1
|
Tosso RD, Zarycz MNC, Schiel A, Goicoechea Moro L, Baldoni HA, Angelina E, Enriz RD. Evaluating the conformational space of the active site of D 2 dopamine receptor. Scope and limitations of the standard docking methods. J Comput Chem 2022; 43:1298-1312. [PMID: 35638694 DOI: 10.1002/jcc.26938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/11/2022] [Accepted: 05/08/2022] [Indexed: 11/05/2022]
Abstract
We report here for the first time the potential energy surfaces (PES) of phenyletilamine (PEA) and meta-tyramine (m-OH-PEA) at the D2 dopamine receptor (D2DR) binding site. PESs not only allow us to observe all the critical points of the surface (minimums, maximums, and transition states), but also to note the ease or difficulty that each local minima have for their conformational inter-conversions and therefore know the conformational flexibility that these ligands have in their active sites. Taking advantage of possessing this valuable information, we analyze how accurate a standard docking study is in these cases. Our results indicate that although we have to be careful in how to carry out this type of study and to consider performing some extra-simulations, docking calculations can be satisfactory. In order to analyze in detail the different molecular interactions that are stabilizing the different ligand-receptor (L-R) complexes, we carried out quantum theory of atoms in molecules (QTAIM) computations and NMR shielding calculations. Although some of these techniques are a bit tedious and require more computational time, our results demonstrate the importance of performing computational simulations using different types of combined techniques (docking/MD/hybrid QM-MM/QTAIM and NMR shielding calculations) in order to obtain more accurate results. Our results allow us to understand in details the molecular interactions stabilizing and destabilizing the different L-R complexes reported here. Thus, the different activities observed for dopamine (DA), m-OH-PEA, and PEA can be clearly explained at molecular level.
Collapse
Affiliation(s)
- Rodrigo D Tosso
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Argentina
| | - M Natalia C Zarycz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Argentina
| | - Ayelén Schiel
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Argentina
| | - Luisa Goicoechea Moro
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Argentina
| | - Héctor A Baldoni
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis; Instituto de Matemáticas, San Luis, Argentina
| | - Emilio Angelina
- Laboratorio de Estructura Molecular y Propiedades, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Instituto de Química Básica y Aplicada, Corrientes, Argentina
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas, San Luis, Argentina
| |
Collapse
|
2
|
Ågren R, Sahlholm K. Evidence for Two Modes of Binding of the Negative Allosteric Modulator SB269,652 to the Dopamine D2 Receptor. Biomedicines 2021; 10:biomedicines10010022. [PMID: 35052702 PMCID: PMC8772941 DOI: 10.3390/biomedicines10010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
SB269,652 has been described as the first negative allosteric modulator (NAM) of the dopamine D2 receptor (D2R), however, the binding mode and allosteric mechanism of action of this ligand remain incompletely understood. SB269,652 comprises an orthosteric, primary pharmacophore and a secondary (or allosteric) pharmacophore joined by a hydrophilic cyclohexyl linker and is known to form corresponding interactions with the orthosteric binding site (OBS) and the secondary binding pocket (SBP) in the D2R. Here, we observed a surprisingly low potency of SB269,652 to negatively modulate the D2R-mediated activation of G protein-coupled inward-rectifier potassium channels (GIRK) and decided to perform a more detailed investigation of the interaction between dopamine and SB269,652. The results indicated that the SB269,652 inhibitory potency is increased 6.6-fold upon ligand pre-incubation, compared to the simultaneous co-application with dopamine. Mutagenesis experiments implicated both S193 in the OBS and E95 in the SBP in the effect of pre-application. The present findings extend previous knowledge about how SB269,652 competes with dopamine at the D2R and may be useful for the development of novel D2R ligands, such as antipsychotic drug candidates.
Collapse
Affiliation(s)
- Richard Ågren
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
- Correspondence: (R.Å.); (K.S.)
| | - Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
- Wallenberg Centre for Molecular Medicine, Department of Integrative Medical Biology, Umeå University, 90187 Umea, Sweden
- Correspondence: (R.Å.); (K.S.)
| |
Collapse
|
3
|
Stepniewski TM, Mancini A, Ågren R, Torrens-Fontanals M, Semache M, Bouvier M, Sahlholm K, Breton B, Selent J. Mechanistic insights into dopaminergic and serotonergic neurotransmission - concerted interactions with helices 5 and 6 drive the functional outcome. Chem Sci 2021; 12:10990-11003. [PMID: 34522296 PMCID: PMC8386650 DOI: 10.1039/d1sc00749a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/15/2021] [Indexed: 01/14/2023] Open
Abstract
Brain functions rely on neurotransmitters that mediate communication between billions of neurons. Disruption of this communication can result in a plethora of psychiatric and neurological disorders. In this work, we combine molecular dynamics simulations, live-cell biosensor and electrophysiological assays to investigate the action of the neurotransmitter dopamine at the dopaminergic D2 receptor (D2R). The study of dopamine and closely related chemical probes reveals how neurotransmitter binding translates into the activation of distinct subsets of D2R effectors (i.e.: Gi2, GoB, Gz and β-arrestin 2). Ligand interactions with key residues in TM5 (S5.42) and TM6 (H6.55) in the D2R binding pocket yield a dopamine-like coupling signature, whereas exclusive TM5 interaction is typically linked to preferential G protein coupling (in particular GoB) over β-arrestin. Further experiments for serotonin receptors indicate that the reported molecular mechanism is shared by other monoaminergic neurotransmitter receptors. Ultimately, our study highlights how sequence variation in position 6.55 is used by nature to fine-tune β-arrestin recruitment and in turn receptor signaling and internalization of neurotransmitter receptors.
Collapse
Affiliation(s)
- Tomasz Maciej Stepniewski
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM) - Pompeu Fabra University (UPF) Dr Aiguader 88 Barcelona E-08003 Spain
- InterAx Biotech AG, PARK InnovAARE 5234 Villigen Switzerland
| | - Arturo Mancini
- Domain Therapeutics NA Inc 7171 Frederick-Banting Saint-Laurent (QC) H4S 1Z9 Canada
| | - Richard Ågren
- Department of Neuroscience, Karolinska Institute Stockholm Sweden
| | - Mariona Torrens-Fontanals
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM) - Pompeu Fabra University (UPF) Dr Aiguader 88 Barcelona E-08003 Spain
| | - Meriem Semache
- Domain Therapeutics NA Inc 7171 Frederick-Banting Saint-Laurent (QC) H4S 1Z9 Canada
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Université de Montréal Montreal QC H3C 3J7 Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal Montréal Québec H3T 1J4 Canada
| | - Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institute Stockholm Sweden
- Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University 90187 Umeå Sweden
| | - Billy Breton
- Domain Therapeutics NA Inc 7171 Frederick-Banting Saint-Laurent (QC) H4S 1Z9 Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal Montréal Québec H3T 1J4 Canada
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM) - Pompeu Fabra University (UPF) Dr Aiguader 88 Barcelona E-08003 Spain
| |
Collapse
|
4
|
Dopamine D 2 Receptor Agonist Binding Kinetics-Role of a Conserved Serine Residue. Int J Mol Sci 2021; 22:ijms22084078. [PMID: 33920848 PMCID: PMC8071183 DOI: 10.3390/ijms22084078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 01/03/2023] Open
Abstract
The forward (kon) and reverse (koff) rate constants of drug–target interactions have important implications for therapeutic efficacy. Hence, time-resolved assays capable of measuring these binding rate constants may be informative to drug discovery efforts. Here, we used an ion channel activation assay to estimate the kons and koffs of four dopamine D2 receptor (D2R) agonists; dopamine (DA), p-tyramine, (R)- and (S)-5-OH-dipropylaminotetralin (DPAT). We further probed the role of the conserved serine S1935.42 by mutagenesis, taking advantage of the preferential interaction of (S)-, but not (R)-5-OH-DPAT with this residue. Results suggested similar koffs for the two 5-OH-DPAT enantiomers at wild-type (WT) D2R, both being slower than the koffs of DA and p-tyramine. Conversely, the kon of (S)-5-OH-DPAT was estimated to be higher than that of (R)-5-OH-DPAT, in agreement with the higher potency of the (S)-enantiomer. Furthermore, S1935.42A mutation lowered the kon of (S)-5-OH-DPAT and reduced the potency difference between the two 5-OH-DPAT enantiomers. Kinetic Kds derived from the koff and kon estimates correlated well with EC50 values for all four compounds across four orders of magnitude, strengthening the notion that our assay captured meaningful information about binding kinetics. The approach presented here may thus prove valuable for characterizing D2R agonist candidate drugs.
