Inhalation , Rats , Drug Discovery , Structure-Activity Relationship , Male , Pulmonary Disease, Chronic Obstructive/drug therapy
2.
Sci Rep ; 13(1): 6116, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37059833

ABSTRACT

Mass spectrometry (MS)-based proteomics has recently attracted the attention from forensic pathologists. This work is the first report of the development of a shotgun bottom-up proteomic approach based on rapid protein extraction and nano-liquid chromatography/high-resolution mass spectrometry applied to full-thickness human skin for the differential analysis of normal and ecchymotic tissues to identify new biomarkers for bruise characterization and dating. We identified around 2000 proteins from each pooled extract. The method showed excellent precision on independent replicates, with Pearson correlation coefficients always higher than 95%. Glycophorin A, a known biomarker of vital wounds from immunochemical studies, was identified only in ecchymotic tissues, as confirmed by Western blotting analysis. This finding suggests that this protein can be used as a MS-detectable biomarker of wound vitality. By focusing on skin samples from individuals with known wound dating, besides Glycophorin A, other proteins differentially expressed in ecchymotic samples and dependant on wound age were identified, although further analysis on larger datasets are needed to validate these findings. This study paves the way for an in-depth investigation of the potential of MS-based techniques for wound examination in forensic pathology, overcoming the limitations of immunochemical assays.


Subject(s)
Glycophorins , Tandem Mass Spectrometry , Humans , Tandem Mass Spectrometry/methods , Proteomics/methods , Forensic Pathology , Proteins/metabolism , Biomarkers
3.
Biomolecules ; 12(9)2022 09 07.
Article in English | MEDLINE | ID: mdl-36139099

ABSTRACT

ATP-binding cassette (ABC) transporters are a large superfamily of membrane transporters that facilitate the translocation of different substrates. While ABC transporters are clearly expressed in various tumor cells where they can play a role in drug extrusion, the presence of these transporters in normal lung tissues is still controversial. Here, we performed an analysis of ABC transporters in EpiAlveolarTM, a recently developed model of human alveoli, by defining the expression and activity of MDR1, BCRP, and MRPs. Immortalized primary epithelial cells hAELVi (human alveolar epithelial lentivirus-immortalized cells) were employed for comparison. Our data underline a close homology between these two models, where none of the ABC transporters here studied are expressed on the apical membrane and only MRP1 is clearly detectable and functional at the basolateral side. According to these findings, we can conclude that other thus-far-unidentified transporter/s involved in drug efflux from alveolar epithelium deserve investigations.


Subject(s)
ATP-Binding Cassette Transporters , Alveolar Epithelial Cells , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate , Alveolar Epithelial Cells/metabolism , Humans , Membrane Transport Proteins , Neoplasm Proteins/metabolism
4.
J Med Chem ; 65(15): 10233-10250, 2022 08 11.
Article in English | MEDLINE | ID: mdl-35901125

ABSTRACT

The development of molecules embedding two distinct pharmacophores acting as muscarinic antagonists and ß2 agonists (MABAs) promises to be an excellent opportunity to reduce formulation issues and boost efficacy through cross-talk and allosteric interactions. Herein, we report the results of our drug discovery campaign aimed at improving the therapeutic index of a previous MABA series by exploiting the super soft-drug concept. The incorporation of a metabolic liability, stable at the site of administration but undergoing rapid systemic metabolism, to generate poorly active and quickly eliminated fragments was pursued. Our SAR studies yielded MABA 29, which demonstrated a balanced in vivo profile up to 24 h, high instability in plasma and the liver, as well as sustained exposure in the lung. In vitro safety and non-GLP toxicity studies supported the nomination of 29 (CHF-6366) as a clinical candidate, attesting to the successful development of a novel super-soft MABA compound.


