1
|
Scarano N, Espinoza S, Brullo C, Cichero E. Computational Methods for the Discovery and Optimization of TAAR1 and TAAR5 Ligands. Int J Mol Sci 2024; 25:8226. [PMID: 39125796 PMCID: PMC11312273 DOI: 10.3390/ijms25158226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
G-protein-coupled receptors (GPCRs) represent a family of druggable targets when treating several diseases and continue to be a leading part of the drug discovery process. Trace amine-associated receptors (TAARs) are GPCRs involved in many physiological functions with TAAR1 having important roles within the central nervous system (CNS). By using homology modeling methods, the responsiveness of TAAR1 to endogenous and synthetic ligands has been explored. In addition, the discovery of different chemo-types as selective murine and/or human TAAR1 ligands has helped in the understanding of the species-specificity preferences. The availability of TAAR1-ligand complexes sheds light on how different ligands bind TAAR1. TAAR5 is considered an olfactory receptor but has specific involvement in some brain functions. In this case, the drug discovery effort has been limited. Here, we review the successful computational efforts developed in the search for novel TAAR1 and TAAR5 ligands. A specific focus on applying structure-based and/or ligand-based methods has been done. We also give a perspective of the experimental data available to guide the future drug design of new ligands, probing species-specificity preferences towards more selective ligands. Hints for applying repositioning approaches are also discussed.
Collapse
Affiliation(s)
- Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (C.B.)
| | - Stefano Espinoza
- Department of Health Sciences and Research Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), 28100 Novara, Italy;
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (C.B.)
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (C.B.)
| |
Collapse
|
2
|
Grossi G, Scarano N, Musumeci F, Tonelli M, Kanov E, Carbone A, Fossa P, Gainetdinov RR, Cichero E, Schenone S. Discovery of a Novel Chemo-Type for TAAR1 Agonism via Molecular Modeling. Molecules 2024; 29:1739. [PMID: 38675561 PMCID: PMC11052455 DOI: 10.3390/molecules29081739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The search for novel effective TAAR1 ligands continues to draw great attention due to the wide range of pharmacological applications related to TAAR1 targeting. Herein, molecular docking studies of known TAAR1 ligands, characterized by an oxazoline core, have been performed in order to identify novel promising chemo-types for the discovery of more active TAAR1 agonists. In particular, the oxazoline-based compound S18616 has been taken as a reference compound for the computational study, leading to the development of quite flat and conformationally locked ligands. The choice of a "Y-shape" conformation was suggested for the design of TAAR1 ligands, interacting with the protein cavity delimited by ASP103 and aromatic residues such as PHE186, PHE195, PHE268, and PHE267. The obtained results allowed us to preliminary in silico screen an in-house series of pyrimidinone-benzimidazoles (1a-10a) as a novel scaffold to target TAAR1. Combined ligand-based (LBCM) and structure based (SBCM) computational methods suggested the biological evaluation of compounds 1a-10a, leading to the identification of derivatives 1a-3a (hTAAR1 EC50 = 526.3-657.4 nM) as promising novel TAAR1 agonists.
Collapse
Affiliation(s)
- Giancarlo Grossi
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.G.); (N.S.); (F.M.); (M.T.); (A.C.); (P.F.); (S.S.)
| | - Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.G.); (N.S.); (F.M.); (M.T.); (A.C.); (P.F.); (S.S.)
| | - Francesca Musumeci
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.G.); (N.S.); (F.M.); (M.T.); (A.C.); (P.F.); (S.S.)
| | - Michele Tonelli
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.G.); (N.S.); (F.M.); (M.T.); (A.C.); (P.F.); (S.S.)
| | - Evgeny Kanov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia (R.R.G.)
- St. Petersburg University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anna Carbone
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.G.); (N.S.); (F.M.); (M.T.); (A.C.); (P.F.); (S.S.)
| | - Paola Fossa
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.G.); (N.S.); (F.M.); (M.T.); (A.C.); (P.F.); (S.S.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia (R.R.G.)
- St. Petersburg University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.G.); (N.S.); (F.M.); (M.T.); (A.C.); (P.F.); (S.S.)
| | - Silvia Schenone
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.G.); (N.S.); (F.M.); (M.T.); (A.C.); (P.F.); (S.S.)
| |
Collapse
|
3
|
Liao S, Pino MJ, Deleon C, Lindner-Jackson M, Wu C. Interaction analyses of hTAAR1 and mTAAR1 with antagonist EPPTB. Life Sci 2022; 300:120553. [PMID: 35452636 DOI: 10.1016/j.lfs.2022.120553] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022]
Abstract
Trace amine-associated receptor 1 (TAAR1) plays a critical role in regulating monoaminergic activity. EPPTB is the only known selective potent antagonist of the mouse (m) TAAR1 presently, while it was shown to be weak at antagonizing human (h) TAAR1. The lack of high-resolution structure of TAAR1 hinders the understanding of the differences in the interaction modes between EPPTB and m/hTARR1. The purpose of this study is to probe these interaction modes using homology modeling, molecular docking, molecular dynamics (MD) simulations, and molecular mechanics-generalized Born surface area (MM-GBSA) binding energy calculations. Eight populated conformers of hTAAR1-EPPTB complex were observed during the MD simulations and could be used in structure-based virtual screening in future. The MM-GBSA binding energy of hTAAR1-EPPTB complex (-96.5 kcal/mol) is larger than that of mTAAR1-EPPTB complex (-106.7 kcal/mol), which is consistent with the experimental finding that EPPTB has weaker binding affinity to hTAAR1. The several residues in binding site of hTAAR1 (F1544.56, T1945.42 and I2907.39) are different from these of mTAAR1 (Y1534.56, A1935.42 and Y2877.39), which might contribute to the binding affinity difference. Our docking analysis on another hTAAR1 antagonist Compound 3 has found that 1). this compound binds in different pockets of our mTAAR1 and hTAAR1 homology models with a slightly stronger binding affinity to hTAAR1; 2). both antagonists bind to a very similar pocket of hTAAR1.