Collapse
|
5
|
Structure of the dopamine D 2 receptor in complex with the antipsychotic drug spiperone. Nat Commun 2020; 11:6442. [PMID: 33353947 PMCID: PMC7755896 DOI: 10.1038/s41467-020-20221-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 11/19/2020] [Indexed: 12/26/2022] Open
Abstract
In addition to the serotonin 5-HT2A receptor (5-HT2AR), the dopamine D2 receptor (D2R) is a key therapeutic target of antipsychotics for the treatment of schizophrenia. The inactive state structures of D2R have been described in complex with the inverse agonists risperidone (D2Rris) and haloperidol (D2Rhal). Here we describe the structure of human D2R in complex with spiperone (D2Rspi). In D2Rspi, the conformation of the extracellular loop (ECL) 2, which composes the ligand-binding pocket, was substantially different from those in D2Rris and D2Rhal, demonstrating that ECL2 in D2R is highly dynamic. Moreover, D2Rspi exhibited an extended binding pocket to accommodate spiperone’s phenyl ring, which probably contributes to the selectivity of spiperone to D2R and 5-HT2AR. Together with D2Rris and D2Rhal, the structural information of D2Rspi should be of value for designing novel antipsychotics with improved safety and efficacy. The dopamine D2 receptor (D2R) is a GPCR and an important drug target for schizophrenia treatment. Here, the authors present the crystal structure of human D2R in complex with the antipsychotic drug spiperone, which is of interest for designing antipsychotics with improved receptor selectivity.
Collapse
|
6
|
Rojas S, Parravicini O, Vettorazzi M, Tosso R, Garro A, Gutiérrez L, Andújar S, Enriz R. Combined MD/QTAIM techniques to evaluate ligand-receptor interactions. Scope and limitations. Eur J Med Chem 2020; 208:112792. [PMID: 32949964 DOI: 10.1016/j.ejmech.2020.112792] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/29/2022]
Abstract
In medicinal chemistry, it is extremely important to evaluate, as accurately as possible, the molecular interactions involved in the formation of different ligand-receptor (L-R) complexes. Evaluating the different molecular interactions by quantum mechanics calculations is not a simple task, since formation of an L-R complex is a dynamic process. In this case, the use of combined techniques of molecular dynamics (MD) and quantum calculations is one the best possible approaches. In this work we report a comparative study using combined MD and QTAIM (Quantum Theory of Atoms In Molecules) calculations for five biological systems with different levels of structural complexity. We have studied Acetylcholinesterase (AChE), D2 Dopamine Receptor (D2DR), beta Secretase (BACE1), Dihydrofolate Reductase (DHFR) and Sphingosine Kinase 1 (SphK1). In these molecular targets, we have analyzed different ligands with diverse structural characteristics. The inhibitory activities of most of them have been previously measured in our laboratory. Our results indicate that QTAIM calculations can be extremely useful for in silico studies. It is possible to obtain very accurate information about the strength of the molecular interactions that stabilize the formation of the different L-R complexes. Better correlations can be obtained between theoretical and experimental data by using QTAIM calculations, allowing us to discriminate among ligands with similar affinities. QTAIM analysis gives fairly accurate information for weak interactions which are not well described by MD simulations. QTAIM study also allowed us to evaluate and determine which parts of the ligand need to be modified in order to increase its interactions with the molecular target. In this study we have discussed the importance of combined MD/QTAIM calculations for this type of simulations, showing their scopes and limitations.
Collapse
Affiliation(s)
- Sebastián Rojas
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Oscar Parravicini
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Marcela Vettorazzi
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Rodrigo Tosso
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Adriana Garro
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Lucas Gutiérrez
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Sebastián Andújar
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Ricardo Enriz
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina.
| |
Collapse
|
7
|
Zhou Y, Cao C, He L, Wang X, Zhang XC. Crystal structure of dopamine receptor D4 bound to the subtype selective ligand, L745870. eLife 2019; 8:e48822. [PMID: 31750832 PMCID: PMC6872212 DOI: 10.7554/elife.48822] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple subtypes of dopamine receptors within the GPCR superfamily regulate neurological processes through various downstream signaling pathways. A crucial question about the dopamine receptor family is what structural features determine the subtype-selectivity of potential drugs. Here, we report the 3.5-angstrom crystal structure of mouse dopamine receptor D4 (DRD4) complexed with a subtype-selective antagonist, L745870. Our structure reveals a secondary binding pocket extended from the orthosteric ligand-binding pocket to a DRD4-specific crevice located between transmembrane helices 2 and 3. Additional mutagenesis studies suggest that the antagonist L745870 prevents DRD4 activation by blocking the relative movement between transmembrane helices 2 and 3. These results expand our knowledge of the molecular basis for the physiological functions of DRD4 and assist new drug design.
Collapse
Affiliation(s)
- Ye Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Can Cao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Lingli He
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Xianping Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Xuejun Cai Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
8
|
Vass M, Podlewska S, de Esch IJP, Bojarski AJ, Leurs R, Kooistra AJ, de Graaf C. Aminergic GPCR-Ligand Interactions: A Chemical and Structural Map of Receptor Mutation Data. J Med Chem 2018; 62:3784-3839. [PMID: 30351004 DOI: 10.1021/acs.jmedchem.8b00836] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The aminergic family of G protein-coupled receptors (GPCRs) plays an important role in various diseases and represents a major drug discovery target class. Structure determination of all major aminergic subfamilies has enabled structure-based ligand design for these receptors. Site-directed mutagenesis data provides an invaluable complementary source of information for elucidating the structural determinants of binding of different ligand chemotypes. The current study provides a comparative analysis of 6692 mutation data points on 34 aminergic GPCR subtypes, covering the chemical space of 540 unique ligands from mutagenesis experiments and information from experimentally determined structures of 52 distinct aminergic receptor-ligand complexes. The integrated analysis enables detailed investigation of structural receptor-ligand interactions and assessment of the transferability of combined binding mode and mutation data across ligand chemotypes and receptor subtypes. An overview is provided of the possibilities and limitations of using mutation data to guide the design of novel aminergic receptor ligands.
Collapse
Affiliation(s)
- Márton Vass
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS) , VU University Amsterdam , 1081HZ Amsterdam , The Netherlands
| | - Sabina Podlewska
- Department of Medicinal Chemistry, Institute of Pharmacology , Polish Academy of Sciences , Smętna 12 , PL31-343 Kraków , Poland
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS) , VU University Amsterdam , 1081HZ Amsterdam , The Netherlands
| | - Andrzej J Bojarski
- Department of Medicinal Chemistry, Institute of Pharmacology , Polish Academy of Sciences , Smętna 12 , PL31-343 Kraków , Poland
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS) , VU University Amsterdam , 1081HZ Amsterdam , The Netherlands
| | - Albert J Kooistra
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS) , VU University Amsterdam , 1081HZ Amsterdam , The Netherlands.,Department of Drug Design and Pharmacology , University of Copenhagen , Universitetsparken 2 , 2100 Copenhagen , Denmark
| | - Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS) , VU University Amsterdam , 1081HZ Amsterdam , The Netherlands.,Sosei Heptares , Steinmetz Building, Granta Park, Great Abington , Cambridge CB21 6DG , U.K
| |
Collapse
|
9
|
Draper-Joyce CJ, Michino M, Verma RK, Klein Herenbrink C, Shonberg J, Kopinathan A, Scammells PJ, Capuano B, Thal DM, Javitch JA, Christopoulos A, Shi L, Lane JR. The structural determinants of the bitopic binding mode of a negative allosteric modulator of the dopamine D 2 receptor. Biochem Pharmacol 2018; 148:315-328. [PMID: 29325769 DOI: 10.1016/j.bcp.2018.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/02/2018] [Indexed: 01/11/2023]
Abstract
SB269652 is a negative allosteric modulator of the dopamine D2 receptor (D2R) yet possesses structural similarity to ligands with a competitive mode of interaction. In this study, we aimed to understand the ligand-receptor interactions that confer its allosteric action. We combined site-directed mutagenesis with molecular dynamics simulations using both SB269652 and derivatives from our previous structure activity studies. We identify residues within the conserved orthosteric binding site (OBS) and a secondary binding pocket (SBP) that determine affinity and cooperativity. Our results indicate that interaction with the SBP is a requirement for allosteric pharmacology, but that both competitive and allosteric derivatives of SB269652 can display sensitivity to the mutation of a glutamate residue (E952.65) within the SBP. Our findings provide the molecular basis for the differences in affinity between SB269652 derivatives, and reveal how changes to interactions made by the primary pharmacophore of SB269652 in the orthosteric pocket can confer changes in the interactions made by the secondary pharmacophore in the SBP. Our insights provide a structure-activity framework towards rational optimization of bitopic ligands for D2R with tailored competitive versus allosteric properties.
