12 results on '"Juan A. Sánchez-Arias"'
Search Results
2. Targeting CB1 and GPR55 Endocannabinoid Receptors as a Potential Neuroprotective Approach for Parkinson’s Disease
- Author
-
Gemma Navarro, José Luis Lanciego, Rafael Franco, Juan A. Sánchez-Arias, Julen Oyarzabal, Eva Martínez-Pinilla, Alberto J Rico, and David Aguinaga
- Subjects
0301 basic medicine ,Agonist ,Cannabinoid receptor ,medicine.drug_class ,Cell growth ,Chemistry ,Neuroscience (miscellaneous) ,Substantia nigra ,Endocannabinoid system ,Neuroprotection ,Cell biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,030104 developmental biology ,0302 clinical medicine ,Neurology ,medicine ,Receptor ,030217 neurology & neurosurgery ,G protein-coupled receptor - Abstract
Cannabinoid CB1 receptors (CB1R) and the GPR55 receptor are expressed in striatum and are potential targets in the therapy of Parkinson's disease (PD), one of the most prevalent neurodegenerative diseases in developed countries. The aim of this paper was to address the potential of ligands acting on those receptors to prevent the action of a neurotoxic agent, MPP+, that specifically affects neurons of the substantia nigra due to uptake via the dopamine DAT transporter. The SH-SY5Y cell line model was used as it expresses DAT and, therefore, is able to uptake MPP+ that inhibits complex I of the respiratory mitochondrial chain and leads to cell death. Cells were transfected with cDNAs coding for either or both receptors. Receptors in cotransfected cells formed heteromers as indicated by the in situ proximity ligation assays. Cell viability was assayed by oxygen rate consumption and by the bromide-based MTT method. Assays of neuroprotection using two concentrations of MPP+ showed that cells expressing receptor heteromers were more resistant to the toxic effect. After correction by effects on cell proliferation, the CB1R antagonist, SR141716, afforded an almost full neuroprotection in CB1R-expressing cells even when a selective agonist, ACEA, was present. In contrast, SR141716 was not effective in cells expressing CB1/GPR55 heteromeric complexes. In addition, an agonist of GPR55, CID1792197, did not enhance neuroprotection in GPR55-expressing cells. These results show that neurons expressing heteromers are more resistant to cell death but question the real usefulness of CB1R, GPR55, and their heteromers as targets to afford PD-related neuroprotection.
- Published
- 2019
- Full Text
- View/download PDF
3. Design, synthesis, biological evaluation and in vivo testing of dual phosphodiesterase 5 (PDE5) and histone deacetylase 6 (HDAC6)-selective inhibitors for the treatment of Alzheimer's disease
- Author
-
Ana Garcia-Osta, Irene de Miguel, Carolina García-Barroso, Julen Oyarzabal, Elena Sáez, Obdulia Rabal, Marta Pérez-González, Mar Cuadrado-Tejedor, Wu Wei, Maria Espelosin, Tan Haizhong, Ana Ugarte, Ander Estella-Hermoso de Mendoza, Susana Ursua, Juan A. Sánchez-Arias, and Xu Musheng
- Subjects
0301 basic medicine ,Gene isoform ,Disease ,Pharmacology ,Histone Deacetylase 6 ,Cell Line ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,In vivo ,Drug Discovery ,Humans ,Biological evaluation ,Cyclic Nucleotide Phosphodiesterases, Type 5 ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Organic Chemistry ,Pde5 inhibition ,General Medicine ,Phosphodiesterase 5 Inhibitors ,HDAC6 ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Design synthesis ,Drug Design ,cGMP-specific phosphodiesterase type 5 ,Neuroglia ,030217 neurology & neurosurgery - Abstract
We have identified chemical probes that act as dual phosphodiesterase 5 (PDE5) and histone deacetylase 6 (HDAC6)-selective inhibitors (>1 log unit difference versus class I HDACs) to decipher the contribution of HDAC isoforms to the positive impact of dual-acting PDE5 and HDAC inhibitors on mouse models of Alzheimer's disease (AD) and fine-tune this systems therapeutics approach. Structure- and knowledge-based approaches led to the design of first-in-class molecules with the desired target compound profile: dual PDE5 and HDAC6-selective inhibitors. Compound 44b, which fulfilled the biochemical, functional and ADME-Tox profiling requirements and exhibited adequate pharmacokinetic properties, was selected as pharmacological tool compound and tested in a mouse model of AD (Tg2576) in vivo.