Subject(s)
Muscarinic Antagonists , Pulmonary Disease, Chronic Obstructive , Administration, Inhalation , Adrenergic beta-2 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Agonists/therapeutic use , Bronchodilator Agents/therapeutic use , Drug Discovery , Humans , Lung , Muscarinic Antagonists/pharmacology , Muscarinic Antagonists/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy
5.
Biochem Biophys Res Commun ; 576: 27-32, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34478916

ABSTRACT

Alveolar epithelium, besides exerting a key role in gas exchange and surfactant production, plays important functions in host defense and inflammation. Pathological conditions associated to alveolar dysfunction include Acute Respiratory Distress Syndrome (ARDS), asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). The use of predictive in vitro models of human alveolar epithelium is nowadays required for the study of disease mechanisms, as well as of pharmacokinetic parameters of pulmonary drugs delivery. Here, we employed a novel 3D model of human alveoli, namely EpiAlveolar™, consisting of primary alveolar epithelial cells, pulmonary endothelial cells and fibroblasts, that reflects properly the in vivo-like conditions. In EpiAlveolar™ we performed a characterization of Organic Cation Transporters (OCTs and OCTNs) expression and activity and we found that OCTN2, OCT1 and OCT3 are expressed on the basolateral membrane; instead, ATB0,+ transporter for cationic and neutral amino acids, which shares with OCTN2 the affinity for carnitine as substrate, is readily detectable and functional at the apical side. We also show that these transporters differentially interact with anticholinergic drugs. Overall, our findings reveal close similarities of EpiAlveolar™ with the tracheal/bronchial epithelium (EpiAirway™ model) and entrust this alveolar tissue as a potential tool for the screening of biopharmaceuticals molecules.


Subject(s)
Alveolar Epithelial Cells/metabolism , Endothelial Cells/metabolism , Fibroblasts/metabolism , Lung/cytology , Organic Cation Transport Proteins/metabolism , Coculture Techniques , Humans , Lung/metabolism , Primary Cell Culture
6.
Bioorg Med Chem Lett ; 41: 127975, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33753262

ABSTRACT

The targeting of both the muscarinic and ß-adrenergic pathways is a well validated therapeutic approach for the treatment of chronic obstructive pulmonary disease (COPD). In this communication we report our effort to incorporate two pharmacologies into a single chemical entity, whose characteristic must be suitable for a once daily inhaled administration. Contextually, we aimed at a locally acting therapy with limited systemic absorption to minimize side effects. Our lung-tailored design of bifunctional compounds that combine the muscarinic and ß-adrenergic pharmacologies by the elaboration of the muscarinic inhibitor 7, successfully led to the potent, pharmacologically balanced muscarinic antagonist and ß2 agonist (MABA) 13.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Bronchodilator Agents/pharmacology , Drug Discovery , Muscarinic Antagonists/pharmacology , Pulmonary Disease, Chronic Obstructive/drug therapy , Administration, Inhalation , Adrenergic beta-2 Receptor Agonists/administration & dosage , Bronchodilator Agents/administration & dosage , Dose-Response Relationship, Drug , Humans , Molecular Structure , Muscarinic Antagonists/administration & dosage , Pulmonary Disease, Chronic Obstructive/metabolism , Receptor, Muscarinic M3/antagonists & inhibitors , Receptor, Muscarinic M3/metabolism , Receptors, Adrenergic, beta-2/metabolism , Structure-Activity Relationship
7.
Mol Nutr Food Res ; 64(16): e2000289, 2020 08.
Article in English | MEDLINE | ID: mdl-32640069

ABSTRACT

SCOPE: Urolithin A and B are gut metabolites of ellagic acid and ellagitannins associated with many beneficial effects. Evidence in vitro pointed to their potential as estrogenic modulators. However, both molecular mechanisms and biological targets involved in such activity are still poorly characterized, preventing a comprehensive understanding of their bioactivity in living organisms. This study aimed at rationally identifying novel biological targets underlying the estrogenic-modulatory activity of urolithins. METHODS AND RESULTS: The work relies on an in silico/in vitro target fishing study coupling molecular modeling with biochemical and cell-based assays. Estrogen sulfotransferase and 17ß-hydroxysteroid dehydrogenase are identified as potentially subject to inhibition by the investigated urolithins. The inhibition of the latter undergoes experimental confirmation either in a cell-free or cell-based assay, validating computational outcomes. CONCLUSIONS: The work describes target fishing as an effective tool to identify unexpected targets of food bioactives detailing the interaction at a molecular level. Specifically, it described, for the first time, 17ß-hydroxysteroid dehydrogenase as a target of urolithins and highlighted the need of further investigations to widen the understanding of urolithins as estrogen modulators in living organisms.