Collapse
Affiliation(s)
- Christopher J Draper-Joyce
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Mayako Michino
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Ravi Kumar Verma
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Carmen Klein Herenbrink
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Jeremy Shonberg
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Anitha Kopinathan
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Ben Capuano
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - David M Thal
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Jonathan A Javitch
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York State Psychiatric Institute, New York, NY 10032, United States; Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York State Psychiatric Institute, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States.
| | - J Robert Lane
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), 399 Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
10
|
Investigating the structural impact of S311C mutation in DRD2 receptor by molecular dynamics & docking studies. Biochimie 2016; 123:52-64. [DOI: 10.1016/j.biochi.2016.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/16/2016] [Indexed: 01/11/2023]
|
11
|
Salmas RE, Yurtsever M, Stein M, Durdagi S. Modeling and protein engineering studies of active and inactive states of human dopamine D2 receptor (D2R) and investigation of drug/receptor interactions. Mol Divers 2015; 19:321-32. [DOI: 10.1007/s11030-015-9569-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 01/11/2015] [Indexed: 01/11/2023]
|
12
|
Angelina E, Andujar S, Moreno L, Garibotto F, Párraga J, Peruchena N, Cabedo N, Villecco M, Cortes D, Enriz RD. 3-Chlorotyramine Acting as Ligand of the D2
Dopamine Receptor. Molecular Modeling, Synthesis and D2
Receptor Affinity. Mol Inform 2014; 34:28-43. [PMID: 27490860 DOI: 10.1002/minf.201400093] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/04/2014] [Indexed: 12/12/2022]
|
13
|
Bhattacharya S, Lam AR, Li H, Balaraman G, Niesen MJM, Vaidehi N. Critical analysis of the successes and failures of homology models of G protein-coupled receptors. Proteins 2013; 81:729-39. [PMID: 23042299 PMCID: PMC3785289 DOI: 10.1002/prot.24195] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/17/2012] [Accepted: 09/21/2012] [Indexed: 12/22/2022]
Abstract
We present a critical assessment of the performance of our homology model refinement method for G protein-coupled receptors (GPCRs), called LITICon that led to top ranking structures in a recent structure prediction assessment GPCRDOCK2010. GPCRs form the largest class of drug targets for which only a few crystal structures are currently available. Therefore, accurate homology models are essential for drug design in these receptors. We submitted five models each for human chemokine CXCR4 (bound to small molecule IT1t and peptide CVX15) and dopamine D3DR (bound to small molecule eticlopride) before the crystal structures were published. Our models in both CXCR4/IT1t and D3/eticlopride assessments were ranked first and second, respectively, by ligand RMSD to the crystal structures. For both receptors, we developed two types of protein models: homology models based on known GPCR crystal structures, and ab initio models based on the prediction method MembStruk. The homology-based models compared better to the crystal structures than the ab initio models. However, a robust refinement procedure for obtaining high accuracy structures is needed. We demonstrate that optimization of the helical tilt, rotation, and translation is vital for GPCR homology model refinement. As a proof of concept, our in-house refinement program LITiCon captured the distinct orientation of TM2 in CXCR4, which differs from that of adrenoreceptors. These findings would be critical for refining GPCR homology models in future.
Collapse
MESH Headings
- Binding Sites
- Dopamine Antagonists/chemistry
- Dopamine Antagonists/pharmacology
- Humans
- Ligands
- Molecular Docking Simulation
- Protein Conformation
- Receptors, Adrenergic, beta-2/chemistry
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, CXCR4/chemistry
- Receptors, CXCR4/metabolism
- Receptors, Dopamine D3/chemistry
- Receptors, Dopamine D3/metabolism
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/metabolism
- Salicylamides/chemistry
- Salicylamides/pharmacology
- Structural Homology, Protein
Collapse
Affiliation(s)
- Supriyo Bhattacharya
- Division of Immunology, Beckman Research Institute of the City of Hope, Duarte, California 91010, USA
| | | | | | | | | | | |
Collapse
|
14
|
Malo M, Persson R, Svensson P, Luthman K, Brive L. Development of 7TM receptor-ligand complex models using ligand-biased, semi-empirical helix-bundle repacking in torsion space: application to the agonist interaction of the human dopamine D2 receptor. J Comput Aided Mol Des 2013; 27:277-91. [PMID: 23553533 PMCID: PMC3639355 DOI: 10.1007/s10822-013-9640-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 03/20/2013] [Indexed: 11/30/2022]
Abstract
Prediction of 3D structures of membrane proteins, and of G-protein coupled receptors (GPCRs) in particular, is motivated by their importance in biological systems and the difficulties associated with experimental structure determination. In the present study, a novel method for the prediction of 3D structures of the membrane-embedded region of helical membrane proteins is presented. A large pool of candidate models are produced by repacking of the helices of a homology model using Monte Carlo sampling in torsion space, followed by ranking based on their geometric and ligand-binding properties. The trajectory is directed by weak initial restraints to orient helices towards the original model to improve computation efficiency, and by a ligand to guide the receptor towards a chosen conformational state. The method was validated by construction of the β1 adrenergic receptor model in complex with (S)-cyanopindolol using bovine rhodopsin as template. In addition, models of the dopamine D2 receptor were produced with the selective and rigid agonist (R)-N-propylapomorphine ((R)-NPA) present. A second quality assessment was implemented by evaluating the results from docking of a library of 29 ligands with known activity, which further discriminated between receptor models. Agonist binding and recognition by the dopamine D2 receptor is interpreted using the 3D structure model resulting from the approach. This method has a potential for modeling of all types of helical transmembrane proteins for which a structural template with sequence homology sufficient for homology modeling is not available or is in an incorrect conformational state, but for which sufficient empirical information is accessible.
Collapse
Affiliation(s)
- Marcus Malo
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-412 96 Göteborg, Sweden
| | - Ronnie Persson
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-412 96 Göteborg, Sweden
| | - Peder Svensson
- NeuroSearch Sweden AB, Arvid Wallgrens Backe 20, SE-413 46 Göteborg, Sweden
- Present Address: Astra Zeneca R&D Mölndal, SE-431 83 Mölndal, Sweden
| | - Kristina Luthman
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-412 96 Göteborg, Sweden
| | - Lars Brive
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-412 96 Göteborg, Sweden
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Box 440, SE-405 30 Göteborg, Sweden
- Cygnal Bioscience, Björnvägen 15, SE-435 43 Pixbo, Sweden
| |
Collapse
|
15
|
Jain ZJ, Kankate RS, Chaudhari BN, Kakad RD. Action of benzimidazolo-piperazinyl derivatives on dopamine receptors. Med Chem Res 2013. [DOI: 10.1007/s00044-012-0055-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
16
|
Yang B, Ni J, Zeng Z, Shi B, You W, Ke C. Cloning and characterization of the dopamine like receptor in the oyster Crassostrea angulata: expression during the ovarian cycle. Comp Biochem Physiol B Biochem Mol Biol 2012; 164:168-75. [PMID: 23274282 DOI: 10.1016/j.cbpb.2012.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/20/2012] [Accepted: 12/20/2012] [Indexed: 01/11/2023]
Abstract
We cloned and characterized a complete cDNA encoding a dopamine receptor (DAR) named Ca-DA1R from Fujian oyster, Crassostrea angulata. The 2843 bp long cDNA sequence includes a 916-bp 5'-UTR, the 1197 bp ORF which encodes a putative protein of 399 amino acids, and a 729 bp 3'-UTR. The Ca-DA1R sequence possesses typical characteristics of a D1 receptor: two main features being a short third intracellular loop and a long inner COOH-terminal tail domain. Using a real-time PCR approach, expression profiles of Ca-DA1R were analyzed in adult tissues and during the four stages of ovarian development. Ca-DA1R was expressed ubiquitously, although transcript levels varied between tissues, with higher mRNA levels detected in the ovary, labial palps and mantle. During the four stages of ovarian development, Ca-DA1R mRNA expression level was higher in the proliferation stage than in the other three stages during the ovary cycle. In situ hybridization results reveal that the Ca-DA1R mRNA is mainly expressed in the epithelium of the gonoducts. These observations suggest that Ca-DA1R binding of DA probably plays an important role in early ovarian development and via regulating oocyte locomotion cooperates with the 5-HT receptor system during the ovarian cycle in C. angulata.
Collapse
Affiliation(s)
- Bingye Yang
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen 361005, PR China
| | | | | | | | | | | |
Collapse
|
17
|
Gogoi S, Biswas S, Modi G, Antonio T, Reith MEA, Dutta AK. Novel bivalent ligands for D2/D3 dopamine receptors: Significant co-operative gain in D2 affinity and potency. ACS Med Chem Lett 2012; 3:991-996. [PMID: 23275802 DOI: 10.1021/ml3002117] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
This report describes development of a series of novel bivalent molecules with a pharmacophore derived from the D2/D3 agonist 5-OH-DPAT. Spacer length in the bivalent compounds had a pronounced influence on affinity for D2 receptors. A 23-fold increase of D2 affinity was observed at a spacer length of 9 or 10 (compounds 11d and 14b) compared to monovalent 5-OH-DPAT (Ki; 2.5 and 2.0 vs. 59 nM for 11d and 14b vs. 5-OH-DPAT, respectively). Functional potency of 11d and 14b indicated a 24- and 94-fold increase in potency at the D2 receptor compared to 5-OH-DPAT (EC50; 1.7 and 0.44 vs. 41 nM for 11d and 14b vs. 5-OH-DPAT, respectively). These are the most potent bivalent agonists for D2 receptor known to date. This synergism is consonant with cooperative interaction at the two orthosteric binding sites in the homodimeric receptor.