- Published
- 2018
- Full Text
- View/download PDF
4. Impact of Scaffold Exploration on Novel Dual-Acting Histone Deacetylases and Phosphodiesterase 5 Inhibitors for the Treatment of Alzheimer’s Disease
- Author
-
Ana Ugarte, Julen Oyarzabal, Tan Haizhong, Mar Cuadrado-Tejedor, Obdulia Rabal, Xu Musheng, Elena Sáez, Wu Wei, Irene de Miguel, Juan A. Sánchez-Arias, Ana Garcia-Osta, Marta Pérez-González, Susana Ursua, and Maria Espelosin
- Subjects
Models, Molecular ,0301 basic medicine ,Scaffold ,Physiology ,Sildenafil ,Cognitive Neuroscience ,Pharmacology ,Biochemistry ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Adenosine Triphosphate ,Alzheimer Disease ,In vivo ,medicine ,Animals ,Humans ,Cyclic GMP ,Cell Line, Transformed ,Cyclic Nucleotide Phosphodiesterases, Type 5 ,Neurons ,biology ,Chemistry ,Cell Biology ,General Medicine ,Phosphodiesterase 5 Inhibitors ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Histone ,3',5'-Cyclic-AMP Phosphodiesterases ,Vardenafil ,cGMP-specific phosphodiesterase type 5 ,Toxicity ,Leukocytes, Mononuclear ,Microsomes, Liver ,biology.protein ,Histone deacetylase ,Neuroglia ,medicine.drug - Abstract
A novel systems therapeutics approach, involving simultaneous inhibition of phosphodiesterase 5 (PDE5) and histone deacetylase (HDAC), has been validated as a potentially novel therapeutic strategy for the treatment of Alzheimer's disease (AD). First-in-class dual inhibitors bearing a sildenafil core have been very recently reported, and the lead molecule 7 has proven this strategy in AD animal models. Because scaffolds may play a critical role in primary activities and ADME-Tox profiling as well as on intellectual property, we have explored alternative scaffolds (vardenafil- and tadalafil-based cores) and evaluated their impact on critical parameters such as primary activities, permeability, toxicity, and in vivo (pharmacokinetics and functional response in hippocampus) to identify a potential alternative lead molecule bearing a different chemotype for in vivo testing.