Collapse
Affiliation(s)
- Siyan Liao
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Michael James Pino
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States of America
| | - Catherine Deleon
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States of America
| | - Maurice Lindner-Jackson
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States of America
| | - Chun Wu
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States of America.
| |
Collapse
|
4
|
Decker AM, Brackeen MF, Mohammadkhani A, Kormos CM, Hesk D, Borgland SL, Blough BE. Identification of a Potent Human Trace Amine-Associated Receptor 1 Antagonist. ACS Chem Neurosci 2022; 13:1082-1095. [PMID: 35325532 DOI: 10.1021/acschemneuro.2c00086] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human trace amine-associated receptor subtype 1 (hTAAR1) is a G protein-coupled receptor that has therapeutic potential for multiple diseases, including schizophrenia, drug addiction, and Parkinson's disease (PD). Although several potent agonists have been identified and have shown positive results in various clinical trials for schizophrenia, the discovery of potent hTAAR1 antagonists remains elusive. Herein, we report the results of structure-activity relationship studies that have led to the discovery of a potent hTAAR1 antagonist (RTI-7470-44, 34). RTI-7470-44 exhibited an IC50 of 8.4 nM in an in vitro cAMP functional assay, a Ki of 0.3 nM in a radioligand binding assay, and showed species selectivity for hTAAR1 over the rat and mouse orthologues. RTI-7470-44 displayed good blood-brain barrier permeability, moderate metabolic stability, and a favorable preliminary off-target profile. Finally, RTI-7470-44 increased the spontaneous firing rate of mouse VTA dopaminergic neurons and blocked the effects of the known TAAR1 agonist RO5166017. Collectively, this work provides a promising hTAAR1 antagonist probe that can be used to study TAAR1 pharmacology and the potential therapeutic role in hypodopaminergic diseases such as PD.
Collapse
Affiliation(s)
- Ann M. Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Marcus F. Brackeen
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Aida Mohammadkhani
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Chad M. Kormos
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - David Hesk
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Stephanie L. Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Bruce E. Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
5
|
Heffernan MLR, Herman LW, Brown S, Jones PG, Shao L, Hewitt MC, Campbell JE, Dedic N, Hopkins SC, Koblan KS, Xie L. Ulotaront: A TAAR1 Agonist for the Treatment of Schizophrenia. ACS Med Chem Lett 2022; 13:92-98. [PMID: 35047111 PMCID: PMC8762745 DOI: 10.1021/acsmedchemlett.1c00527] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/22/2021] [Indexed: 12/17/2022] Open
Abstract
![]()
Ulotaront (SEP-363856)
is a trace-amine associated receptor 1 (TAAR1)
agonist with 5-HT1A receptor agonist activity in Phase 3 clinical
development, with FDA Breakthrough Therapy Designation, for the treatment
of schizophrenia. TAAR1 is a G-protein-coupled receptor (GPCR) that
is expressed in cortical, limbic, and midbrain monoaminergic regions.
It is activated by endogenous trace amines, and is believed to play
an important role in modulating dopaminergic, serotonergic, and glutamatergic
circuitry. TAAR1 agonism data are reported herein for ulotaront and
its analogues in comparison to endogenous TAAR1 agonists. In addition,
a human TAAR1 homology model was built around ulotaront to identify
key interactions and attempt to better understand the scaffold-specific
TAAR1 agonism structure–activity relationships.
Collapse
Affiliation(s)
| | - Lee W. Herman
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - Scott Brown
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - Philip G. Jones
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - Liming Shao
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - Michael C. Hewitt
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - John E. Campbell
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - Nina Dedic
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - Seth C. Hopkins
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - Kenneth S. Koblan
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| | - Linghong Xie
- Sunovion Pharmaceuticals Inc, Marlborough, Massachusetts 01752, United States
| |
Collapse
|
6
|
Overlap and Specificity in the Substrate Spectra of Human Monoamine Transporters and Organic Cation Transporters 1, 2, and 3. Int J Mol Sci 2021; 22:ijms222312816. [PMID: 34884618 PMCID: PMC8657982 DOI: 10.3390/ijms222312816] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022] Open
Abstract
Human monoamine transporters (MATs) are cation transporters critically involved in neuronal signal transmission. While inhibitors of MATs have been intensively studied, their substrate spectra have received far less attention. Polyspecific organic cation transporters (OCTs), predominantly known for their role in hepatic and renal drug elimination, are also expressed in the central nervous system and might modulate monoaminergic signaling. Using HEK293 cells overexpressing MATs or OCTs, we compared uptake of 48 compounds, mainly phenethylamine and tryptamine derivatives including matched molecular pairs, across noradrenaline, dopamine and serotonin transporters and OCTs (1, 2, and 3). Generally, MATs showed surprisingly high transport activities for numerous analogs of neurotransmitters, but their substrate spectra were limited by molar mass. Human OCT2 showed the broadest substrate spectrum, and also the highest overlap with MATs substrates. Comparative kinetic analyses revealed that the radiotracer meta-iodobenzylguanidine had the most balanced uptake across all six transporters. Matched molecular pair analyses comparing MAT and OCT uptake using the same methodology could provide a better understanding of structural determinants for high cell uptake by MATs or OCTs. The data may result in a better understanding of pharmacokinetics and toxicokinetics of small molecular organic cations and, possibly, in the development of more specific radiotracers for MATs.