Collapse
Affiliation(s)
- Sanjib Gogoi
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | - Swati Biswas
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | - Gyan Modi
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | | | | | - Aloke K. Dutta
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
18
|
Li B, Li W, Du P, Yu KQ, Fu W. Molecular Insights into the D1R Agonist and D2R/D3R Antagonist Effects of the Natural Product (−)-Stepholidine: Molecular Modeling and Dynamics Simulations. J Phys Chem B 2012; 116:8121-30. [DOI: 10.1021/jp3049235] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Bian Li
- Department of Medicinal Chemistry & Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Li
- Department of Medicinal Chemistry & Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peng Du
- Department of Medicinal Chemistry & Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Kun Qian Yu
- Drug Discovery and Design Center,
State Key Laboratory of Drug Research, Shanghai Institute of Materia
Medica, Chinese Academy of Sciences, Shanghai
201203, China
| | - Wei Fu
- Department of Medicinal Chemistry & Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
19
|
Fowler JC, Bhattacharya S, Urban JD, Vaidehi N, Mailman RB. Receptor conformations involved in dopamine D(2L) receptor functional selectivity induced by selected transmembrane-5 serine mutations. Mol Pharmacol 2012; 81:820-31. [PMID: 22416052 PMCID: PMC3362898 DOI: 10.1124/mol.111.075457] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 03/13/2012] [Indexed: 11/22/2022] Open
Abstract
Although functional selectivity is now widely accepted, the molecular basis is poorly understood. We have studied how aspects of transmembrane region 5 (TM5) of the dopamine D(2L) receptor interacts with three rationally selected rigid ligands (dihydrexidine, dinapsoline, and dinoxyline) and the reference compounds dopamine and quinpirole. As was expected from homology modeling, mutation of three TM5 serine residues to alanine (S5.42A, S5.43A, S5.46A) had little effect on antagonist affinity. All three mutations decreased the affinity of the agonist ligands to different degrees, S5.46A being somewhat less affected. Four functions [adenylate cyclase (AC), extracellular signal-regulated kinase 1/2 phosphorylation (MAPK), arachidonic acid release (AA), and guanosine 5'-O-(3-thio)triphosphate binding (GTPγS)] were assessed. The intrinsic activity (IA) of quinpirole was unaffected by any of the mutations, whereas S5.42A and S5.46A mutations abolished the activity of dopamine and the three rigid ligands, although dihydrexidine retained IA at MAPK function only with S5.42A. Remarkably, S5.43A did not markedly affect IA for AC and MAPK for any of the ligands and eliminated AA activity for dinapsoline and dihydrexidine but not dinoxyline. These data suggest that this mutation did not disrupt the overall conformation or signaling ability of the mutant receptors but differentially affected ligand activation. Computational studies indicate that these D(2) agonists stabilize multiple receptor conformations. This has led to models showing the stabilized conformations and interhelical and receptor-ligand contacts corresponding to the different activation pathways stabilized by various agonists. These data provide a basis for understanding D(2L) functional selectivity and rationally discovering functionally selective D(2) drugs.
Collapse
Affiliation(s)
- J Corey Fowler
- Division of Medicinal Chemistry and Toxicology Curriculum, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | | | | | | |
Collapse
|
20
|
Malo M, Brive L, Luthman K, Svensson P. Investigation of D₂ receptor-agonist interactions using a combination of pharmacophore and receptor homology modeling. ChemMedChem 2012; 7:471-82, 338. [PMID: 22315215 PMCID: PMC3382189 DOI: 10.1002/cmdc.201100545] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 01/05/2012] [Indexed: 01/21/2023]
Abstract
A combined modeling approach was used to identify structural factors that underlie the structure–activity relationships (SARs) of full dopamine D2 receptor agonists and structurally similar inactive compounds. A 3D structural model of the dopamine D2 receptor was constructed, with the agonist (−)-(R)-2-OH-NPA present in the binding site during the modeling procedure. The 3D model was evaluated and compared with our previously published D2 agonist pharmacophore model. The comparison revealed an inconsistency between the projected hydrogen bonding feature (Ser-TM5) in the pharmacophore model and the TM5 region in the structure model. A new refined pharmacophore model was developed, guided by the shape of the binding site in the receptor model and with less emphasis on TM5 interactions. The combination of receptor and pharmacophore modeling also identified the importance of His3936.55 for agonist binding. This convergent 3D pharmacophore and protein structure modeling strategy is considered to be general and can be highly useful in less well-characterized systems to explore ligand–receptor interactions. The strategy has the potential to identify weaknesses in the individual models and thereby provides an opportunity to improve the discriminating predictivity of both pharmacophore searches and structure-based virtual screens.
Collapse
Affiliation(s)
- Marcus Malo
- Department of Chemistry, Medicinal Chemistry, University of Gothenburg, 41296 Göteborg, Sweden
| | | | | | | |
Collapse
|
21
|
Sahlholm K, Barchad-Avitzur O, Marcellino D, Gómez-Soler M, Fuxe K, Ciruela F, Arhem P. Agonist-specific voltage sensitivity at the dopamine D2S receptor--molecular determinants and relevance to therapeutic ligands. Neuropharmacology 2011; 61:937-49. [PMID: 21752340 DOI: 10.1016/j.neuropharm.2011.06.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 05/20/2011] [Accepted: 06/24/2011] [Indexed: 10/18/2022]
Abstract
Voltage sensitivity has been demonstrated for some GPCRs. At the dopamine D(2S) receptor, this voltage sensitivity is agonist-specific; some agonists, including dopamine, exhibit decreased potency at depolarized potentials, whereas others are not significantly affected. In the present study, we examined some of the receptor-agonist interactions contributing to these differences, and investigated how dopamine D(2S) receptor voltage sensitivity affects clinically used dopamine agonists. GIRK channel activation in voltage-clamped Xenopus oocytes was used as readout of receptor activation. Structurally distinct agonists and complementary site-directed mutagenesis of the receptor's binding site were used to investigate the role of agonist-receptor interactions. We also confirmed that the depolarization-induced decrease of dopamine potency in GIRK activation is correlated by decreased binding of radiolabeled dopamine, and by decreased potency in G protein activation. In the mutagenesis experiments, a conserved serine residue as well as the conserved aspartate in the receptor's binding site were found to be important for voltage sensitive potency of dopamine. Furthermore, the voltage sensitivity of the receptor had distinct effects on different therapeutic D(2) agonists. Depolarization decreased the potency of several compounds, whereas for others, efficacy was reduced. For some agonists, both potency and efficacy were diminished, whereas for others still, neither parameter was significantly altered. The present work identifies some of the ligand-receptor interactions which determine agonist-specific effects of voltage at the dopamine D(2S) receptor. The observed differences between therapeutic agonists might be clinically relevant, and make them potential tools for investigating the roles of dopamine D(2) receptor voltage sensitivity in native tissue.
Collapse
|
22
|
Tetrahydroisoquinolines acting as dopaminergic ligands. A molecular modeling study using MD simulations and QM calculations. J Mol Model 2011; 18:419-31. [DOI: 10.1007/s00894-011-1061-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 03/22/2011] [Indexed: 11/24/2022]
|
23
|
Ortore G, Tuccinardi T, Orlandini E, Martinelli A. Different Binding Modes of Structurally Diverse Ligands for Human D3DAR. J Chem Inf Model 2010; 50:2162-75. [DOI: 10.1021/ci100290f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Gabriella Ortore
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Tiziano Tuccinardi
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Elisabetta Orlandini
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Adriano Martinelli
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
24
|
Kortagere S, Cheng SY, Antonio T, Zhen J, Reith MEA, Dutta AK. Interaction of novel hybrid compounds with the D3 dopamine receptor: Site-directed mutagenesis and homology modeling studies. Biochem Pharmacol 2010; 81:157-63. [PMID: 20833147 DOI: 10.1016/j.bcp.2010.08.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 08/05/2010] [Accepted: 08/30/2010] [Indexed: 12/14/2022]
Abstract
The dopamine D3 receptor has been implicated as a potential target for drug development in various complex psychiatric disorders including psychosis, drug dependence, and Parkinson's disease. In our overall goal to develop molecules with preferential affinity at D3 receptors, we undertook a hybrid drug development approach by combining a known dopamine agonist moiety with a substituted piperazine fragment. In the present study, three compounds produced this way with preferential D3 agonist activity, were tested at D3 receptors with mutations in the agonist binding pocket of three residues known to be important for agonist binding activity. At S192A and T369V, the hybrid agonist compounds produced an interaction profile in [(3)H]spiperone binding assays similar to that of the parent 5-OH-DPAT and 7-OH-DPAT molecules. The loss of affinity at the S192A mutant was most prominent for 5-OH-DPAT and its corresponding hybrid compound D237. D110N did not show any radioligand binding. Homology modeling indicated that 7-OH-DPAT-derived D315 uniquely shares H-bonding with Tyr365 which produced favorable interaction and no loss of H-bonding in the S192A mutant, suggesting that agonist activity may not be solely controlled by residues in the binding pocket.
Collapse
Affiliation(s)
- Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
The antipsychotic effectiveness of chlorpromazine and haloperidol started a search for their therapeutic targets. The antipsychotic receptor target turned out to be a dopamine receptor, now cloned as the dopamine D2 receptor. The D2 receptor is the common target for antipsychotics. Antipsychotic clinical doses correlate with their affinities for this receptor. Therapeutic doses of antipsychotics occupy 60 to 80% of brain D2 receptors in patients, but aripiprazole occupies up to 90%. While antipsychotics may take up to six hours to occupy D2 receptors, much clinical improvement occurs within a few days. The receptor has high- and low-affinity states. The D2High state is functional for dopamine-like agonists such as aripiprazole. Most individuals with schizophrenia are supersensitive to dopamine. Animal models of psychosis show that a variety of risk factors, genetic and nongenetic, are associated with behavioral supersensitivity to dopamine, reflected in elevated levels of dopamine D2High receptors. Although antipsychotics such as haloperidol alleviate psychosis and reverse the elevation of D2High receptors, long-term use of traditional antipsychotics can further enhance dopamine supersensitivity in patients. Therefore, switching from a traditional antipsychotic to an agonist antipsychotic such as aripiprazole can result in the emergence of psychotic signs and symptoms. Clozapine and quetiapine do not elicit parkinsonism and rarely result in tardive dyskinesia because they are released from D2 within 12 to 24 hours. Traditional antipsychotics remain attached to D2 receptors for days, preventing relapse, but allowing accumulation that can lead to tardive dyskinesia. Future goals include imaging D2High receptors and desensitizing them in early-stage psychosis.