Subject(s)
Coumarins/pharmacology , Enzyme Inhibitors/pharmacology , Estradiol Dehydrogenases/antagonists & inhibitors , Proteins/metabolism , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/metabolism , Cell-Free System , Computer Simulation , Coumarins/chemistry , Coumarins/metabolism , Humans , Ligands , MCF-7 Cells , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Proteins/chemistry , Sulfotransferases/chemistry , Sulfotransferases/metabolism
8.
Biomedicines ; 8(5)2020 May 19.
Article in English | MEDLINE | ID: mdl-32438722

ABSTRACT

Organic cation transporters (OCTs) and novel organic cation transporters (OCTNs) are responsible for drug delivery in the intestine and kidney; in the lung, OCTs mediate inhaled drugs' transport, although their physiological role in airways remains poorly understood. The studies addressing OCTs/OCTNs in human airways were mostly performed in immortal or transformed cell lines; here, we studied OCTs in EpiAirway™, a recently developed in vitro model of normal bronchial epithelium. Calu-3 monolayers were used for comparison. The activity of OCTs was evaluated by measuring the uptake of 1-methyl-4-phenylpyridinium (MPP+) at the apical and basolateral side of monolayers and protein expression through Western Blot analysis. OCTs and OCTNs expression, along with that of Amino acid Transporter B0,+ (ATB0,+)transporter, was determined by measuring the number of mRNA molecules through quantitative Polymerase Chain Reaction (qPCR). The interaction of the transporters with bronchodilators was also assessed. Results highlight significant differences between Calu-3 cells and EpiAirway™, since, in the latter, OCTs are active only on the basolateral membrane where they interact with the bronchodilator ipratropium. No activity of OCTs is detectable at the apical side; there, the most abundant carrier is, instead, SLC6A14/ATB0,+, that can thus be potentially listed among organic cation transporters responsible for drug delivery in the lung.

9.
Int J Mol Sci ; 21(9)2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32366035

ABSTRACT

The ATP-binding cassette (ABC) transporters P-glycoprotein (MDR1/ABCB1), multidrug resistance-associated protein 1 (MRP1/ABCC1), and breast cancer resistance protein (BCRP/ABCG2) play a crucial role in the translocation of a broad range of drugs; data about their expression and activity in lung tissue are controversial. Here, we address their expression, localization and function in EpiAirway™, a three-dimensional (3D)-model of human airways; Calu-3 cells, a representative in vitro model of bronchial epithelium, are used for comparison. Transporter expression has been evaluated with RT-qPCR and Western blot, the localization with immunocytochemistry, and the activity by measuring the apical-to-basolateral and basolateral-to-apical fluxes of specific substrates in the presence of inhibitors. EpiAirway™ and Calu-3 cells express high levels of MRP1 on the basolateral membrane, while they profoundly differ in terms of BCRP and MDR1: BCRP is detected in EpiAirway™, but not in Calu-3 cells, while MDR1 is expressed and functional only in fully-differentiated Calu-3; in EpiAirway™, MDR1 expression and activity are undetectable, consistently with the absence of the protein in specimens from human healthy bronchi. In summary, EpiAirway™ appears to be a promising tool to study the mechanisms of drug delivery in the bronchial epithelium and to clarify the role of ABC transporters in the modulation of the bioavailability of administered drugs.