Collapse
|
7
|
Decker AM, Mathews KM, Blough BE, Gilmour BP. Validation of a High-Throughput Calcium Mobilization Assay for the Human Trace Amine-Associated Receptor 1. SLAS DISCOVERY 2020; 26:140-150. [PMID: 32734809 DOI: 10.1177/2472555220945279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The human trace amine-associated receptor 1 (hTAAR1) is a G protein-coupled receptor (GPCR) that is widely expressed in monoaminergic nuclei in the central nervous system and has therapeutic potential for multiple diseases, including drug addiction and schizophrenia. Thus, identification of novel hTAAR1 ligands is critical to advancing our knowledge of hTAAR1 function and to the development of therapeutics for a wide range of diseases. Herein we describe the development of a robust, 3-addition high-throughput screening (HTS) calcium mobilization assay using stable CHO-Gαq16-hTAAR1 cells, which functionally couple hTAAR1 to the promiscuous Gαq16 protein and thus allow signal transduction to occur through mobilization of internal calcium. Our previously established 96-well hTAAR1 assay was first miniaturized to the 384-well format and optimized to provide an assay with a Z' factor of 0.84, which is indicative of a robust HTS assay. Using the 3-addition protocol, 22,000 compounds were screened and yielded a ~1% agonist hit rate and a ~0.2% antagonist hit rate. Of the antagonist hits, two confirmed hits are the most potent hTAAR1 antagonists identified to date (IC50 = 206 and 281 nM). While scientists have been studying hTAAR1 for years, the lack of suitable hTAAR1 antagonists has been a major roadblock for studying the basic pharmacology of hTAAR1. Thus, these new ligands will serve as valuable tools to study hTAAR1-mediated signaling mechanisms, therapeutic potential, and in vivo functions.
Collapse
Affiliation(s)
- Ann M Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Kelly M Mathews
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Brian P Gilmour
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| |
Collapse
|
8
|
Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. ARAB J CHEM 2019. [DOI: 10.1016/j.arabjc.2016.09.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
9
|
Khan MF, Verma G, Akhtar W, Shaquiquzzaman M, Akhter M, Rizvi MA, Alam MM. Pharmacophore modeling, 3D-QSAR, docking study and ADME prediction of acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents. ARAB J CHEM 2019. [DOI: 10.1016/j.arabjc.2016.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
10
|
Lu H, Wang N, Li X, Huang Y, Wang J, Zhao Q. Identification of New Potent Human Uncoupling Protein 1 (UCP1) Agonists Using Virtual Screening and
in vitro
Approaches. Mol Inform 2019; 38:e1900030. [PMID: 31264791 DOI: 10.1002/minf.201900030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Hong‐Yuan Lu
- Department of Life Science and BiochemistryShenyang Pharmaceutical University Shenyang 110016 China
- Department of PharmacyGeneral Hospital of Northern Theater Command Shenyang 110840 China
| | - Nan Wang
- Department of Life Science and BiochemistryShenyang Pharmaceutical University Shenyang 110016 China
| | - Xiang Li
- Department of Life Science and BiochemistryShenyang Pharmaceutical University Shenyang 110016 China
| | - Yuan Huang
- Department of Life Science and BiochemistryShenyang Pharmaceutical University Shenyang 110016 China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of EducationShenyang Pharmaceutical University Shenyang 110016 China
| | - Qing‐Chun Zhao
- Department of Life Science and BiochemistryShenyang Pharmaceutical University Shenyang 110016 China
- Department of PharmacyGeneral Hospital of Northern Theater Command Shenyang 110840 China
| |
Collapse
|
11
|
D'Andrea G, Pizzolato G, Gucciardi A, Stocchero M, Giordano G, Baraldi E, Leon A. Different Circulating Trace Amine Profiles in De Novo and Treated Parkinson's Disease Patients. Sci Rep 2019; 9:6151. [PMID: 30992490 PMCID: PMC6467876 DOI: 10.1038/s41598-019-42535-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/29/2019] [Indexed: 12/18/2022] Open
Abstract
Early diagnosis of Parkinson’s disease (PD) remains a challenge to date. New evidence highlights the potential clinical value of circulating trace amines (TAs) in early-stage PD and their involvement in disease progression. A new ultra performance chromatography mass spectrometry (UPLC-MS/MS) method was developed to quantify plasmatic TAs, and the catecholamines and indolamines pertaining to the same biochemical pathways. Three groups of subjects were recruited: 21 de novo, drug untreated, PD patients, 27 in treatment PD patients and 10 healthy subjects as controls. Multivariate and univariate data analyses were applied to reveal metabolic changes among the groups in attempt to discover new putative markers for early PD detection and disease progression. Different circulating levels of tyrosine (p = 0.002), tyramine (p < 0.001), synephrine (p = 0.015), norepinephrine (p = 0.012), metanephrine (p = 0.001), β-phenylethylamine (p = 0.001) and serotonin (p = 0.006) were found among the three groups. While tyramine behaves as a putative biomarker for early-stage PD (AUC = 0.90) tyramine, norepinephrine, and tyrosine appear to act as biomarkers of disease progression (AUC > 0.75). The findings of this pilot cross-sectional study suggest that biochemical anomalies of the aminergic and indolic neurotransmitters occur in PD patients. Compounds within the TAs family may constitute putative markers for early stage detection and progression of PD.
Collapse
Affiliation(s)
| | - Gilberto Pizzolato
- Department of Medical Sciences, Neurology Unit, University of Trieste, Trieste, Italy
| | - Antonina Gucciardi
- Mass Spectrometry and Metabolomic Laboratory, Women's and Children's Health Department, University of Padova, Padova, Italy. .,Fondazione Istituto di Ricerca Pediatrica Cittàdella Speranza, Padova, Italy.