- Published
- 2016
- Full Text
- View/download PDF
5. Discovery of in Vivo Chemical Probes for Treating Alzheimer's Disease: Dual Phosphodiesterase 5 (PDE5) and Class I Histone Deacetylase Selective Inhibitors
- Author
-
Elena Sáez, Julen Oyarzabal, Ander Estella-Hermoso de Mendoza, Maria Espelosin, Xu Musheng, Mar Cuadrado-Tejedor, Ana Ugarte, Ana Garcia-Osta, Susana Ursua, Obdulia Rabal, Marta Pérez-González, Juan A. Sánchez-Arias, Irene de Miguel, Wu Wei, Carolina García-Barroso, and Tan Haizhong
- Subjects
Gene isoform ,Physiology ,Cognitive Neuroscience ,Biochemistry ,Histone Deacetylases ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Alzheimer Disease ,Ic50 values ,Animals ,Humans ,030304 developmental biology ,Cyclic Nucleotide Phosphodiesterases, Type 5 ,0303 health sciences ,biology ,Chemistry ,Target engagement ,Acetylation ,Cell Biology ,General Medicine ,Phosphodiesterase 5 Inhibitors ,Histone Deacetylase Inhibitors ,Disease Models, Animal ,Histone ,cGMP-specific phosphodiesterase type 5 ,biology.protein ,Histone deacetylase ,030217 neurology & neurosurgery - Abstract
In order to determine the contributions of histone deacetylase (HDAC) isoforms to the beneficial effects of dual phosphodiesterase 5 (PDE5) and pan-HDAC inhibitors on in vivo models of Alzheimer’s disease (AD), we have designed, synthesized, and tested novel chemical probes with the desired target compound profile of PDE5 and class I HDAC selective inhibitors. Compared to previous hydroxamate-based series, these molecules exhibit longer residence times on HDACs. In this scenario, shorter or longer preincubation times may have a significant impact on the IC50 values of these compounds and therefore on their corresponding selectivity profiles on the different HDAC isoforms. On the other hand, different chemical series have been explored and, as expected, some pairwise comparisons show a clear impact of the scaffold on biological responses (e.g., 35a vs 40a). The lead identification process led to compound 29a, which shows an adequate ADME-Tox profile and in vivo target engagement (histone acetylation and cA...
- Published
- 2018
6. Discovery of Reversible DNA Methyltransferase and Lysine Methyltransferase G9a Inhibitors with Antitumoral in Vivo Efficacy
- Author
-
Yingying Liu, Edurne San José-Enériz, Leire Garate, Jose A. Martinez-Climent, Raúl F. Pérez, Elena Sáez, Pablo Santamarina, Xabier Agirre, María José García-Barchino, Estíbaliz Miranda, Sergio Roa, Raquel Ordoñez, Amaia Vilas-Zornoza, Irene de Miguel, Juan A. Sánchez-Arias, Ana Ugarte, Musheng Xu, Mario F. Fraga, Obdulia Rabal, Julen Oyarzabal, Felipe Prosper, and Wei Wu
- Subjects
0301 basic medicine ,Methyltransferase ,Protein Conformation ,Lysine ,Antineoplastic Agents ,DNA methyltransferase ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Histocompatibility Antigens ,Drug Discovery ,Animals ,Humans ,Epigenetics ,Enzyme Inhibitors ,DNA Modification Methylases ,biology ,Chemistry ,Methylation ,Histone-Lysine N-Methyltransferase ,Xenograft Model Antitumor Assays ,In vitro ,Molecular Docking Simulation ,030104 developmental biology ,Histone ,Biochemistry ,030220 oncology & carcinogenesis ,Drug Design ,biology.protein ,Molecular Medicine - Abstract
Using knowledge- and structure-based approaches, we designed and synthesized reversible chemical probes that simultaneously inhibit the activity of two epigenetic targets, histone 3 lysine 9 methyltransferase (G9a) and DNA methyltransferases (DNMT), at nanomolar ranges. Enzymatic competition assays confirmed our design strategy: substrate competitive inhibitors. Next, an initial exploration around our hit 11 was pursued to identify an adequate tool compound for in vivo testing. In vitro treatment of different hematological neoplasia cell lines led to the identification of molecules with clear antiproliferative efficacies (GI50 values in the nanomolar range). On the basis of epigenetic functional cellular responses (levels of lysine 9 methylation and 5-methylcytosine), an acceptable therapeutic window (around 1 log unit) and a suitable pharmacokinetic profile, 12 was selected for in vivo proof-of-concept ( Nat. Commun. 2017, 8, 15424). Herein, 12 achieved a significant in vivo efficacy: 70% overall tumor g...