Subject(s)
Bronchi/metabolism , Epithelium/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Blotting, Western , Cell Line, Tumor , Humans , Immunohistochemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Respiratory Mucosa/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
PLoS One ; 15(2): e0228568, 2020.
Article in English | MEDLINE | ID: mdl-32027707

ABSTRACT

In human, OCTN2 (SLC22A5) and ATB0,+ (SLC6A14) transporters mediate the uptake of L-carnitine, essential for the transport of fatty acids into mitochondria and the subsequent degradation by ß-oxidation. Aim of the present study was to characterize L-carnitine transport in EpiAirway™, a 3D organotypic in vitro model of primary human tracheal-bronchial epithelial cells that form a fully differentiated, pseudostratified columnar epithelium at air-liquid interface (ALI) condition. In parallel, Calu-3 monolayers grown at ALI for different times (8d or 21d of culture) were used as comparison. OCTN2 transporter was equally expressed in both models and functional at the basolateral side. ATB0,+ was, instead, highly expressed and active on the apical membrane of EpiAirway™ and only in early-cultures of Calu-3 (8d but not 21d ALI). In both cell models, L-carnitine uptake on the apical side was significantly inhibited by the bronchodilators glycopyrrolate and tiotropium, that hence can be considered substrates of ATB0,+; ipratropium was instead effective on the basolateral side, indicating its interaction with OCTN2. Inflammatory stimuli, such as LPS or TNFα, caused an induction of SLC6A14/ATB0,+ expression in Calu-3 cells, along with a 2-fold increase of L-carnitine uptake only at the apical side; on the contrary SLC22A5/OCTN2 was not affected. As both OCTN2 and ATB0,+, beyond transporting L-carnitine, have a significant potential as delivery systems for drugs, the identification of these transporters in EpiAirway™ can open new fields of investigation in the study of drug inhalation and pulmonary delivery.


Subject(s)
Amino Acid Transport System ASC/physiology , Carnitine/metabolism , Epithelial Cells/chemistry , Respiratory System/cytology , Solute Carrier Family 22 Member 5/physiology , Amino Acid Transport System ASC/analysis , Biological Transport/drug effects , Bronchodilator Agents/pharmacology , Cell Culture Techniques/methods , Cell Polarity , Glycopyrrolate/pharmacology , Humans , Solute Carrier Family 22 Member 5/analysis , Tiotropium Bromide/pharmacology
12.
Mol Ther Methods Clin Dev ; 14: 1-15, 2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31236426

ABSTRACT

Recently, mesenchymal stromal stem cells (MSCs) have been proposed as therapeutic agents because of their promising preclinical features and good safety profile. However, their introduction into clinical practice has been associated with a suboptimal therapeutic profile. In this review, we address the biodistribution of MSCs in preclinical studies with a focus on the current understanding of the pharmacodynamics (PD) and pharmacokinetics (PK) of MSCs as key aspects to overcome unsatisfactory clinical benefits of MSC application. Beginning with evidence of MSC biodistribution and highlighting PK and PD factors, a new PK-PD model is also proposed. According to this theory, MSCs and their released factors are key players in PK, and the efficacy biomarkers are considered relevant for PD in more predictive preclinical investigations. Accounting for the PK-PD relationship in MSC translational research and proposing new models combined with better biodistribution studies could allow realization of the promise of more robust MSC clinical translation.

13.
Front Immunol ; 9: 508, 2018.
Article in English | MEDLINE | ID: mdl-29616026

ABSTRACT

Lysinuric protein intolerance (LPI) is a recessively inherited aminoaciduria caused by mutations of SLC7A7, the gene encoding y+LAT1 light chain of system y+L for cationic amino acid transport. The pathogenesis of LPI is still unknown. In this study, we have utilized a gene silencing approach in macrophages and airway epithelial cells to investigate whether complications affecting lung and immune system are directly ascribable to the lack of SLC7A7 or, rather, mediated by an abnormal accumulation of arginine in mutated cells. When SLC7A7/y+LAT1 was silenced in human THP-1 macrophages and A549 airway epithelial cells by means of short interference RNA (siRNA), a significant induction of the expression and release of the inflammatory mediators IL1ß and TNFα was observed, no matter the intracellular arginine availability. This effect was mainly regulated at transcriptional level through the activation of NFκB signaling pathway. Moreover, since respiratory epithelial cells are the important sources of chemokines in response to pro-inflammatory stimuli, the effect of IL1ß has been addressed on SLC7A7 silenced A549 cells. Results obtained indicated that the downregulation of SLC7A7/y+LAT1 markedly strengthened the stimulatory effect of the cytokine on CCL5/RANTES expression and release without affecting the levels of CXCL8/IL8. Consistently, also the conditioned medium of silenced THP-1 macrophages activated airway epithelial cells in terms of CCL5/RANTES expression due to the presence of elevated amount of proinflammatory cytokines. In conclusion, our results point to a novel thus far unknown function of SLC7A7/y+LAT1, that, under physiological conditions, besides transporting arginine, may act as a brake to restrain inflammation.