| | - Matteo Stocchero
- Mass Spectrometry and Metabolomic Laboratory, Women's and Children's Health Department, University of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Cittàdella Speranza, Padova, Italy
| | - Giuseppe Giordano
- Mass Spectrometry and Metabolomic Laboratory, Women's and Children's Health Department, University of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Cittàdella Speranza, Padova, Italy
| | - Eugenio Baraldi
- Mass Spectrometry and Metabolomic Laboratory, Women's and Children's Health Department, University of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Cittàdella Speranza, Padova, Italy
| | - Alberta Leon
- Research and Innovation (R&I Genetics) s.r.l., Padova, Italy
| |
Collapse
|
12
|
Walcher L, Budde C, Böhm A, Reinach PS, Dhandapani P, Ljubojevic N, Schweiger MW, von der Waydbrink H, Reimers I, Köhrle J, Mergler S. TRPM8 Activation via 3-Iodothyronamine Blunts VEGF-Induced Transactivation of TRPV1 in Human Uveal Melanoma Cells. Front Pharmacol 2018. [DOI: 10.3389/fphar.2018.01234 ecollection 2018.pmid: 30483120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2022] Open
|
13
|
Walcher L, Budde C, Böhm A, Reinach PS, Dhandapani P, Ljubojevic N, Schweiger MW, von der Waydbrink H, Reimers I, Köhrle J, Mergler S. TRPM8 Activation via 3-Iodothyronamine Blunts VEGF-Induced Transactivation of TRPV1 in Human Uveal Melanoma Cells. Front Pharmacol 2018; 9:1234. [PMID: 30483120 PMCID: PMC6243059 DOI: 10.3389/fphar.2018.01234] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/11/2018] [Indexed: 01/17/2023] Open
Abstract
In human uveal melanoma (UM), tumor enlargement is associated with increases in aqueous humor vascular endothelial growth factor-A (VEGF-A) content that induce neovascularization. 3-Iodothyronamine (3-T1AM), an endogenous thyroid hormone metabolite, activates TRP melastatin 8 (TRPM8), which blunts TRP vanilloid 1 (TRPV1) activation by capsaicin (CAP) in human corneal, conjunctival epithelial cells, and stromal cells. We compare here the effects of TRPM8 activation on VEGF-induced transactivation of TRPV1 in an UM cell line (92.1) with those in normal primary porcine melanocytes (PM) since TRPM8 is upregulated in melanoma. Fluorescence Ca2+-imaging and planar patch-clamping characterized functional channel activities. CAP (20 μM) induced Ca2+ transients and increased whole-cell currents in both the UM cell line and PM whereas TRPM8 agonists, 100 μM menthol and 20 μM icilin, blunted such responses in the UM cells. VEGF (10 ng/ml) elicited Ca2+ transients and augmented whole-cell currents, which were blocked by capsazepine (CPZ; 20 μM) but not by a highly selective TRPM8 blocker, AMTB (20 μM). The VEGF-induced current increases were not augmented by CAP. Both 3-T1AM (1 μM) and menthol (100 μM) increased the whole-cell currents, whereas 20 μM AMTB blocked them. 3-T1AM exposure suppressed both VEGF-induced Ca2+ transients and increases in underlying whole-cell currents. Taken together, functional TRPM8 upregulation in UM 92.1 cells suggests that TRPM8 is a potential drug target for suppressing VEGF induced increases in neovascularization and UM tumor growth since TRPM8 activation blocked VEGF transactivation of TRPV1.
Collapse
Affiliation(s)
- Lia Walcher
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Clara Budde
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Arina Böhm
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peter S Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | | | - Nina Ljubojevic
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Markus W Schweiger
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Henriette von der Waydbrink
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ilka Reimers
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Mergler
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
14
|
Lam VM, Mielnik CA, Baimel C, Beerepoot P, Espinoza S, Sukhanov I, Horsfall W, Gainetdinov RR, Borgland SL, Ramsey AJ, Salahpour A. Behavioral Effects of a Potential Novel TAAR1 Antagonist. Front Pharmacol 2018; 9:953. [PMID: 30233365 PMCID: PMC6131539 DOI: 10.3389/fphar.2018.00953] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022] Open
Abstract
The trace amine associated receptor 1 (TAAR1) is a G-protein coupled receptor expressed in the monoaminergic regions of the brain, and represents a potential novel therapeutic target for the treatment of neurological disorders. While selective agonists for TAAR1 have been successfully identified, only one high affinity TAAR1 antagonist has been described thus far. We previously identified four potential low potency TAAR1 antagonists through an in silico screen on a TAAR1 homology model. One of the identified antagonists (compound 22) was predicted to have favorable physicochemical properties, which would allow the drug to cross the blood brain barrier. In vivo studies were therefore carried out and showed that compound 22 potentiates amphetamine- and cocaine-mediated locomotor activity. Furthermore, electrophysiology experiments demonstrated that compound 22 increased firing of dopamine neurons similar to EPPTB, the only known TAAR1 antagonist. In order to assess whether the effects of compound 22 were mediated through TAAR1, experiments were carried out on TAAR1-KO mice. The results showed that compound 22 is able to enhance amphetamine- and cocaine-mediated locomotor activity, even in TAAR1-KO mice, suggesting that the in vivo effects of this compound are not mediated by TAAR1. In collaboration with Psychoactive Drug Screening Program, we attempted to determine the targets for compound 22. Psychoactive Drug Screening Program (PDSP) results suggested several potential targets for compound 22 including, the dopamine, norepinephrine and serotonin transporters; as well as sigma 1 and 2 receptors. Our follow-up studies using heterologous cell systems showed that the dopamine transporter is not a target of compound 22. Therefore, the biological target of compound 22 mediating its psychoactive effects still remains unknown.