- Published
- 2018
7. Glucocerebrosidase mutations and synucleinopathies. Potential role of sterylglucosides and relevance of studying both GBA1 and GBA2 genes
- Author
-
Rafael Franco, Juan A. Sánchez-Arias, Gemma Navarro, José L. Lanciego, and Universitat de Barcelona
- Subjects
0301 basic medicine ,Parkinson's disease ,Lisosomes ,Mini Review ,alpha-synuclein ,GBA1 ,Neuroscience (miscellaneous) ,Mitochondrion ,Biology ,lcsh:RC321-571 ,lcsh:QM1-695 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,neurodegenerative disease ,0302 clinical medicine ,Lysosome ,Malaltia de Parkinson ,medicine ,Genetics ,GCase ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Gene ,Synucleinopathies ,Alpha-synuclein ,Dementia with Lewy bodies ,lcsh:Human anatomy ,medicine.disease ,Cell biology ,Enzymes ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,lysosome ,Parkinson’s disease ,Anatomy ,Enzims ,dementia with Lewy bodies ,Lysosomes ,Glucocerebrosidase ,030217 neurology & neurosurgery ,Genètica ,Neuroscience - Abstract
Gaucher’s disease (GD) is the most prevalent lysosomal storage disorder. GD is caused by homozygous mutations of the GBA1 gene, which codes for beta-glucocerebrosidase (GCase). Although GD primarily affects peripheral tissues, the presence of neurological symptoms has been reported in several GD subtypes. GBA1 mutations have recently deserved increased attention upon the demonstration that both homo- and heterozygous GBA1 mutations represent the most important genetic risk factor for the appearance of synucleinopathies like Parkinson’s disease (PD) and dementia with Lewy bodies (LBD). Although reduced GCase activity leads to alpha-synuclein aggregation, the mechanisms sustaining a role for GCase in alpha-synuclein homeostasis still remain largely unknown. Furthermore, the role to be played by impairment in the physiological function of endoplasmic reticulum, mitochondria and other subcellular membranous components is currently under investigation. Here we focus on the impact of GCase loss-of-function that impact on the levels of sterylglucosides, molecules that are known to trigger a PD-related synucleinopathy upon administration in rats. Moreover, the concurrence of another gene also coding for an enzyme with GCase activity (GBA2 gene) should also be taken into consideration, bearing in mind that in addition to a hydrolytic function, both GCases also share transglycosylation as a second catalytic activity. Accordingly, sterylglycoside levels should also be considered to further assess their impact on the neurodegenerative process. In this regard—and besides GBA1 genotyping—we suggest that screening for GBA2 mutations should be considered, together with analytical measurements of cholesterol glycosides in body fluids, as biomarkers for both PD risk and disease progression.
- Published
- 2018
8. Detailed Exploration around 4-Aminoquinolines Chemical Space to Navigate the Lysine Methyltransferase G9a and DNA Methyltransferase Biological Spaces
- Author
-
Edurne San José-Enériz, Leire Garate, Estíbaliz Miranda, Wei Wu, Xabier Agirre, Irene de Miguel, Jose A. Martinez-Climent, Yingying Liu, Juan A. Sánchez-Arias, Elena Sáez, Musheng Xu, Julen Oyarzabal, Obdulia Rabal, Felipe Prosper, and Sergio Roa
- Subjects
0301 basic medicine ,Methyltransferase ,Protein Conformation ,01 natural sciences ,DNA methyltransferase ,03 medical and health sciences ,Inhibitory Concentration 50 ,In vivo ,Cell Line, Tumor ,Histocompatibility Antigens ,Drug Discovery ,Humans ,Epigenetics ,DNA Modification Methylases ,Cell Proliferation ,chemistry.chemical_classification ,Methylation ,Histone-Lysine N-Methyltransferase ,Chemical space ,0104 chemical sciences ,Molecular Docking Simulation ,010404 medicinal & biomolecular chemistry ,030104 developmental biology ,Enzyme ,Biochemistry ,chemistry ,Drug Design ,DNMT1 ,Aminoquinolines ,Molecular Medicine - Abstract
Epigenetic regulators that exhibit aberrant enzymatic activities or expression profiles are potential therapeutic targets for cancers. Specifically, enzymes responsible for methylation at histone-3 lysine-9 (like G9a) and aberrant DNA hypermethylation (DNMTs) have been implicated in a number of cancers. Recently, molecules bearing a 4-aminoquinoline scaffold were reported as dual inhibitors of these targets and showed a significant in vivo efficacy in animal models of hematological malignancies. Here, we report a detailed exploration around three growing vectors born by this chemotype. Exploring this chemical space led to the identification of features to navigate G9a and DNMT1 biological spaces: not only their corresponding exclusive areas, selective compounds, but also common spaces. Thus, we identified from selective G9a and first-in-class DNMT1 inhibitors, >1 log unit between their IC50 values, with IC50 < 25 nM (e.g., 43 and 26, respectively) to equipotent inhibitors with IC50 < 50 nM for both target...