Subject(s)
Amino Acid Metabolism, Inborn Errors/immunology , Fusion Regulatory Protein 1, Light Chains/metabolism , Inflammation/immunology , Macrophages/immunology , Renal Aminoacidurias/immunology , Respiratory Mucosa/immunology , A549 Cells , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Transport System y+L , Chemokine CCL5/metabolism , Fusion Regulatory Protein 1, Light Chains/genetics , Gene Silencing , Humans , Inflammation/genetics , Interleukin-1beta/metabolism , Mutation/genetics , NF-kappa B/metabolism , Phenotype , RNA, Small Interfering/genetics , Renal Aminoacidurias/genetics , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism
14.
J Leukoc Biol ; 101(3): 665-674, 2017 03.
Article in English | MEDLINE | ID: mdl-27733576

ABSTRACT

l-Carnitine, in addition to playing a fundamental role in the ß-oxidation of fatty acids, has been recently identified as a modulator of immune function, although the mechanisms that underlie this role remain to be clarified. In this study, we addressed the modulation of l-carnitine transport and expression of related transporters during differentiation of human monocytes to macrophages. Whereas monocytes display a modest uptake of l-carnitine, GM-CSF-induced differentiation massively increased the saturable Na+-dependent uptake of l-carnitine. Kinetic and inhibition analyses demonstrate that in macrophage l-carnitine transport is mediated by a high-affinity component (Km ∼4 µM) that is identifiable with the operation of OCTN2 transporter and a low-affinity component (Km > 10 mM) that is identifiable with system A for neutral amino acids. Consistently, both SLC22A5/OCTN2 and SLC38A2/SNAT2 are induced during the differentiation of monocytes to macrophages at gene and protein levels. Elucidation of GM-CSF signaling demonstrates that the cytokine causes the activation of mTOR kinase, leading to the phosphorylation and activation of STAT3, which, in turn, is responsible for OCTN2 transcription. SLC22A5/OCTN2 therefore emerges as a novel member of the set of genes markers of macrophage differentiation.


Subject(s)
Carnitine/metabolism , Cell Differentiation , Macrophages/cytology , Macrophages/metabolism , Organic Cation Transport Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Biological Transport , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Kinetics , Models, Biological , Monocytes/cytology , Organic Cation Transport Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solute Carrier Family 22 Member 5 , Time Factors
15.
Biochim Biophys Acta ; 1858(2): 210-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26607009

ABSTRACT

Carnitine plays a physiologically important role in the ß-oxidation of fatty acids, facilitating the transport of long-chain fatty acids across the inner mitochondrial membrane. Distribution of carnitine within the body tissues is mainly performed by novel organic cation transporter (OCTN) family, including the isoforms OCTN1 (SLC22A4) and OCTN2 (SLC22A5) expressed in human. We performed here a characterization of carnitine transport in human airway epithelial cells A549, Calu-3, NCl-H441, and BEAS-2B, by means of an integrated approach combining data of mRNA/protein expression with the kinetic and inhibition analyses of L-[(3)H]carnitine transport. Carnitine uptake was strictly Na(+)-dependent in all cell models. In A549 and BEAS-2B cells, carnitine uptake was mediated by one high-affinity component (Km<2 µM) identifiable with OCTN2. In both these cell models, indeed, carnitine uptake was maximally inhibited by betaine and strongly reduced by SLC22A5/OCTN2 silencing. Conversely, Calu-3 and NCl-H441 exhibited both a high (Km~20 µM) and a low affinity (Km>1 mM) transport component. While the high affinity component is identifiable with OCTN2, the low affinity uptake is mediated by ATB(0,+), a Na(+), and Cl(-)-coupled transport system for neutral and cationic amino acids, as demonstrated by the inhibition by leucine and arginine, as well as by SLC6A14/ATB(0,+) silencing. The presence of this transporter leads to a massive accumulation of carnitine inside the cells and may be of peculiar relevance in pathologic conditions of carnitine deficiency, such as those associated to OCTN2 defects.