Collapse
Affiliation(s)
- Vincent M Lam
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Catharine A Mielnik
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Corey Baimel
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Pieter Beerepoot
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Boston Children's Hospital, F.M. Kirby Center for Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Stefano Espinoza
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Ilya Sukhanov
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.,Pavlov First Saint Petersburg State Medical University, Valdman Institute of Pharmacology, Saint Petersburg, Russia
| | - Wendy Horsfall
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Amy J Ramsey
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Ali Salahpour
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Guariento S, Tonelli M, Espinoza S, Gerasimov AS, Gainetdinov RR, Cichero E. Rational design, chemical synthesis and biological evaluation of novel biguanides exploring species-specificity responsiveness of TAAR1 agonists. Eur J Med Chem 2018; 146:171-184. [DOI: 10.1016/j.ejmech.2018.01.059] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 11/28/2022]
|
16
|
Berry MD, Gainetdinov RR, Hoener MC, Shahid M. Pharmacology of human trace amine-associated receptors: Therapeutic opportunities and challenges. Pharmacol Ther 2017; 180:161-180. [DOI: 10.1016/j.pharmthera.2017.07.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Targeting species-specific trace amine-associated receptor 1 ligands: to date perspective of the rational drug design process. Future Med Chem 2017; 9:1507-1527. [DOI: 10.4155/fmc-2017-0044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
G-protein-coupled receptors represent main targets of several clinically relevant drugs, playing nowadays a leading part for further drug discovery process. Trace amine-associated receptor's family (TAARs) assumed an intriguing role as druggable target in medicinal chemistry, being TAAR1 the most investigated. Indeed, related ligands proved to be intertwined in several circuits involved in pathological pathways or therapeutic routes. Herein, we highlight relevant efforts in the search of novel agonists, focusing on responsiveness featured by different chemotypes toward rodent and human TAAR1, in order to explore species-specificity preferences. We also discuss the main strategies guiding so far the design of new TAAR1 agonists, giving a perspective of the structure-based methodologies aimed at deriving new insights for more potent and selective derivatives.
Collapse
|
18
|
Liu S, Ji S, Yu ZJ, Wang HL, Cheng X, Li WJ, Jing L, Yu Y, Chen Q, Yang LL, Li GB, Wu Y. Structure-based discovery of new selective small-molecule sirtuin 5 inhibitors. Chem Biol Drug Des 2017; 91:257-268. [PMID: 28756638 DOI: 10.1111/cbdd.13077] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 07/10/2017] [Accepted: 07/15/2017] [Indexed: 02/05/2023]
Abstract
Human sirtuin 5 (SIRT5) is a protein deacylase regulating metabolic pathways and stress responses and is implicated in metabolism-related diseases. Small-molecule inhibitors for SIRT5 are sought as chemical tools and potential therapeutics. Herein, we proposed a customized virtual screening approach targeting catalytically important and unique residues Tyr102 and Arg105 of SIRT5. Of the 20 tested virtual screening hits, six compounds displayed marked inhibitory activities against SIRT5. For the hit compound 19, a series of newly synthesized (E)-2-cyano-N-phenyl-3-(5-phenylfuran-2-yl)acrylamide derivatives/analogues were carried out structure-activity relationship analyses, resulting in new more potent inhibitors, among which 37 displayed the most potent inhibition to SIRT5 with an IC50 value of 5.59 ± 0.75 μM. The biochemical studies revealed that 37 likely acts via competitive inhibition with the succinyl-lysine substrate, rather than the NAD+ cofactor, and it manifested substantial selectivity for SIRT5 over SIRT2 and SIRT6. This study will aid further efforts to develop new selective SIRT5 inhibitors as tools and therapeutics.
Collapse
Affiliation(s)
- Sha Liu
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Sen Ji
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Zhu-Jun Yu
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hua-Li Wang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xu Cheng
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Wei-Jian Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Li Jing
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yamei Yu
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qiang Chen
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ling-Ling Yang
- College of Food and Bioengineering, Xihua University, Sichuan, China
| | - Guo-Bo Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yong Wu
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, and Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Kadam AA, Ellern A, Stanley LM. Enantioselective, Palladium-Catalyzed Conjugate Additions of Arylboronic Acids to Form Bis-benzylic Quaternary Stereocenters. Org Lett 2017; 19:4062-4065. [DOI: 10.1021/acs.orglett.7b01825] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Abhishek A. Kadam
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| | - Arkady Ellern
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| | - Levi M. Stanley
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
20
|
Tonelli M, Espinoza S, Gainetdinov RR, Cichero E. Novel biguanide-based derivatives scouted as TAAR1 agonists: Synthesis, biological evaluation, ADME prediction and molecular docking studies. Eur J Med Chem 2017; 127:781-792. [DOI: 10.1016/j.ejmech.2016.10.058] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 12/14/2022]
|
21
|
Berry MD, Hart S, Pryor AR, Hunter S, Gardiner D. Pharmacological characterization of a high-affinity p-tyramine transporter in rat brain synaptosomes. Sci Rep 2016; 6:38006. [PMID: 27901065 PMCID: PMC5128819 DOI: 10.1038/srep38006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/03/2016] [Indexed: 11/22/2022] Open
Abstract
p-Tyramine is an archetypal member of the endogenous family of monoamines known as trace amines, and is one of the endogenous agonists for trace amine-associated receptor (TAAR)1. While much work has focused on the function of TAAR1, very little is known about the regulation of the endogenous agonists. We have previously reported that p-tyramine readily crosses lipid bilayers and that its release from synaptosomes is non-exocytotic. Such release, however, showed characteristics of modification by one or more transporters. Here we provide the first characterization of such a transporter. Using frontal cortical and striatal synaptosomes we show that p-tyramine passage across synaptosome membranes is not modified by selective inhibition of either the dopamine, noradrenaline or 5-HT transporters. In contrast, inhibition of uptake-2 transporters significantly slowed p-tyramine re-uptake. Using inhibitors of varying selectivity, we identify Organic Cation Transporter 2 (OCT2; SLC22A2) as mediating high affinity uptake of p-tyramine at physiologically relevant concentrations. Further, we confirm the presence of OCT2 protein in synaptosomes. These results provide the first identification of a high affinity neuronal transporter for p-tyramine, and also confirm the recently described localization of OCT2 in pre-synaptic terminals.