- Published
- 2018
9. Neuroprotective Effect of JZL184 in MPP+-Treated SH-SY5Y Cells Through CB2 Receptors
- Author
-
Rafael Franco, Estefanía Rojo-Bustamante, María S. Aymerich, Marta Celorrio, Carmen Molina, and Juan A. Sánchez-Arias
- Subjects
0301 basic medicine ,1-Methyl-4-phenylpyridinium ,SH-SY5Y ,Cannabinoid receptor ,Neurotoxins ,Neuroscience (miscellaneous) ,Pharmacology ,Biology ,Neuroprotection ,Amidohydrolases ,Receptor, Cannabinoid, CB2 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Piperidines ,Receptor, Cannabinoid, CB1 ,Cell Line, Tumor ,Cannabinoid receptor type 2 ,Humans ,Benzodioxoles ,Enzyme Inhibitors ,JZL184 ,MPTP ,Endocannabinoid system ,Monoacylglycerol Lipases ,Monoacylglycerol lipase ,Neuroprotective Agents ,030104 developmental biology ,Neurology ,chemistry ,lipids (amino acids, peptides, and proteins) ,030217 neurology & neurosurgery - Abstract
Growing evidence suggests that the endocannabinoid system plays a role in neuroprotection in Parkinson's disease. Recently, we have shown the neuroprotective effect of monoacylglycerol lipase (MAGL) inhibition with JZL184 in the chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model. However, further investigation is needed to determine the neuroprotective mechanisms of the endocannabinoid system on the nigrostriatal pathway. The aim of this work was to investigate whether the neuroprotective effect of JZL184 in mice could be extended to an in vitro cellular model to further understand the mechanism of action of the drug. The SH-SY5Y cell line was selected based on its dopaminergic-like phenotype and its susceptibility to 1-methyl-4-phenylpyridinium iodide (MPP(+)) toxicity. Furthermore, SH-SY5Y cells express both cannabinoid receptors, CB1 and CB2. The present study describes the neuroprotective effect of MAGL inhibition with JZL184 in SH-SY5Y cells treated with MPP(+). The effect of JZL184 in cell survival was blocked by AM630, a CB2 receptor antagonist, and it was mimicked with JWH133, a CB2 receptor agonist. Rimonabant, a CB1 receptor antagonist, did not affect JZL184-induced cell survival. These results demonstrate that the neuroprotective effect of MAGL inhibition with JZL184 described in animal models of Parkinson's disease could be extended to in vitro models such as SH-SY5Y cells treated with MPP(+). This represents a useful tool to study mechanisms of neuroprotection mediated by MAGL inhibition, and we provide evidence for the possible involvement of CB2 receptors in the improvement of cell survival.