Subject(s)
Carnitine/metabolism , Epithelial Cells/metabolism , Organic Cation Transport Proteins/metabolism , Respiratory Mucosa/metabolism , Amino Acid Transport Systems , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Biological Transport, Active/physiology , Carnitine/genetics , Cell Line, Tumor , Epithelial Cells/cytology , Gene Expression Regulation/physiology , Humans , Organic Cation Transport Proteins/genetics , Respiratory Mucosa/cytology , Solute Carrier Family 22 Member 5 , Symporters
16.
Biochim Biophys Acta ; 1848(7): 1563-72, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25883089

ABSTRACT

Organic cation transporters (OCT1-3) mediate the transport of organic cations including inhaled drugs across the cell membrane, although their role in lung epithelium hasn't been well understood yet. We address here the expression and functional activity of OCT1-3 in human airway epithelial cells A549, Calu-3 and NCl-H441. Kinetic and inhibition analyses, employing [(3)H]1-methyl-4-phenylpyridinium (MPP+) as substrate, and the compounds quinidine, prostaglandine E2 (PGE2) and corticosterone as preferential inhibitors of OCT1, OCT2, and OCT3, respectively, have been performed. A549 cells present a robust MPP+ uptake mediated by one high-affinity component (Km~50µM) which is identifiable with OCT3. Corticosterone, indeed, completely inhibits MPP+ transport, while quinidine and PGE2 are inactive and SLC22A3/OCT3 silencing with siRNA markedly lowers MPP+ uptake. Conversely, Calu-3 exhibits both a high (Km<20µM) and a low affinity (Km>0.6mM) transport components, referable to OCT3 and OCT1, respectively, as demonstrated by the inhibition analysis performed at proper substrate concentrations and confirmed by the use of specific siRNA. These transporters are active also when cells are grown under air-liquid interface (ALI) conditions. Only a very modest saturable MPP+ uptake is measurable in NCl-H441 cells and the inhibitory effect of quinidine points to OCT1 as the subtype functionally involved in this model. Finally, the characterization of MPP+ transport in human bronchial BEAS-2B cells suggests that OCT1 and OCT3 are operative. These findings could help to identify in vitro models to be employed for studies concerning the specific involvement of each transporter in drug transportation.


Subject(s)
Epithelial Cells/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/metabolism , 1-Methyl-4-phenylpyridinium/metabolism , 1-Methyl-4-phenylpyridinium/pharmacokinetics , Biological Transport/drug effects , Biological Transport/genetics , Cell Line , Cell Line, Tumor , Corticosterone/pharmacology , Dinoprostone/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics , Lung/cytology , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 1/genetics , Organic Cation Transporter 2 , Quinidine/pharmacology , RNA Interference , Time Factors
17.
Xenobiotica ; 45(8): 693-710, 2015.
Article in English | MEDLINE | ID: mdl-25733029

ABSTRACT

1. The metabolism of CHF 6001, a novel PDE4 inhibitor, was determined in vitro in mouse, rat, dog, monkey and human microsomes and hepatocytes and in vivo in plasma, urine, feces and bile of rats after intravenous and intratracheal administration. 2. The behavior of CHF 6001 in microsomes and hepatocytes changed across species. CYP3A4/5 isoenzymes were identified to be the primary enzymes responsible for the metabolism of CHF 6001 in human liver microsomes. 3. In the rat, CHF 6001 was found extensively metabolized in urine, feces and bile, but not in plasma, where CHF 6001 was the main compound present. The metabolite profiles were different in the four biological matrices from both qualitative and quantitative point of view. 4. CHF 6001 was metabolized through hydrolysis with the formation of the alcohol CHF 5956, loss of a chlorine atom, loss of the N-oxide, hydroxylation, loss of the cyclopropylmethyl group in the alcohol moiety, conjugation with glucuronic acid, glutathione and cysteine-glycine. 5. The major metabolite present in the bile was isolated and characterized by nuclear magnetic resonance analysis. It derived from CHF 6001 through contraction of the pyridine-N-oxide ring to N-hydroxy pyrrole and conjugation with glucuronic acid.