Collapse
Affiliation(s)
- Philip Seeman
- Pharmacology Department, Faculty of Medicine, University of Toronto, Canada.
| |
Collapse
|
26
|
Rondou P, Haegeman G, Van Craenenbroeck K. The dopamine D4 receptor: biochemical and signalling properties. Cell Mol Life Sci 2010; 67:1971-86. [PMID: 20165900 PMCID: PMC11115718 DOI: 10.1007/s00018-010-0293-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 01/19/2010] [Accepted: 01/26/2010] [Indexed: 01/20/2023]
Abstract
Dopamine is an important neurotransmitter that regulates several key functions in the brain, such as motor output, motivation and reward, learning and memory, and endocrine regulation. Dopamine does not mediate fast synaptic transmission, but rather modulates it by triggering slow-acting effects through the activation of dopamine receptors, which belong to the G-protein-coupled receptor superfamily. Besides activating different effectors through G-protein coupling, dopamine receptors also signal through interaction with a variety of proteins, collectively termed dopamine receptor-interacting proteins. We focus on the dopamine D4 receptor, which contains an important polymorphism in its third intracellular loop. This polymorphism has been the subject of numerous studies investigating links with several brain disorders, such as attention-deficit hyperactivity disorder and schizophrenia. We provide an overview of the structure, signalling properties and regulation of dopamine D4 receptors, and briefly discuss their physiological and pathophysiological role in the brain.
Collapse
Affiliation(s)
- Pieter Rondou
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University (UGent), K.L. Ledeganckstraat 35, 9000 Ghent, Belgium
- Present Address: Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Medical Research Building, De Pintelaan 185, 9000 Ghent, Belgium
| | - Guy Haegeman
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University (UGent), K.L. Ledeganckstraat 35, 9000 Ghent, Belgium
| | - Kathleen Van Craenenbroeck
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University (UGent), K.L. Ledeganckstraat 35, 9000 Ghent, Belgium
| |
Collapse
|
27
|
Cummings DF, Ericksen SS, Goetz A, Schetz JA. Transmembrane segment five serines of the D4 dopamine receptor uniquely influence the interactions of dopamine, norepinephrine, and Ro10-4548. J Pharmacol Exp Ther 2010; 333:682-95. [PMID: 20215412 PMCID: PMC2879936 DOI: 10.1124/jpet.109.164962] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 03/05/2010] [Indexed: 11/22/2022] Open
Abstract
Conserved serines of transmembrane segment (TM) five (TM5) are critical for the interactions of endogenous catecholamines with alpha(1)- and alpha(2)-adrenergic, beta(2)-adrenergic, and D1, D2, and D3 dopamine receptors. The unique high-affinity interaction of the D4 dopamine receptor subtype with both norepinephrine and dopamine, and the fact that TM5 serine interactions have never been studied for this receptor subtype, led us to investigate the interactions of ligands with D4 receptor TM5 serines. Serine-to-alanine mutations at positions 5.42 and 5.46 drastically decreased affinities of dopamine and norepinephrine for the D4 receptor. The D4-S5.43A receptor mutant had substantially reduced affinity for norepinephrine, but a modest loss of affinity for dopamine. In functional assays of cAMP accumulation, norephinephrine was unable to activate any of the mutant receptors, even though the agonist quinpirole displayed wild-type functional properties for all of them. Dopamine was unable to activate the S5.46A mutant and had reduced potency for the S5.43A mutant and reduced potency and efficacy for the S5.42A mutant. In contrast, Ro10-4548 [RAC-2'-2-hydroxy-3-4-(4-hydroxy-2-methoxyphenyl)-1-piperazinyl-propoxy-acetanilide], a catechol-like antagonist of the wild-type receptor unexpectedly functions as an agonist of the S5.43A mutant. Other noncatechol ligands had similar properties for mutant and wild-type receptors. This is the first example of a dopamine receptor point mutation selectively changing the receptor's interaction with a specific antagonist to that of an agonist, and together with other data, provides evidence, supported by molecular modeling, that catecholamine-type agonism is induced by different ligand-specific configurations of intermolecular H-bonds with the TM5 conserved serines.
Collapse
Affiliation(s)
- David F Cummings
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107-2699, USA
| | | | | | | |
Collapse
|
28
|
Pettersson F, Pontén H, Waters N, Waters S, Sonesson C. Synthesis and evaluation of a set of 4-phenylpiperidines and 4-phenylpiperazines as D2 receptor ligands and the discovery of the dopaminergic stabilizer 4-[3-(methylsulfonyl)phenyl]-1-propylpiperidine (huntexil, pridopidine, ACR16). J Med Chem 2010; 53:2510-20. [PMID: 20155917 DOI: 10.1021/jm901689v] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Modification of the partial dopamine type 2 receptor (D(2)) agonist 3-(1-benzylpiperidin-4-yl)phenol (9a) generated a series of novel functional D(2) antagonists with fast-off kinetic properties. A representative of this series, pridopidine (4-[3-(methylsulfonyl)phenyl]-1-propylpiperidine; ACR16, 12b), bound competitively with low affinity to D(2) in vitro, without displaying properties essential for interaction with D(2) in the inactive state, thereby allowing receptors to rapidly regain responsiveness. In vivo, neurochemical effects of 12b were similar to those of D(2) antagonists, and in a model of locomotor hyperactivity, 12b dose-dependently reduced activity. In contrast to classic D(2) antagonists, 12b increased spontaneous locomotor activity in partly habituated animals. The "agonist-like" kinetic profile of 12b, combined with its lack of intrinsic activity, induces a functional state-dependent D(2) antagonism that can vary with local, real-time dopamine concentration fluctuations around distinct receptor populations. These properties may contribute to its unique "dopaminergic stabilizer" characteristics, differentiating 12b from D(2) antagonists and partial D(2) agonists.
Collapse
Affiliation(s)
- Fredrik Pettersson
- NeuroSearch Sweden AB, Arvid Wallgrens Backe 20, S-413 46 Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
29
|
Malo M, Brive L, Luthman K, Svensson P. Selective pharmacophore models of dopamine D(1) and D(2) full agonists based on extended pharmacophore features. ChemMedChem 2010; 5:232-46. [PMID: 20077461 DOI: 10.1002/cmdc.200900398] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This study is focused on the identification of structural features that determine the selectivity of dopamine receptor agonists toward D(1) and D(2) receptors. Selective pharmacophore models were developed for both receptors. The models were built by using projected pharmacophoric features that represent the main agonist interaction sites in the receptor (the Ser residues in TM5 and the Asp in TM3), a directional aromatic feature in the ligand, a feature with large positional tolerance representing the positively charged nitrogen in the ligand, and sets of excluded volumes reflecting the shapes of the receptors. The sets of D(1) and D(2) ligands used for modeling were carefully selected from published sources and consist of structurally diverse, conformationally rigid full agonists as active ligands together with structurally related inactives. The robustness of the models in discriminating actives from inactives was tested against four ensembles of conformations generated by using different established methods and different force fields. The reasons for the selectivity can be attributed to both geometrical differences in the arrangement of the features, e.g., different tilt angels of the pi system, as well as shape differences covered by the different sets of excluded volumes. This work provides useful information for the design of new D(1) and D(2) agonists and also for comparative homology modeling of D(1) and D(2) receptors. The approach is general and could therefore be applied to other ligand-protein interactions for which no experimental protein structure is available.
Collapse
Affiliation(s)
- Marcus Malo
- Department of Chemistry, Medicinal Chemistry, University of Gothenburg, 41296 Göteborg, Sweden
| | | | | | | |
Collapse
|
30
|
McRobb FM, Capuano B, Crosby IT, Chalmers DK, Yuriev E. Homology Modeling and Docking Evaluation of Aminergic G Protein-Coupled Receptors. J Chem Inf Model 2010; 50:626-37. [DOI: 10.1021/ci900444q] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Fiona M. McRobb
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052 Australia
| | - Ben Capuano
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052 Australia
| | - Ian T. Crosby
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052 Australia
| | - David K. Chalmers
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052 Australia
| | - Elizabeth Yuriev
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052 Australia
| |
Collapse
|
31
|
Sipos A, Mueller FKU, Lehmann J, Berényi S, Antus S. Synthesis and Pharmacological Evaluation of Thiazole and Isothiazole Derived Apomorphines. Arch Pharm (Weinheim) 2009; 342:557-68. [DOI: 10.1002/ardp.200900100] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
32
|
Taman A, Ribeiro P. Investigation of a dopamine receptor in Schistosoma mansoni: functional studies and immunolocalization. Mol Biochem Parasitol 2009; 168:24-33. [PMID: 19545592 DOI: 10.1016/j.molbiopara.2009.06.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/05/2009] [Accepted: 06/11/2009] [Indexed: 11/15/2022]
Abstract
A dopamine receptor (SmD2) was cloned from adult Schistosoma mansoni. The receptor has the classical heptahelical topology of class A (rhodopsin-like) G protein-coupled receptors (GPCR) and shares sequence homology with D2-like receptors from other species. The full length SmD2 cDNA was expressed in the yeast Saccharomyces cerevisiae and mammalian HEK293 cells. Functional assays in both expression systems revealed that SmD2 was responsive to dopamine in a dose-dependent manner, whereas other structurally related amines had no effect. Activation of SmD2 in mammalian cells caused an elevation in intracellular cAMP but not calcium, suggesting that the receptor coupled to Gs and the stimulation of adenylate cyclase. Pharmacological studies showed that the S. mansoni dopamine receptor was inhibited by apomorphine, a classical dopamine agonist, as well as known dopaminergic antagonists, including chlorpromazine, spiperone and haloperidol. SmD2 immunoreactivity was detected in membrane protein fractions of S. mansoni cercaria, in vitro transformed schistosomula and adult parasites, using a specific peptide antibody. When tested by confocal immunofluorescence, SmD2 was detected in the subtegumental somatic musculature and acetabulum of all larval stages tested. In the adults, SmD2 was enriched in the somatic muscles and, to a lesser extent, the muscular lining of the caecum. The results suggest that SmD2 is an important component of the neuromuscular system in schistosomes.