Collapse
Affiliation(s)
- Mark D Berry
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Shannon Hart
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Anthony R Pryor
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Samantha Hunter
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Danielle Gardiner
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| |
Collapse
|
22
|
Cichero E, Tonelli M. New insights into the structure of the trace amine-associated receptor 2: Homology modelling studies exploring the binding mode of 3-iodothyronamine. Chem Biol Drug Des 2016; 89:790-796. [DOI: 10.1111/cbdd.12903] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 10/19/2016] [Accepted: 10/24/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Elena Cichero
- Department of Pharmacy; University of Genoa; Genoa Italy
| | | |
Collapse
|
23
|
Inhibition of Urease by Disulfiram, an FDA-Approved Thiol Reagent Used in Humans. Molecules 2016; 21:molecules21121628. [PMID: 27898047 PMCID: PMC6274061 DOI: 10.3390/molecules21121628] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 12/22/2022] Open
Abstract
Urease is a nickel-dependent amidohydrolase that catalyses the decomposition of urea into carbamate and ammonia, a reaction that constitutes an important source of nitrogen for bacteria, fungi and plants. It is recognized as a potential antimicrobial target with an impact on medicine, agriculture, and the environment. The list of possible urease inhibitors is continuously increasing, with a special interest in those that interact with and block the flexible active site flap. We show that disulfiram inhibits urease in Citrullus vulgaris (CVU), following a non-competitive mechanism, and may be one of this kind of inhibitors. Disulfiram is a well-known thiol reagent that has been approved by the FDA for treatment of chronic alcoholism. We also found that other thiol reactive compounds (l-captopril and Bithionol) and quercetin inhibits CVU. These inhibitors protect the enzyme against its full inactivation by the thiol-specific reagent Aldrithiol (2,2'-dipyridyl disulphide, DPS), suggesting that the three drugs bind to the same subsite. Enzyme kinetics, competing inhibition experiments, auto-fluorescence binding experiments, and docking suggest that the disulfiram reactive site is Cys592, which has been proposed as a "hinge" located in the flexible active site flap. This study presents the basis for the use of disulfiram as one potential inhibitor to control urease activity.
Collapse
|
24
|
Chiellini G, Nesi G, Sestito S, Chiarugi S, Runfola M, Espinoza S, Sabatini M, Bellusci L, Laurino A, Cichero E, Gainetdinov RR, Fossa P, Raimondi L, Zucchi R, Rapposelli S. Hit-to-Lead Optimization of Mouse Trace Amine Associated Receptor 1 (mTAAR1) Agonists with a Diphenylmethane-Scaffold: Design, Synthesis, and Biological Study. J Med Chem 2016; 59:9825-9836. [DOI: 10.1021/acs.jmedchem.6b01092] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Giulia Nesi
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Simona Sestito
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Sara Chiarugi
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | | | - Stefano Espinoza
- Department
of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | | | | | - Annunziatina Laurino
- Department
of NEUROFARBA, Section of Pharmacology, University of Florence, 50139 Florence, Italy
| | - Elena Cichero
- Department
of Pharmacy, University of Genoa, 16126, Genoa, Italy
| | - Raul R. Gainetdinov
- Institute
of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
- Skolkovo Institute of Science and Technology (Skoltech), Skolkovo, Moscow Region, 143025, Russia
| | - Paola Fossa
- Department
of Pharmacy, University of Genoa, 16126, Genoa, Italy
| | - Laura Raimondi
- Department
of NEUROFARBA, Section of Pharmacology, University of Florence, 50139 Florence, Italy
| | - Riccardo Zucchi
- Department
of Pathology, University of Pisa, 56126 Pisa, Italy
| | | |
Collapse
|
25
|
Synthesis of Pyrazole-Thiobarbituric Acid Derivatives: Antimicrobial Activity and Docking Studies. Molecules 2016; 21:molecules21101337. [PMID: 27735850 PMCID: PMC6274314 DOI: 10.3390/molecules21101337] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/25/2016] [Accepted: 09/30/2016] [Indexed: 11/17/2022] Open
Abstract
A one-pot reaction was described that results in various pyrazole-thiobarbituric acid derivatives as new pharmacophore agents. These new heterocycles were synthesized in high yields with a broad substrate scope under mild reaction conditions in water mediated by NHEt2. The molecular structures of the synthesized compounds were assigned based on different spectroscopic techniques. The new compounds were evaluated for their antibacterial and antifungal activity. Compounds 4h and 4l were the most active compounds against C. albicans with MIC = 4 µg/L. Compound 4c exhibited the best activity against S. aureus and E. faecalis with MIC = 16 µg/L. However, compounds 4l and 4o were the most active against B. subtilis with MIC = 16 µg/L. Molecular docking studies for the final compounds and standard drugs were performed using the OpenEye program.
Collapse
|
26
|
Meduru H, Wang YT, Tsai JJP, Chen YC. Finding a Potential Dipeptidyl Peptidase-4 (DPP-4) Inhibitor for Type-2 Diabetes Treatment Based on Molecular Docking, Pharmacophore Generation, and Molecular Dynamics Simulation. Int J Mol Sci 2016; 17:ijms17060920. [PMID: 27304951 PMCID: PMC4926453 DOI: 10.3390/ijms17060920] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/30/2016] [Accepted: 06/02/2016] [Indexed: 12/28/2022] Open
Abstract
Dipeptidyl peptidase-4 (DPP-4) is the vital enzyme that is responsible for inactivating intestinal peptides glucagon like peptide-1 (GLP-1) and Gastric inhibitory polypeptide (GIP), which stimulates a decline in blood glucose levels. The aim of this study was to explore the inhibition activity of small molecule inhibitors to DPP-4 following a computational strategy based on docking studies and molecular dynamics simulations. The thorough docking protocol we applied allowed us to derive good correlation parameters between the predicted binding affinities (pKi) of the DPP-4 inhibitors and the experimental activity values (pIC50). Based on molecular docking receptor-ligand interactions, pharmacophore generation was carried out in order to identify the binding modes of structurally diverse compounds in the receptor active site. Consideration of the permanence and flexibility of DPP-4 inhibitor complexes by means of molecular dynamics (MD) simulation specified that the inhibitors maintained the binding mode observed in the docking study. The present study helps generate new information for further structural optimization and can influence the development of new DPP-4 inhibitors discoveries in the treatment of type-2 diabetes.