Subject(s)
Bile/metabolism , Hepatocytes/metabolism , Microsomes, Liver/metabolism , Phosphodiesterase 4 Inhibitors/metabolism , Phosphoric Diester Hydrolases/metabolism , Pyrroles/metabolism , Administration, Intravenous , Animals , Chromatography, High Pressure Liquid , Dogs , Feces/chemistry , Haplorhini , Humans , Male , Mice , Phosphodiesterase 4 Inhibitors/administration & dosage , Phosphodiesterase 4 Inhibitors/blood , Phosphodiesterase 4 Inhibitors/urine , Rats , Species Specificity , Tandem Mass Spectrometry
18.
Pharmacol Res ; 55(4): 318-28, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17292621

ABSTRACT

Long-term use of non-steroidal anti-inflammatory drugs (NSAIDs) may delay or prevent the onset of Alzheimer's disease (AD). A subset of NSAIDs, including flurbiprofen, has been shown to selectively inhibit the production of beta-amyloid(1-42) (Abeta42), independently from their cyclooxygenase (COX) inhibiting activity. We evaluated the in vitro and in vivo profiles of CHF5022 and CHF5074, two flurbiprofen analogues. The in vitro Abeta inhibiting activity was evaluated in a human neuroglioma cell line (H4) carrying the double Swedish mutation (K595N/M596L) of the human amyloid precursor protein (APPsw). The in vitro anti-COX activity was evaluated using human recombinant enzymes isolated from transfected Sf-9 cells. The in vivo pharmacokinetic and pharmacodynamic profiles of the two compounds were evaluated in young APPsw transgenic mice (Tg2576) after oral gavage (100 or 300mgkg(-1) day(-1) for 4-5 days) and after medicated diet (375ppm for 4 weeks). R-Flurbiprofen was used as comparator. In vitro, CHF5022 and CHF5074 were found to be 3- and 7-fold more potent than R-flurbiprofen in inhibiting Abeta42 secretion (IC(50)s of 92, 40 and 268microM, respectively). Differently from R-flurbiprofen, CHF5022 and CHF5074 did not affect COX-1 (at 100microM) and COX-2 (at 300microM) activity. Similarly to R-flurbiprofen, no significant alteration in the expression profile of a subset of Notch intracellular domain-responsive genes was observed with either CHF5022 or CHF5074. In Tg2576 mice, CHF5022 was well tolerated when administered by oral gavage (100mgkg(-1) day(-1) for 5 days) or by medicated diet (56mg kg(-1) day(-1) for 4 weeks). R-Flurbiprofen was poorly tolerated in the diet (32mgkg(-1) day(-1)) with 55% of the animals dying during the first week of treatment. After 4-5 days of oral gavage, CHF5022 and CHF5074 plasma and brain levels at 3h were found to increase with the dose, leading to brain concentrations of about 10% and 5% of the corresponding plasma concentrations, respectively. In animals fed for 4 weeks with compound-supplemented diet, mean plasma (580microM) and brain (20microM) Cyrillic) concentrations of CHF5022 were 8 and 15 times higher than those of R-flurbiprofen. Plasma Abeta42 concentration was dose-dependently decreased by CHF5022 and CHF5074. Brain Abeta levels (formic acid-extractable) were not significantly affected by either compound, although Abeta42 levels tended to inversely correlate (P=0.105) with CHF5022 concentration in the brain. CHF5022 and CHF5074 thus appear to have a promising in vitro and in vivo profile. This warrants further evaluation of their long-term effects on Abeta brain pathology.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain/drug effects , Cyclopropanes/pharmacology , Flurbiprofen/analogs & derivatives , Flurbiprofen/pharmacology , Nootropic Agents/pharmacology , Peptide Fragments/metabolism , Administration, Oral , Amyloid beta-Protein Precursor/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 1/drug effects , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Cyclopropanes/administration & dosage , Cyclopropanes/pharmacokinetics , Dose-Response Relationship, Drug , Down-Regulation , Flurbiprofen/administration & dosage , Flurbiprofen/pharmacokinetics , Gene Expression/drug effects , Humans , Insecta/cytology , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation , Nootropic Agents/administration & dosage , Nootropic Agents/pharmacokinetics , Time Factors , Transfection
19.
J Med Chem ; 48(18): 5705-20, 2005 Sep 08.
Article in English | MEDLINE | ID: mdl-16134939