Collapse
Affiliation(s)
- Amira Taman
- Institute of Parasitology, McGill University, Macdonald Campus, 21,111 Lakeshore Road, Sainte Anne de Bellevue, Quebec, Canada
| | | |
Collapse
|
33
|
Shiba K, Torashima T, Hirai H, Ogawa K, Akhter N, Nakajima K, Kinuya S, Mori H. Potential usefulness of D2R reporter gene imaging by IBF as gene therapy monitoring for cerebellar neurodegenerative diseases. J Cereb Blood Flow Metab 2009; 29:434-40. [PMID: 19002197 DOI: 10.1038/jcbfm.2008.137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We investigated a gene expression imaging method to examine the level of therapeutic gene expression in the cerebellum. Using a human immunodeficiency virus derived lentivial vector, we expressed the dopamine D(2) receptor (D(2)R) as a reporter protein to mouse cerebellar Purkinje cells. Biodistribution and ex vivo autoradiography studies were performed by giving [(125)I]5-iodo-7-N-[(1-ethyl-2-pyrrolidinyl)methyl]carboxamide-2,3-dihydrobenzofuran ([(125)I]IBF) (1.85 MBq), as a radioactive D(2)R ligand, to model mice expressing the D(2)R with an HA tag (HA-D(2)R) in the cerebellum. In this study, [(125)I]IBF was bound to the D(2)R expressed in the cerebellum of the model mice selectively. Immunostaining was performed to confirm the HA-D(2)R expression in the cerebellum of the model mice. A significant correlation (r=0.900, P<0.001) between areas that expressed HA-D(2)R by immunostaining and areas in which [(125)I]IBF accumulated by the ex vivo autoradiograms was found. These results indicated that radioiodinated IBF is useful as a reporter probe to detect D(2)R reporter gene expression, which can be used for monitoring therapeutic gene expression in the cerebellum.
Collapse
Affiliation(s)
- Kazuhiro Shiba
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa University, Kanazawa, Ishikawa, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Brown DA, Kharkar PS, Parrington I, Reith ME, Dutta AK. Structurally constrained hybrid derivatives containing octahydrobenzo[g or f]quinoline moieties for dopamine D2 and D3 receptors: binding characterization at D2/D3 receptors and elucidation of a pharmacophore model. J Med Chem 2008; 51:7806-7819. [PMID: 19053758 PMCID: PMC2607046 DOI: 10.1021/jm8008629] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A series of structurally constrained analogues based on hybrid compounds containing octahydrobenzo[g or f]quinoline moieties were designed, synthesized, and characterized for their binding to dopamine D2 and D3 receptors expressed in HEK-293 cells. Among the newly developed constrained molecules, trans-octahydrobenzo[f]quinolin-7-ol (8) exhibited the highest affinity for D2 and D3 receptors, the (-)-isomer being the eutomer. Interestingly, this hybrid constrained version 8 showed significant affinity over the corresponding nonhybrid version 1 (representing a constrained version of the aminotetralin structure only) when assayed under same conditions (K(i) of 49.1 and 14.9 nM for 8 vs 380 and 96.0 nM for 1 at D2 and D3, respectively). Similar results were found with other lead hybrid compounds, indicating a contribution of the piperazine moiety in the observed enhanced affinity. On the basis of the data of new lead constrained derivatives and other lead hybrid derivatives developed by us, a unique pharmacophore model was proposed consisting of three pharmacophoric centers, two with aromatic/hydrophobic and one with cationic features.
Collapse
Affiliation(s)
- Dennis A. Brown
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202
| | - Prashant S. Kharkar
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202
| | - Ingrid Parrington
- New York University School of Medicine, Millhauser Laboratories, Department of Psychiatry, New York, NY 10016
| | - Maarten E.A. Reith
- New York University School of Medicine, Millhauser Laboratories, Department of Psychiatry, New York, NY 10016
| | - Aloke K. Dutta
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202
| |
Collapse
|
35
|
Sahlholm K, Marcellino D, Nilsson J, Fuxe K, Arhem P. Voltage-sensitivity at the human dopamine D2S receptor is agonist-specific. Biochem Biophys Res Commun 2008; 377:1216-21. [PMID: 18983826 DOI: 10.1016/j.bbrc.2008.10.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2008] [Accepted: 10/26/2008] [Indexed: 01/23/2023]
Abstract
Recently, we and others have shown that agonist potencies at some, but not all, G protein-coupled receptors are voltage-sensitive. Several of those studies employed electrophysiology assays in Xenopus oocytes with G protein-coupled potassium channels as a readout. Using this assay, we have now obtained evidence that voltage-sensitivity at the dopamine D(2S) receptor is agonist-specific. Whereas the potency of dopamine at the D(2S) receptor is decreased by depolarization, the potencies of beta-phenethylamine, p- and m-tyramine are voltage-insensitive. Furthermore, both monohydroxylated and non-hydroxylated N,N-dipropyl-2-aminotetralin compounds are voltage-sensitive. Differential activation of G protein subtypes or differential ratios between effector and active G protein do not underlie this agonist-selective voltage-sensitivity. This is the first demonstration of voltage-sensitive and voltage-insensitive behaviour of different agonists acting via the same receptor.
Collapse
Affiliation(s)
- Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
36
|
Selent J, López L, Sanz F, Pastor M. Multi-receptor binding profile of clozapine and olanzapine: a structural study based on the new beta2 adrenergic receptor template. ChemMedChem 2008; 3:1194-8. [PMID: 18465762 DOI: 10.1002/cmdc.200800074] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Jana Selent
- Research Unit on Biomedical Informatics (GRIB), IMIM/Universitat Pompeu Fabra, Avinguda Dr. Aiguader 88, 08003 Barcelona, Spain
| | | | | | | |
Collapse
|
37
|
Dukat M, Mosier PD, Kolanos R, Roth BL, Glennon RA. Binding of serotonin and N1-benzenesulfonyltryptamine-related analogs at human 5-HT6 serotonin receptors: receptor modeling studies. J Med Chem 2008; 51:603-11. [PMID: 18201064 DOI: 10.1021/jm070910s] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A population of 100 graphics models of the human 5-HT6 serotonin receptor was constructed based on the structure of bovine rhodopsin. The endogenous tryptamine-based agonist serotonin (5-HT; 1) and the benzenesulfonyl-containing tryptamine-derived 5-HT6 receptor antagonist MS-245 (4a) were automatically docked with each of the 100 receptor models using a genetic algorithm approach. Similar studies were conducted with the more selective 5-HT6 receptor agonist EMDT (5) and optical isomers of EMDT-related analog 8, as well as with optical isomers of MS-245 (4a)-related and benzenesulfonyl-containing pyrrolidine 6 and aminotetralin 7. Although associated with the same general aromatic/hydrophobic binding cluster, 5-HT (1) and MS-245 (4a) were found to preferentially bind with distinct receptor conformations, and did so with different binding orientations (i.e., poses). A 5-HT pose/model was found to be common to EMDT (5) and its analogs, whereas that identified for MS-245 (4a) was found common to benzenesulfonyl-containing compounds. Specific amino acid residues were identified that can participate in binding, and evaluation of a sulfenamide analog of MS-245 indicates for the first time that the presence of the sulfonyl oxygen atoms enhances receptor affinity. The results indicate that the presence or absence of an N1-benzenesulfonyl group is a major determinant of the manner in which tryptamine-related agents bind at 5-HT6 serotonin receptors.