- Published
- 2015
- Full Text
- View/download PDF
10. Design, Synthesis, and Biological Evaluation of Novel Matrix Metalloproteinase Inhibitors As Potent Antihemorrhagic Agents: From Hit Identification to an Optimized Lead
- Author
-
Haizhong Tan, M. Belzunce, Josune Orbe, Juan A. Sánchez-Arias, Wei Wu, Ana Ugarte, Musheng Xu, Jose A. Rodriguez, Agustina Salicio, Hongyu Ma, José A. Páramo, Obdulia Rabal, and Julen Oyarzabal
- Subjects
Male ,Models, Molecular ,Cell Membrane Permeability ,Matrix metalloproteinase inhibitor ,Stereochemistry ,medicine.medical_treatment ,Antihemorrhagic ,Hemorrhage ,Matrix Metalloproteinase Inhibitors ,Matrix metalloproteinase ,Pharmacology ,Hydroxamic Acids ,Mice ,Structure-Activity Relationship ,In vivo ,Bleeding time ,Drug Discovery ,Fibrinolysis ,medicine ,Animals ,Humans ,Structure–activity relationship ,ADME ,Molecular Structure ,medicine.diagnostic_test ,Chemistry ,Matrix Metalloproteinases ,Mice, Inbred C57BL ,Drug Design ,Benzamides ,Molecular Medicine - Abstract
Growing evidence suggests that matrix metalloproteinases (MMP) are involved in thrombus dissolution; then, considering that new therapeutic strategies are required for controlling hemorrhage, we hypothesized that MMP inhibition may reduce bleeding by delaying fibrinolysis. Thus, we designed and synthesized a novel series of MMP inhibitors to identify potential candidates for acute treatment of bleeding. Structure-based and knowledge-based strategies were utilized to design this novel chemical series, α-spiropiperidine hydroxamates, of potent and soluble (75 μg/mL) pan-MMP inhibitors. The initial hit, 12, was progressed to an optimal lead 19d. Racemic 19d showed a remarkable in vitro phenotypic response and outstanding in vivo efficacy; in fact, the mouse bleeding time at 1 mg/kg was 0.85 min compared to 29.28 min using saline. In addition, 19d displayed an optimal ADME and safety profile (e.g., no thrombus formation). Its corresponding enantiomers were separated, leading to the preclinical candidate 5 (described in Drug Annotations series, J. Med. Chem. 2015, ).
- Published
- 2015
- Full Text
- View/download PDF
11. Design, Synthesis, and Biological Evaluation of First-in-Class Dual Acting Histone Deacetylases (HDACs) and Phosphodiesterase 5 (PDE5) Inhibitors for the Treatment of Alzheimer's Disease
- Author
-
Ana Ugarte, Elena Sáez, Ana Garcia-Osta, Irene de Miguel, Marta Pérez-González, Wu Wei, Tan Haizhong, Ander Estella-Hermoso de Mendoza, Mar Cuadrado-Tejedor, Julen Oyarzabal, Obdulia Rabal, Susana Ursua, Xu Musheng, Juan A. Sánchez-Arias, Maria Espelosin, and Carolina García-Barroso
- Subjects
0301 basic medicine ,Models, Molecular ,Disease ,Pharmacology ,CREB ,Histone Deacetylases ,Cell Line ,03 medical and health sciences ,Mice ,In vivo ,Alzheimer Disease ,Drug Discovery ,Animals ,Humans ,Cyclic AMP Response Element-Binding Protein ,Cells, Cultured ,Cyclic Nucleotide Phosphodiesterases, Type 5 ,biology ,Chemistry ,Acetylation ,Phosphodiesterase 5 Inhibitors ,In vitro ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Histone ,cGMP-specific phosphodiesterase type 5 ,Drug Design ,biology.protein ,Molecular Medicine ,Phosphorylation - Abstract
Simultaneous inhibition of phosphodiesterase 5 (PDE5) and histone deacetylases (HDAC) has recently been validated as a potentially novel therapeutic approach for Alzheimer's disease (AD). To further extend this concept, we designed and synthesized the first chemical series of dual acting PDE5 and HDAC inhibitors, and we validated this systems therapeutics approach. Following the implementation of structure- and knowledge-based approaches, initial hits were designed and were shown to validate our hypothesis of dual in vitro inhibition. Then, an optimization strategy was pursued to obtain a proper tool compound for in vivo testing in AD models. Initial hits were translated into molecules with adequate cellular functional responses (histone acetylation and cAMP/cGMP response element-binding (CREB) phosphorylation in the nanomolar range), an acceptable therapeutic window (1 log unit), and the ability to cross the blood-brain barrier, leading to the identification of 7 as a candidate for in vivo proof-of-concept testing ( Cuadrado-Tejedor, M.; Garcia-Barroso, C.; Sánchez-Arias, J. A.; Rabal, O.; Mederos, S.; Ugarte, A.; Franco, R.; Segura, V.; Perea, G.; Oyarzabal, J.; Garcia-Osta, A. Neuropsychopharmacology 2016 , in press, doi: 10.1038/npp.2016.163 ).
