Molecular Determinants of Allosteric Modulation at the M1 Muscarinic Acetylcholine Receptor*

Background: BQCA is a selective allosteric modulator of the M1 mAChR. Results: Residues that govern BQCA activity were identified using mutagenesis and molecular modeling. Conclusion: BQCA likely occupies a pocket overlapping prototypical mAChR modulators and gains selectivity through cooperativity with orthosteric ligands. Significance: Understanding the structural basis of BQCA function can provide insight into the design of more tailored allosteric ligands. Benzylquinolone carboxylic acid (BQCA) is an unprecedented example of a selective positive allosteric modulator of acetylcholine at the M1 muscarinic acetylcholine receptor (mAChR). To probe the structural basis underlying its selectivity, we utilized site-directed mutagenesis, analytical modeling, and molecular dynamics to delineate regions of the M1 mAChR that govern modulator binding and transmission of cooperativity. We identified Tyr-852.64 in transmembrane domain 2 (TMII), Tyr-179 and Phe-182 in the second extracellular loop (ECL2), and Glu-3977.32 and Trp-4007.35 in TMVII as residues that contribute to the BQCA binding pocket at the M1 mAChR, as well as to the transmission of cooperativity with the orthosteric agonist carbachol. As such, the BQCA binding pocket partially overlaps with the previously described “common” allosteric site in the extracellular vestibule of the M1 mAChR, suggesting that its high subtype selectivity derives from either additional contacts outside this region or through a subtype-specific cooperativity mechanism. Mutation of amino acid residues that form the orthosteric binding pocket caused a loss of carbachol response that could be rescued by BQCA. Two of these residues (Leu-1023.29 and Asp-1053.32) were also identified as indirect contributors to the binding affinity of the modulator. This new insight into the structural basis of binding and function of BQCA can guide the design of new allosteric ligands with tailored pharmacological properties.

[1]  Albert C. Pan,et al.  Structural basis for modulation of a G-protein-coupled receptor by allosteric drugs , 2013, Nature.

[2]  Wei Zhang,et al.  A point‐charge force field for molecular mechanics simulations of proteins based on condensed‐phase quantum mechanical calculations , 2003, J. Comput. Chem..

[3]  A. Christopoulos Assessing the distribution of parameters in models of ligand-receptor interaction: to log or not to log. , 1998, Trends in pharmacological sciences.

[4]  J. Black,et al.  Operational models of pharmacological agonism , 1983, Proceedings of the Royal Society of London. Series B. Biological Sciences.

[5]  E C Hulme,et al.  The Functional Topography of Transmembrane Domain 3 of the M1 Muscarinic Acetylcholine Receptor, Revealed by Scanning Mutagenesis* , 1999, The Journal of Biological Chemistry.

[6]  Laxmikant V. Kalé,et al.  Scalable molecular dynamics with NAMD , 2005, J. Comput. Chem..

[7]  R. Stevens,et al.  Structural Features for Functional Selectivity at Serotonin Receptors , 2013, Science.

[8]  P. Sexton,et al.  Structural Determinants of Allosteric Agonism and Modulation at the M4 Muscarinic Acetylcholine Receptor , 2010, The Journal of Biological Chemistry.

[9]  P. Sexton,et al.  A Monod-Wyman-Changeux Mechanism Can Explain G Protein-coupled Receptor (GPCR) Allosteric Modulation* , 2011, The Journal of Biological Chemistry.

[10]  P. Sexton,et al.  Allosteric GPCR modulators: taking advantage of permissive receptor pharmacology. , 2007, Trends in pharmacological sciences.

[11]  M. Brann,et al.  Identification of a Ligand-dependent Switch within a Muscarinic Receptor* , 1998, The Journal of Biological Chemistry.

[12]  A. Kruse,et al.  Structure of the human M2 muscarinic acetylcholine receptor bound to an antagonist , 2011, Nature.

[13]  S. Jadhav,et al.  Discovery and characterization of novel subtype-selective allosteric agonists for the investigation of M(1) receptor function in the central nervous system. , 2010, ACS chemical neuroscience.

[14]  J. Saldanha,et al.  Structure and activation of muscarinic acetylcholine receptors. , 2001, Biochemical Society transactions.

[15]  Arthur Christopoulos,et al.  Orthosteric and Allosteric Modes of Interaction of Novel Selective Agonists of the M1 Muscarinic Acetylcholine Receptor , 2010, Molecular Pharmacology.