Collapse
Affiliation(s)
- Małgorzata Dukat
- Department of Medicinal Chemistry, School of Pharmacy, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0540, USA
| | | | | | | | | |
Collapse
|
38
|
Kummer C, Winkeler A, Dittmar C, Bauer B, Rueger MA, Rueckriem B, Heneka MT, Vollmar S, Wienhard K, Fraefel C, Heiss WD, Jacobs AH. Multitracer Positron Emission Tomographic Imaging of Exogenous Gene Expression Mediated by a Universal Herpes Simplex Virus 1 Amplicon Vector. Mol Imaging 2007. [DOI: 10.2310/7290.2007.00015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Christiane Kummer
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Alexandra Winkeler
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Claus Dittmar
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Bernd Bauer
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Maria Adele Rueger
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Benedikt Rueckriem
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Michael T. Heneka
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Stefan Vollmar
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Klaus Wienhard
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Cornel Fraefel
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Wolf-Dieter Heiss
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| | - Andreas H. Jacobs
- From the Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Center for Molecular Medicine, and Department of Neurology, University of Cologne, Cologne, Germany
| |
Collapse
|
39
|
Fu W, Shen J, Luo X, Zhu W, Cheng J, Yu K, Briggs JM, Jin G, Chen K, Jiang H. Dopamine D1 receptor agonist and D2 receptor antagonist effects of the natural product (-)-stepholidine: molecular modeling and dynamics simulations. Biophys J 2007; 93:1431-41. [PMID: 17468175 PMCID: PMC1948031 DOI: 10.1529/biophysj.106.088500] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
(-)-Stepholidine (SPD), an active ingredient of the Chinese herb Stephania, is the first compound found to have dual function as a dopamine receptor D1 agonist and D2 antagonist. Insights into dynamical behaviors of D1 and D2 receptors and their interaction modes with SPD are crucial in understanding the structural and functional characteristics of dopamine receptors. In this study a computational approach, integrating protein structure prediction, automated molecular docking, and molecular dynamics simulations were employed to investigate the dual action mechanism of SPD on the D1 and D2 receptors, with the eventual aim to develop new drugs for treating diseases affecting the central nervous system such as schizophrenia. The dynamics simulations revealed the surface features of the electrostatic potentials and the conformational "open-closed" process of the binding entrances of two dopamine receptors. Potential binding conformations of D1 and D2 receptors were obtained, and the D1-SPD and D2-SPD complexes were generated, which are in good agreement with most of experimental data. The D1-SPD structure shows that the K-167_EL-2-E-302_EL-3 (EL-2: extracellular loop 2; EL-3: extracellular loop 3) salt bridge plays an important role for both the conformational change of the extracellular domain and the binding of SPD. Based on our modeling and simulations, we proposed a mechanism of the dual action of SPD and a subsequent signal transduction model. Further mutagenesis and biophysical experiments are needed to test and improve our proposed dual action mechanism of SPD and signal transduction model.
Collapse
Affiliation(s)
- Wei Fu
- Drug Discovery and Design Centre, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lan H, Durand CJ, Teeter MM, Neve KA. Structural determinants of pharmacological specificity between D(1) and D(2) dopamine receptors. Mol Pharmacol 2006; 69:185-94. [PMID: 16236817 DOI: 10.1124/mol.105.017244] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To test the hypothesis that pharmacological differentiation between D(1) and D(2) dopamine receptors results from interactions of selective ligands with nonconserved residues lining the binding pocket, we mutated amino acid residues in the D(2) receptor to the corresponding aligned residues in the D(1) receptor and vice versa and expressed the receptors in human embryonic kidney 293 cells. Determinations of the affinity of the 14 mutant D(2) receptors and 11 mutant D(1) receptors for D(1)- and D(2)-selective antagonists, and rhodopsin-based homology models of the two receptors, identified two residues whose direct interactions with certain ligands probably contribute to ligand selectivity. The D(1) receptor mutant W99(3.28)F showed dramatically increased affinity for several D(2)-selective antagonists, particularly spiperone (225-fold), whereas the D(2) receptor mutant Y417(7.43)W had greatly decreased affinity for benzamide ligands such as raclopride (200-fold) and sulpiride (125-fold). The binding of the D(1)-selective ligand R-(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine (SCH23390) was unaffected, indicating that SCH23390 makes little contact with these ancillary pocket residues. Mutation of A/V(5.39) caused modest but consistent and reciprocal changes in affinity of the receptors for D(1) and D(2)-selective ligands, perhaps reflecting altered packing of the interface of helices 5 and 6. We also obtained some evidence that residues in the second extracellular loop contribute to ligand binding. We conclude that additional determinants of D(1)/D(2) receptor-selective binding are located either in that loop or in the transmembrane helices but, like residue 5.39, indirectly influence the interactions of selective ligands with conserved residues by altering the shape of the primary and ancillary binding pockets.
Collapse
Affiliation(s)
- Hongxiang Lan
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | |
Collapse
|
41
|
Sugiura M, Fuke S, Suo S, Sasagawa N, Van Tol HHM, Ishiura S. Characterization of a novel D2-like dopamine receptor with a truncated splice variant and a D1-like dopamine receptor unique to invertebrates from Caenorhabditis elegans. J Neurochem 2005; 94:1146-57. [PMID: 16001968 DOI: 10.1111/j.1471-4159.2005.03268.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have cloned two novel Caenorhabditis elegans dopamine receptors, DOP-3 and DOP-4. DOP-3 shows high sequence homology with other D2-like dopamine receptors. As a result of alternative splicing, a truncated splice variant of DOP-3, DOP-3nf, was produced. Because of the in-frame insertion of a stop codon in the third intracellular loop, DOP-3nf lacks the sixth and seventh transmembrane domains that are found in the full-length DOP-3 receptor. Reporter gene assay showed that DOP-3 attenuates forskolin-stimulated cAMP formation in response to dopamine stimulation, whereas DOP-3nf does not. When DOP-3 was coexpressed with DOP-3nf, the ability to inhibit forskolin-stimulated cAMP formation was reduced. DOP-4 shows high sequence homology with D1-like dopamine receptors unique to invertebrates, which are distinct from mammalian D1-like dopamine receptors. Reporter gene assay showed that DOP-4 stimulates cAMP accumulation in response to dopamine stimulation. These two receptors provide new opportunities to understand dopaminergic signaling at the molecular level.
Collapse
Affiliation(s)
- Mai Sugiura
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Meguro-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Floresca CZ, Schetz JA. Dopamine receptor microdomains involved in molecular recognition and the regulation of drug affinity and function. J Recept Signal Transduct Res 2005; 24:207-39. [PMID: 15521362 DOI: 10.1081/rrs-200032088] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
A cationic protonatable amine moiety on dopaminergic ligands forms a high affinity reinforced ionic bond with an anionic aspartic acid at position 3.32 of dopamine receptors. When present, catechol hydroxyls of the ligands form hydrogen bonds with serines at position 5.42, 5.43, and 5.46, and this network of hydrogen bonds serves to orient ligands in the binding-site crevice and increase their binding affinity. A steric clash between aromatic moieties of the ligands and aromatic amino acids of the receptor (e.g., H6.55, F6.52 or F6.51 and W6.48) is likely to be propagated in domino-like fashion along the length of TM6, which is believed to trigger activation of the receptor. Specifically, it is the change in the conformation of W6.48 from an orientation perpendicular to the plane of the lipid membrane to one that is parallel that is believed to result in activation. Molecular determinants that mediate the D4/D2-selectivity of many extremely D4-selective 1,4-DAP ligands, include a nonconserved cluster of bulky amino acids at the TM2/TM3 interface (positions 2.61, 3.28 and 3.29).
Collapse
Affiliation(s)
- Christina Z Floresca
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107-2699, USA
| | | |
Collapse
|
43
|
Hjerde E, Dahl SG, Sylte I. Atypical and typical antipsychotic drug interactions with the dopamine D2 receptor. Eur J Med Chem 2005; 40:185-94. [PMID: 15694653 DOI: 10.1016/j.ejmech.2004.10.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2004] [Revised: 09/27/2004] [Accepted: 10/15/2004] [Indexed: 11/21/2022]
Abstract
A model of the dopamine D2 receptor was used to study the receptor interactions of dopamine, the typical antipsychotics haloperidol and loxapine, and the atypical antipsychotics clozapine and melperone. The atypical antipsychotics interacted with the halogen atom of the ring system in the direction of the transmembrane helices (TMHs) 2, 3 and 7, while the typical had the corresponding halogen atom in the direction of TMH5. Molecular dynamics simulations indicated that the average helical displacement upon binding increased in the order: typical < atypical < dopamine. Upon binding, the atypical induced larger displacements into TMH5 than did the typical. The typical had stronger non-bonded interactions with the receptor than had the atypical, which is in agreement with the experimental observation that the atypical antipsychotic drugs dissociate faster from the receptor than the typical antipsychotic drugs.