Collapse
Affiliation(s)
- Harika Meduru
- Department of Bioinformatics and Medical Engineering, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan.
| | - Yeng-Tseng Wang
- Department of Biochemistry, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan.
| | - Jeffrey J P Tsai
- Department of Bioinformatics and Medical Engineering, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan.
| | - Yu-Ching Chen
- Department of Bioinformatics and Medical Engineering, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan.
| |
Collapse
|
27
|
Chen KB, Chen KC, Chang YL, Chang KL, Chang PC, Chang TT, Chen YC. In Silico Investigation of Traditional Chinese Medicine for Potential Lead Compounds as SPG7 Inhibitors against Coronary Artery Disease. Molecules 2016; 21:molecules21050588. [PMID: 27164068 PMCID: PMC6273800 DOI: 10.3390/molecules21050588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/22/2016] [Accepted: 04/29/2016] [Indexed: 11/16/2022] Open
Abstract
Coronary artery disease (CAD) is the most common cause of heart attack and the leading cause of mortality in the world. It is associated with mitochondrial dysfunction and increased level of reactive oxygen species production. According to the Ottawa Heart Genomics Study genome-wide association study, a recent research identified that Q688 spastic paraplegia 7 (SPG7) variant is associated with CAD as it bypasses the regulation of tyrosine phosphorylation of AFG3L2 and enhances the processing and maturation of SPG7 protein. This study aims to identify potential compounds isolated from Traditional Chinese Medicines (TCMs) as potential lead compounds for paraplegin (SPG7) inhibitors. For the crystallographic structure of paraplegin, the disordered disposition of key amino acids in the binding site was predicted using the PONDR-Fit protocol before virtual screening. The TCM compounds saussureamine C and 3-(2-carboxyphenyl)-4(3H)-quinazolinone, have potential binding affinities with stable H-bonds and hydrophobic contacts with key residues of paraplegin. A molecular dynamics simulation was performed to validate the stability of the interactions between each candidate and paraplegin under dynamic conditions. Hence, we propose these compounds as potential candidates as lead drug from the compounds isolated from TCM for further study in drug development process with paraplegin protein for coronary artery disease.
Collapse
Affiliation(s)
- Kuen-Bao Chen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan.
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan.
- Department of Anesthesiology, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Kuan-Chung Chen
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| | - Ya-Lin Chang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan.
| | - Kun-Lung Chang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan.
- Department of Pharmacy, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan.
| | - Pei-Chun Chang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan.
| | - Tung-Ti Chang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan.
- Department of Chinese Pediatrics, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Yu-Chian Chen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan.
- Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung 40402, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan.
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
28
|
Simmler LD, Buchy D, Chaboz S, Hoener MC, Liechti ME. In Vitro Characterization of Psychoactive Substances at Rat, Mouse, and Human Trace Amine-Associated Receptor 1. J Pharmacol Exp Ther 2016; 357:134-44. [PMID: 26791601 DOI: 10.1124/jpet.115.229765] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/19/2016] [Indexed: 11/22/2022] Open
Abstract
Trace amine-associated receptor 1 (TAAR1) has been implicated in the behavioral effects of amphetamine-type stimulant drugs in rodents. TAAR1 has also been suggested as a target for novel medications to treat psychostimulant addiction. We previously reported that binding affinities at TAAR1 can differ between structural analogs of psychostimulants, and species differences have been observed. In this study, we complement our previous findings with additional substances and the determination of functional activation potencies. In summary, we present here pharmacological in vitro profiles of 101 psychoactive substances at human, rat, and mouse TAAR1. p-Tyramine, β-phenylethylamine, and tryptamine were included as endogenous comparator compounds. Functional cAMP measurements and radioligand displacement assays were conducted with human embryonic kidney 293 cells that expressed human, rat, or mouse TAAR1. Most amphetamines, phenethylamine, and aminoindanes exhibited potentially physiologically relevant rat and mouse TAAR1 activation (EC50 < 5 µM) and showed full or partial (Emax < 80%) agonist properties. Cathinone derivatives, including mephedrone and methylenedioxypyrovalerone, exhibited weak (EC50 = 5-10 µM) to negligible (EC50 > 10 µM) binding properties at TAAR1. Pipradrols, including methylphenidate, exhibited no affinity for TAAR1. We found considerable species differences in activity at TAAR1 among the highly active ligands, with a rank order of rat > mouse > human. This characterization provides information about the pharmacological profile of psychoactive substances. The species differences emphasize the relevance of clinical studies to translationally complement rodent studies on the role of TAAR1 activity for psychoactive substances.
Collapse
Affiliation(s)
- Linda D Simmler
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland (L.D.S., M.E.L.); and Neuroscience Research, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (D.B., S.C., M.C.H)
| | - Danièle Buchy
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland (L.D.S., M.E.L.); and Neuroscience Research, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (D.B., S.C., M.C.H)
| | - Sylvie Chaboz
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland (L.D.S., M.E.L.); and Neuroscience Research, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (D.B., S.C., M.C.H)
| | - Marius C Hoener
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland (L.D.S., M.E.L.); and Neuroscience Research, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (D.B., S.C., M.C.H)
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland (L.D.S., M.E.L.); and Neuroscience Research, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (D.B., S.C., M.C.H)
| |
Collapse
|
29
|
Cichero E, Espinoza S, Tonelli M, Franchini S, Gerasimov AS, Sorbi C, Gainetdinov RR, Brasili L, Fossa P. A homology modelling-driven study leading to the discovery of the first mouse trace amine-associated receptor 5 (TAAR5) antagonists. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00490j] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The computational study here proposed allowed us to discovery for the first time two TAAR5 antagonist, selective over the TAAR1 receptor.