[16]  A. Christopoulos,et al.  Fitting Models to Biological Data Using Linear and Nonlinear Regression: A Practical Guide to Curve Fitting , 2004 .

[17]  Willem Soudijn,et al.  Allosteric modulation of G-protein-coupled receptors , 2001 .

[18]  Hualiang Jiang,et al.  Structure of the CCR5 Chemokine Receptor–HIV Entry Inhibitor Maraviroc Complex , 2013, Science.

[19]  Oliver P. Ernst,et al.  Crystal structure of metarhodopsin II , 2011, Nature.

[20]  S. Jadhav,et al.  Discovery and characterization of novel subtype-selective allosteric agonists for the investigation of M(1) receptor function in the central nervous system. , 2010, ACS chemical neuroscience.

[21]  Xi-Ping Huang,et al.  Critical Amino Acid Residues of the Common Allosteric Site on the M2 Muscarinic Acetylcholine Receptor: More Similarities than Differences between the Structurally Divergent Agents Gallamine and Bis(ammonio)alkane-Type Hexamethylene-bis-[dimethyl-(3-phthalimidopropyl)ammonium]dibromide , 2005, Molecular Pharmacology.

[22]  B. Roth,et al.  New Insights into the Function of M4 Muscarinic Acetylcholine Receptors Gained Using a Novel Allosteric Modulator and a DREADD (Designer Receptor Exclusively Activated by a Designer Drug) , 2008, Molecular Pharmacology.

[23]  S. Lazareno,et al.  Probing of the location of the allosteric site on m1 muscarinic receptors by site-directed mutagenesis. , 1995, Molecular pharmacology.

[24]  Thomas M Frimurer,et al.  PheVI:09 (Phe6.44) as a Sliding Microswitch in Seven-transmembrane (7TM) G Protein-coupled Receptor Activation* , 2012, The Journal of Biological Chemistry.

[25]  S. Rasmussen,et al.  Crystal Structure of the β2Adrenergic Receptor-Gs protein complex , 2011, Nature.

[26]  C. Langmead,et al.  Structure-Function Studies of Allosteric Agonism at M2 Muscarinic Acetylcholine Receptors , 2007, Molecular Pharmacology.

[27]  Roberto Maass-Moreno,et al.  Fitting Models to Biological Data Using Linear and Nonlinear Regression: A Practical Guide to Curve Fitting.ByHarvey Motulskyand, Arthur Christopoulos.Oxford and New York: Oxford University Press. $65.00 (hardcover); $29.95 (paper). 351 p; ill.; index. ISBN: 0–19–517179–9 (hc); 0–19–517180–2 (pb). 2 , 2005 .

[28]  A. Leslie,et al.  Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation , 2011, Nature.

[29]  R. Stevens,et al.  High-Resolution Crystal Structure of an Engineered Human β2-Adrenergic G Protein–Coupled Receptor , 2007, Science.

[30]  Albert C. Pan,et al.  Structure and Dynamics of the M3 Muscarinic Acetylcholine Receptor , 2012, Nature.

[31]  E. Hulme,et al.  GPCR activation: a mutagenic spotlight on crystal structures. , 2013, Trends in pharmacological sciences.

[32]  Craig W. Lindsley,et al.  Selective activation of the M1 muscarinic acetylcholine receptor achieved by allosteric potentiation , 2009, Proceedings of the National Academy of Sciences.

[33]  C. Langmead,et al.  Roof and Floor of the Muscarinic Binding Pocket: Variations in the Binding Modes of Orthosteric Ligands , 2007, Molecular Pharmacology.

[34]  K. Mohr,et al.  Allosteric Interactions with Muscarinic Acetylcholine Receptors: Complex Role of the Conserved Tryptophan M2422Trp in a Critical Cluster of Amino Acids for Baseline Affinity, Subtype Selectivity, and Cooperativity , 2006, Molecular Pharmacology.

[35]  John Ellis,et al.  Allosteric site on muscarinic acetylcholine receptors: identification of two amino acids in the muscarinic M2 receptor that account entirely for the M2/M5 subtype selectivities of some structurally diverse allosteric ligands in N-methylscopolamine-occupied receptors. , 2003, Molecular pharmacology.

[36]  M. Quirk,et al.  A Selective Allosteric Potentiator of the M1 Muscarinic Acetylcholine Receptor Increases Activity of Medial Prefrontal Cortical Neurons and Restores Impairments in Reversal Learning , 2009, The Journal of Neuroscience.