Collapse
Affiliation(s)
- Erik Hjerde
- Department of Pharmacology, Institute of Medical Biology, University of Tromsø, N-9037 Tromsø, Norway
| | | | | |
Collapse
|
44
|
Shacham S, Marantz Y, Bar-Haim S, Kalid O, Warshaviak D, Avisar N, Inbal B, Heifetz A, Fichman M, Topf M, Naor Z, Noiman S, Becker OM. PREDICT modeling and in-silico screening for G-protein coupled receptors. Proteins 2005; 57:51-86. [PMID: 15326594 DOI: 10.1002/prot.20195] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
G-protein coupled receptors (GPCRs) are a major group of drug targets for which only one x-ray structure is known (the nondrugable rhodopsin), limiting the application of structure-based drug discovery to GPCRs. In this paper we present the details of PREDICT, a new algorithmic approach for modeling the 3D structure of GPCRs without relying on homology to rhodopsin. PREDICT, which focuses on the transmembrane domain of GPCRs, starts from the primary sequence of the receptor, simultaneously optimizing multiple 'decoy' conformations of the protein in order to find its most stable structure, culminating in a virtual receptor-ligand complex. In this paper we present a comprehensive analysis of three PREDICT models for the dopamine D2, neurokinin NK1, and neuropeptide Y Y1 receptors. A shorter discussion of the CCR3 receptor model is also included. All models were found to be in good agreement with a large body of experimental data. The quality of the PREDICT models, at least for drug discovery purposes, was evaluated by their successful utilization in in-silico screening. Virtual screening using all three PREDICT models yielded enrichment factors 9-fold to 44-fold better than random screening. Namely, the PREDICT models can be used to identify active small-molecule ligands embedded in large compound libraries with an efficiency comparable to that obtained using crystal structures for non-GPCR targets.
Collapse
|
45
|
Abstract
The development of noninvasive imaging technologies designed specifically for use with small animals has provided new paradigms for cancer research. Traditional molecular biology techniques are being melded with noninvasive imaging technologies to develop a new research domain, "molecular imaging." One of the most exciting advances in this research area is the adaptation and application of conventional reporter-gene imaging techniques, used extensively by cell and molecular biologists, to living animals. Using these new assays, investigators can image noninvasively, repeatedly, and quantitatively the location, magnitude, and duration of reporter-gene expression in living animals. This review will describe the instrumentation used for noninvasive imaging of reporter genes, the reporter genes developed for noninvasive imaging with radio-nuclide-based assays such as positron emission tomography, and the reporter genes used for optically based noninvasive assays using sensitive charged-coupled device cameras. Applications of noninvasive, whole-animal imaging to gene therapy for cancer, to cell-based therapy for cancer, to lymphocyte activation, to cancer progression and dissemination in engrafted models, to tumor initiation, promotion and metastasis in conditional murine models of cancer induction, and to the noninvasive monitoring of tumor responses to a variety of therapies are described. New developments in multimodality molecular imaging are discussed, and the potential utility of noninvasive reporter gene expression in the diagnosis and management of human cancer is presented.
Collapse
Affiliation(s)
- Harvey R Herschman
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Johnsson Comprehensive Cancer Center, Los Angeles, California 90095, USA
| |
Collapse
|
46
|
Abstract
The binding site in G protein-coupled cationic biogenic amine receptors is formed in the cleft of the seven transmembrane segments. Upon binding the ligand, the receptors are activated or inactivated through the conformational changes of the transmembrane segments. G protein-coupled receptors bind four functionally distinct ligands; inverse agonists, antagonists, partial agonists, and full agonists. Hence, putative structural models for biogenic amine receptors corresponding to the ligand function (inverse agonist-, antagonist-, partial agonist-, and full agonist-bound receptor models) were built by using photointermediate models in the rhodopsin photocascade (M. Ishiguro et al. ChemBioChem. 2004, 5, 298-310). The ligand-receptor recognition of each was examined by modeling receptor-ligand complexes with functional ligands. The complex models suggested that each functional ligand binds the corresponding receptor structure and that ligand-specific interactions contribute to stabilization of the corresponding receptor structure.
Collapse
MESH Headings
- Amino Acids/analysis
- Computational Biology
- Ligands
- Models, Molecular
- Protein Transport/physiology
- Receptor, Serotonin, 5-HT2A/chemistry
- Receptor, Serotonin, 5-HT2A/metabolism
- Receptors, Adrenergic/chemistry
- Receptors, Adrenergic/metabolism
- Receptors, Biogenic Amine/agonists
- Receptors, Biogenic Amine/metabolism
- Receptors, Dopamine/chemistry
- Receptors, Dopamine/metabolism
- Receptors, Histamine H2/chemistry
- Receptors, Histamine H2/metabolism
- Receptors, Muscarinic/chemistry
- Receptors, Muscarinic/metabolism
Collapse
Affiliation(s)
- Masaji Ishiguro
- Suntory Institute for Bioorganic Research, 1-1 Wakayamadai, Shimamoto, Osaka 618-8503, Japan.
| |
Collapse
|
47
|
Cordeaux Y, IJzerman AP, Hill SJ. Coupling of the human A1 adenosine receptor to different heterotrimeric G proteins: evidence for agonist-specific G protein activation. Br J Pharmacol 2004; 143:705-14. [PMID: 15302686 PMCID: PMC1575922 DOI: 10.1038/sj.bjp.0705925] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The present study investigates the effect of varying ligand structure on the ability of agonists to activate guanine nucleotide-binding proteins of the Gi, Gs and Gq families via the A(1) adenosine receptor. In CHO cells expressing this receptor, inhibition or potentiation of forskolin-stimulated cAMP accumulation was used as an end point to measure the activation of Gi and, in Pertussis toxin (PTX)-treated cells, Gs, respectively. Stimulation of inositol phosphate accumulation in PTX-treated cells was used as an index of Gq activation. CPA (N(6)-cyclopentyladenosine), NECA (5'-N-ethyl-carboxyamidoadenosine) and eight analogues of these ligands presented a range of guanine nucleotide-binding protein (G-protein)-activating profiles. Some ligands could only activate Gi (e.g. 2'deoxyCPA), some primarily Gi and Gs (and only weakly Gq) (e.g. 3'deoxyCPA), highlighting the importance of the ribose hydroxyls in agonist activation of multiple G proteins. CHA (N(6)-cyclohexyladenosine) activated Gi, Gs and Gq, but was more efficacious than CPA in activating Gs. The NECA analogues 5'-N-cyclopropyl-carboxamidoadenosine, 5'-N-cyclobutyl-carboxamidoadenosine and 5'-N-cyclopentyl-carboxamidoadenosine (CPeCA) also activated all three G proteins, although their ability to activate Gs and Gq (relative to CPA) was reduced with increasing substituent size, such that CPeCA produced only a small stimulation (at 100 microM) at Gq, but was a full agonist, relative to CPA, at Gi and Gs. This study suggests that the A(1) adenosine receptor can adopt agonist-specific conformations, arising from small changes in ligand structure, which lead to the differential activation of Gi, Gs and Gq.
Collapse
Affiliation(s)
- Yolande Cordeaux
- Institute of Cell Signalling, Medical School, University of Nottingham, Nottingham NG7 2UH
| | - Adriaan P IJzerman
- Leiden/Amsterdam Centre for Drug research, Division of Medicinal Chemistry, Leiden, Netherlands
| | - Stephen J Hill
- Institute of Cell Signalling, Medical School, University of Nottingham, Nottingham NG7 2UH
- Author for correspondence:
| |
Collapse
|
48
|
Abstract
Non-invasive, quantitative and repetitive imaging of biological processes in living animals is rapidly changing the way in which many experiments in models of human disease and normal physiological processes are conducted. This review summarizes the newest molecular imaging approaches to analyzing reporter gene expression, with particular emphasis on pre-clinical cancer research. Alternative modes of imaging are summarized, followed by descriptions of the major reporter gene systems now used for radionuclide imaging in vivo of gene expression. Several somatic delivery paradigms for co-ordinate expression of therapeutic and imaging genes are presented, and our own emphasis on the dopamine D2 receptor and Herpes Simplex Virus Type I thymidine kinase reporter genes are detailed.
Collapse
Affiliation(s)
- Harvey R Herschman
- Department of Biological Chemistry, Director for Basic Research, UCLA Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.
| |
Collapse
|
49
|
Oliveira L, Paiva PB, Paiva ACM, Vriend G. Sequence analysis reveals how G protein-coupled receptors transduce the signal to the G protein. Proteins 2003; 52:553-60. [PMID: 12910455 DOI: 10.1002/prot.10489] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Sequence entropy-variability plots based on alignments of very large numbers of sequences-can indicate the location in proteins of the main active site and modulator sites. In the previous article in this issue, we applied this observation to a series of well-studied proteins and concluded that it was possible to detect most of the residues with a known functional role. Here, we apply the method to rhodopsin-like G protein-coupled receptors. Our conclusion is that G protein binding is the main evolutionary constraint on these receptors, and that other ligands, such as agonists, act as modulators. The activation of the receptors can be described as a simple, two-step process, and the residues involved in signal transduction can be identified.
Collapse
Affiliation(s)
- Laerte Oliveira
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
50
|
Abstract
Positron emission tomography (PET) has been used clinically to measure enzyme reactions, ligand-receptor interactions, cellular metabolism and cell proliferation. Until recently, however, PET has not been suitable for small animal models because of resolution limitations. Development of micro-PET instrumentation for small animal imaging and the availability of positron-emitting tracers has made this technology accessible for the non-invasive, quantitative and repetitive imaging of biological function in living animals. The development of new probes and positron-imaging based reporter genes has extended micro-PET applications to investigations of metabolism, enzyme activity, receptor-ligand interactions, protein-protein interactions, gene expression, adoptive cell therapy and somatic gene therapy. Because small animal PET is immediately extrapolatable to the clinic, laboratory advances should rapidly be translated to clinical practice.
Collapse
Affiliation(s)
- Harvey R Herschman
- Department of Biological Chemistry, Molecular Biology Institute, Crump Institute and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, 90095, USA.
| |
Collapse
|