ABSTRACT

Flurbiprofen, a nonsteroidal antiinflammatory drug (NSAID), has been recently described to selectively inhibit beta-amyloid(1)(-)(42) (Abeta42) secretion, the most toxic component of the senile plaques present in the brain of Alzheimer patients. The use of this NSAID in Alzheimer's disease (AD) is hampered by a significant gastrointestinal toxicity associated with cyclooxygenase (COX) inhibition. New flurbiprofen analogues were synthesized, with the aim of increasing Abeta42 inhibitory potency while removing anti-COX activity. In vitro ADME developability parameters were taken into account in order to identify optimized compounds at an early stage of the project. Appropriate substitution patterns at the alpha position of flurbiprofen allowed for the complete removal of anti-COX activity, while modifications at the terminal phenyl ring resulted in increased inhibitory potency on Abeta42 secretion. In rats, some of the compounds appeared to be well absorbed after oral administration and to penetrate into the central nervous system. Studies in a transgenic mice model of AD showed that selected compounds significantly decreased plasma Abeta42 concentrations. These new flurbiprofen analogues represent potential drug candidates to be developed for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Flurbiprofen/analogs & derivatives , Flurbiprofen/chemical synthesis , Peptide Fragments/antagonists & inhibitors , Administration, Oral , Alzheimer Disease/blood , Alzheimer Disease/genetics , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blood-Brain Barrier/metabolism , Caco-2 Cells , Cell Line, Tumor , Cell Membrane Permeability , Cyclooxygenase Inhibitors/pharmacology , Flurbiprofen/pharmacology , Glioma , Humans , Immunoassay , In Vitro Techniques , Injections, Intravenous , Mice , Mice, Transgenic , Peptide Fragments/blood , Peptide Fragments/metabolism , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stereoisomerism , Structure-Activity Relationship
20.
Comb Chem High Throughput Screen ; 7(8): 745-56, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15578936

ABSTRACT

A cell-based assay using homogeneous time-resolved fluorescence has been developed for high throughput screening of putative beta-amyloid (Abeta)production inhibitors. In this assay, total Abeta is detected by simply adding two commercially available antibody complexes. The first was a biotinylated monoclonal antibody (4G8), specifically recognizing an epitope comprising the residues 17-24 of the Abetapeptide, complexed with europium cryptate-streptavidin conjugate. The second was a polyclonal antibody (BioS-N), raised against the N-terminus of the Abeta peptide, complexed with an allophycocyanin-anti rabbit antibody conjugate. Binding of the two complexes to the Abeta peptide brought europium cryptate (fluorescence donor) and allophycocyanin (fluorescence acceptor) into close proximity, consequently a fluorescent resonance energy transfer signal was produced upon excitation at 337 nm. The resulting fluorescence signal (665 nm) was then detected using a Discovery or a ViewLux reader. Detection of Abeta by the proposed method is possible at concentrations of approximately 1 nM. The method was employed for the detection of Abeta secreted from a stable transfected human neuroglioma cell line (H4) overexpressing a mutated form of the human amyloid precursor protein (APP695NL) and developed for robotic automation. At optimized conditions, signal-to-background ratios exceeding 5 and Z' factors around 0.7 were achieved in a 384-well format. High throughput screening of 56,913 potential Abeta production inhibitors led to identification of new non-cytotoxic and cell permeable compounds with potencies in the submicromolar range.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal , Cells, Cultured , Chromatography, Liquid , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Fluorescent Dyes , Humans , Immunoassay , Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...