Collapse
Affiliation(s)
- Elena Cichero
- Department of Pharmacy
- University of Genoa
- 3, 16132 Genoa
- Italy
| | - Stefano Espinoza
- Department of Neuroscience and Brain Technologies
- Istituto Italiano di Tecnologia
- Genoa
- Italy
| | - Michele Tonelli
- Department of Pharmacy
- University of Genoa
- 3, 16132 Genoa
- Italy
| | - Silvia Franchini
- Department of Life Sciences
- University of Modena and Reggio Emilia
- 41125 Modena
- Italy
| | | | - Claudia Sorbi
- Department of Life Sciences
- University of Modena and Reggio Emilia
- 41125 Modena
- Italy
| | - Raul R. Gainetdinov
- Department of Neuroscience and Brain Technologies
- Istituto Italiano di Tecnologia
- Genoa
- Italy
- Institute of Translational Biomedicine
| | - Livio Brasili
- Department of Life Sciences
- University of Modena and Reggio Emilia
- 41125 Modena
- Italy
| | - Paola Fossa
- Department of Pharmacy
- University of Genoa
- 3, 16132 Genoa
- Italy
| |
Collapse
|
30
|
Liu HC, Tang SZ, Lu S, Ran T, Wang J, Zhang YM, Xu AY, Lu T, Chen YD. Studies on [5,6]-Fused Bicyclic Scaffolds Derivatives as Potent Dual B-RafV600E/KDR Inhibitors Using Docking and 3D-QSAR Approaches. Int J Mol Sci 2015; 16:24451-74. [PMID: 26501259 PMCID: PMC4632759 DOI: 10.3390/ijms161024451] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 09/22/2015] [Accepted: 10/07/2015] [Indexed: 01/07/2023] Open
Abstract
Research and development of multi-target inhibitors has attracted increasing attention as anticancer therapeutics. B-RafV600E synergistically works with vascular endothelial growth factor receptor 2 (KDR) to promote the occurrence and progression of cancers, and the development of dual-target drugs simultaneously against these two kinds of kinase may offer a better treatment advantage. In this paper, docking and three-dimensional quantitative structure activity relationship (3D-QSAR) studies were performed on a series of dual B-Raf/KDR inhibitors with a novel hinge-binding group, [5,6]-fused bicyclic scaffold. Docking studies revealed optimal binding conformations of these compounds interacting with both B-Raf and KDR. Based on these conformations, comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) 3D-QSAR models were constructed, and the best CoMFA (q²=0.542, r²=0.989 for B-Raf; q²=0.768, r²=0.991 for KDR) and CoMSIA models (q²=0.519, r²=0.992 for B-Raf; q²=0.849, r²=0.993 for KDR) were generated. Further external validations confirmed their predictability, yielding satisfactory correlation coefficients (r²pred=0.764 (CoMFA), r²pred=0.841 (CoMSIA) for B-Raf, r²pred=0.912 (CoMFA), r²pred=0.846 (CoMSIA) for KDR, respectively). Through graphical analysis and comparison on docking results and 3D-QSAR contour maps, key amino acids that affect the ligand-receptor interactions were identified and structural features influencing the activities were discussed. New potent derivatives were designed, and subjected to preliminary pharmacological evaluation. The study may offer useful references for the modification and development of novel dual B-Raf/KDR inhibitors.
Collapse
Affiliation(s)
- Hai-Chun Liu
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
| | - San-Zhi Tang
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
| | - Ting Ran
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
| | - Jian Wang
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
| | - Yan-Min Zhang
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
| | - An-Yang Xu
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
| | - Tao Lu
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211169, China.
| | - Ya-Dong Chen
- School of Science, China Pharmaceutical University, Nanjing 211169, China.
| |
Collapse
|
31
|
Lam VM, Espinoza S, Gerasimov AS, Gainetdinov RR, Salahpour A. In-vivo pharmacology of Trace-Amine Associated Receptor 1. Eur J Pharmacol 2015; 763:136-42. [DOI: 10.1016/j.ejphar.2015.06.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/26/2015] [Accepted: 06/15/2015] [Indexed: 11/30/2022]
|
32
|
Lam VM, Rodríguez D, Zhang T, Koh EJ, Carlsson J, Salahpour A. Discovery of trace amine-associated receptor 1 ligands by molecular docking screening against a homology model. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00400d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An in silico screen of a TAAR1 homology model identifies novel ligands.
Collapse
Affiliation(s)
- V. M. Lam
- Department of Pharmacology and Toxicology
- University of Toronto
- Toronto
- Canada
| | - D. Rodríguez
- Science for Life Laboratory
- Department of Biochemistry and Biophysics and Center for Biomembrane Research
- Stockholm University
- SE-106 91 Stockholm
- Sweden
| | - T. Zhang
- Department of Pharmacology and Toxicology
- University of Toronto
- Toronto
- Canada
| | - E. J. Koh
- Department of Pharmacology and Toxicology
- University of Toronto
- Toronto
- Canada
| | - J. Carlsson
- Science for Life Laboratory
- Department of Medicinal Chemistry
- BMC
- Uppsala University
- SE-751 23 Uppsala
| | - A. Salahpour
- Department of Pharmacology and Toxicology
- University of Toronto
- Toronto
- Canada
| |
Collapse
|
33
|
Cichero E, Menozzi G, Guariento S, Fossa P. Ligand-based homology modelling of the human CB2 receptor SR144528 antagonist binding site: a computational approach to explore the 1,5-diaryl pyrazole scaffold. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00333d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
SR144528 docking mode into the LBHM of the human CB2 receptor antagonist binding site.
Collapse
Affiliation(s)
- Elena Cichero
- Department of Pharmacy
- University of Genoa
- 3 - 16132 Genoa
- Italy
| | - Giulia Menozzi
- Department of Pharmacy
- University of Genoa
- 3 - 16132 Genoa
- Italy
| | - Sara Guariento
- Department of Pharmacy
- University of Genoa
- 3 - 16132 Genoa
- Italy
| | - Paola Fossa
- Department of Pharmacy
- University of Genoa
- 3 - 16132 Genoa
- Italy
| |
Collapse
|