[37]  J W Saldanha,et al.  Transmembrane Domains 4 and 7 of the M1Muscarinic Acetylcholine Receptor Are Critical for Ligand Binding and the Receptor Activation Switch* , 2001, The Journal of Biological Chemistry.

[38]  Junmei Wang,et al.  Development and testing of a general amber force field , 2004, J. Comput. Chem..

[39]  S. Rasmussen,et al.  Structure of a nanobody-stabilized active state of the β2 adrenoceptor , 2010, Nature.

[40]  P. Sexton,et al.  Probe Dependence in the Allosteric Modulation of a G Protein-Coupled Receptor: Implications for Detection and Validation of Allosteric Ligand Effects , 2012, Molecular Pharmacology.

[41]  A. Christopoulos,et al.  Development of M1 mAChR allosteric and bitopic ligands: prospective therapeutics for the treatment of cognitive deficits. , 2013, ACS chemical neuroscience.

[42]  Arthur Christopoulos,et al.  A Novel Mechanism of G Protein-coupled Receptor Functional Selectivity , 2008, Journal of Biological Chemistry.

[43]  J. Ballesteros,et al.  [19] Integrated methods for the construction of three-dimensional models and computational probing of structure-function relations in G protein-coupled receptors , 1995 .

[44]  Ruben Abagyan,et al.  The GPCR Network: a large-scale collaboration to determine human GPCR structure and function , 2012, Nature Reviews Drug Discovery.

[45]  Catherine Marquer,et al.  Structural Model of Ligand-G Protein-coupled Receptor (GPCR) Complex Based on Experimental Double Mutant Cycle Data , 2011, The Journal of Biological Chemistry.

[46]  Arthur Christopoulos,et al.  Signalling bias in new drug discovery: detection, quantification and therapeutic impact , 2012, Nature Reviews Drug Discovery.

[47]  Helgi B. Schiöth,et al.  Structural diversity of G protein-coupled receptors and significance for drug discovery , 2008, Nature Reviews Drug Discovery.

[48]  M. Babu,et al.  Molecular signatures of G-protein-coupled receptors , 2013, Nature.

[49]  P. Sexton,et al.  Allosteric Modulation of a Chemogenetically Modified G Protein-Coupled Receptor , 2013, Molecular Pharmacology.

[50]  T. Blundell,et al.  Comparative protein modelling by satisfaction of spatial restraints. , 1993, Journal of molecular biology.

[51]  P. Sexton,et al.  The Role of Transmembrane Domain 3 in the Actions of Orthosteric, Allosteric, and Atypical Agonists of the M4 Muscarinic Acetylcholine Receptor , 2011, Molecular Pharmacology.

[52]  J. Thompson,et al.  The CLUSTAL_X windows interface: flexible strategies for multiple sequence alignment aided by quality analysis tools. , 1997, Nucleic acids research.

[53]  J Andrew McCammon,et al.  Activation and dynamic network of the M2 muscarinic receptor , 2013, Proceedings of the National Academy of Sciences.

[54]  C. Lindsley,et al.  Subtype-selective allosteric modulators of muscarinic receptors for the treatment of CNS disorders. , 2009, Trends in pharmacological sciences.

[55]  P. Sexton,et al.  Molecular Mechanisms of Action and In Vivo Validation of an M4 Muscarinic Acetylcholine Receptor Allosteric Modulator with Potential Antipsychotic Properties , 2010, Neuropsychopharmacology.

[56]  U. Holzgrabe,et al.  Allosteric ligands for G protein‐coupled receptors: A novel strategy with attractive therapeutic opportunities , 2010, Medicinal research reviews.

[57]  Esther Kellenberger,et al.  Exploration of the Orthosteric/Allosteric Interface in Human M1 Muscarinic Receptors by Bitopic Fluorescent Ligands , 2013, Molecular Pharmacology.

[58]  Shailesh N Mistry,et al.  Synthesis and pharmacological profiling of analogues of benzyl quinolone carboxylic acid (BQCA) as allosteric modulators of the M1 muscarinic receptor. , 2013, Journal of medicinal chemistry.

[59]  A. Christopoulos,et al.  A structure-activity analysis of biased agonism at the dopamine D2 receptor. , 2013, Journal of medicinal chemistry.

[60]  Arthur Christopoulos,et al.  Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders , 2009, Nature Reviews Drug Discovery.