- Published
- 2016
12. Two Affinity Sites of the Cannabinoid Subtype 2 Receptor Identified by a Novel Homogeneous Binding Assay
- Author
-
Eva Martínez-Pinilla, Irene de Miguel, Obdulia Rabal, Marta Zamarbide, Gemma Navarro, Rafael Franco, Juan A. Sánchez-Arias, Irene Reyes-Resina, José L. Lanciego, and Julen Oyarzabal
- Subjects
0301 basic medicine ,Cannabinoid receptor ,Indoles ,medicine.medical_treatment ,Ligands ,Chrysenes ,Receptors, G-Protein-Coupled ,Receptor, Cannabinoid, CB2 ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,medicine ,Humans ,Binding site ,Receptor ,Protein Structure, Quaternary ,Receptors, Cannabinoid ,Fluorescent Dyes ,Pharmacology ,Binding Sites ,Chemistry ,Ligand binding assay ,Endocannabinoid system ,Molecular Docking Simulation ,030104 developmental biology ,HEK293 Cells ,GPR55 ,Biochemistry ,Molecular Medicine ,GPR18 ,Biological Assay ,Cannabinoid ,Protein Multimerization ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Endocannabinoids act on G protein-coupled receptors that are considered potential targets for a variety of diseases. There are two different cannabinoid receptor types: ligands for cannabinoid type 2 receptors (CB2Rs) show more promise than those for cannabinoid type 1 receptors (CB1Rs) because they lack psychotropic actions. However, the complex pharmacology of these receptors, coupled with the lipophilic nature of ligands, is delaying the translational success of medications targeting the endocannabinoid system. We here report the discovery and synthesis of a fluorophore-conjugated CB2R-selective compound, CM-157 (3-[[4-[2-tert-butyl-1-(tetrahydropyran-4-ylmethyl)benzimidazol-5-yl]sulfonyl-2-pyridyl]oxy]propan-1-amine), which was useful for pharmacological characterization of CB2R by using a time-resolved fluorescence resonance energy transfer assay. This methodology does not require radiolabeled compounds and may be undertaken in homogeneous conditions and in living cells (i.e., without the need to isolate receptor-containing membranes). The affinity of the labeled compound was similar to that of the unlabeled molecule. Time-resolved fluorescence resonance energy transfer assays disclosed a previously unreported second affinity site and showed conformational changes in CB2R forming receptor heteromers with G protein-coupled receptor GPR55, a receptor for l-α-lysophosphatidylinositol. The populations displaying subnanomolar and nanomolar affinities were undisclosed in competitive assays using a well known cannabinoid receptor ligand, AM630 (1-[2-(morpholin-4-yl)ethyl]-2-methyl-3-(4-methoxybenzoyl)-6-iodoindole), and TH-chrysenediol, not previously tested on binding to cannabinoid receptors. Variations in binding parameters upon formation of dimers with GPR55 may reflect decreases in binding sites or alterations of the quaternary structure of the macromolecular G protein-coupled receptor complexes. In summary, the homogeneous binding assay described here may serve to better characterize agonist binding to CB2R and to identify specific properties of CB2R on living cells.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.