丁香实验_LOGO
登录
提问
我要登录
|免费注册
点赞
收藏
wx-share
分享

Quantification of Allosteric Interactions at G Protein–Coupled Receptors Using Radioligand Binding Assays

互联网

1207
  • Abstract
  • Table of Contents
  • Materials
  • Figures
  • Literature Cited

Abstract

 

Allosteric interactions involve the simultaneous binding of two ligands to the same receptor. An allosteric modulator causes a conformational change in the receptor protein that yields a change in the binding or signaling of an orthosteric agent, i.e., an agonist or competitive antagonist that binds to the endogenous agonist binding site. Because of the complex nature of allosteric phenomena, the detection and quantification of their effects on orthosteric ligand binding relies on the use of both equilibrium and non?equilibrium assays to ensure proper interpretation of the findings. Outlined in this unit are the most common experimental approaches for measuring allosteric effects on orthosteric ligand affinity at G protein?coupled receptors. There is also a discussion of the analysis of experimental data derived from such assays. Curr. Protoc. Pharmacol. 52:1.22.1?1.22.41. © 2011 by John Wiley & Sons, Inc.

Keywords: allosterism; allosteric interaction; cooperativity; radioligand binding; G protein coupled?receptors; dissociation kinetics; non?equilibrium

     
 
GO TO THE FULL PROTOCOL:
PDF or HTML at Wiley Online Library

Table of Contents

  • Introduction
  • Basic Protocol 1: Measurement of Allosteric Modulation of Radioligand Binding: Saturation Experiments
  • Alternate Protocol 1: Determination of the Affinity Ratio
  • Basic Protocol 2: Measurement of Allosteric Modulation of Radioligand Binding: Inhibition (or Potentiation) Experiments
  • Alternate Protocol 2: Measurement of Allosteric Modulation of Radioligand Binding: Inhibition (or Potentiation) Experiments Under Non‐Equilibrium Conditions
  • Basic Protocol 3: Measurement of Allosteric Modulation of Radioligand Binding to Cloned Receptors in Membranes: Dissociation Kinetic Studies Using Isotopic Dilution
  • Alternate Protocol 3: “Two‐Point” Kinetic Experiments
  • Alternate Protocol 4: Measurement of Allosteric Modulation of Radioligand Binding: Dissociation Kinetic Studies Using “Infinite Dilution” in Buffer
  • Support Protocol 1: Data Analysis
  • Commentary
  • Literature Cited
  • Figures
  • Tables
     
 
GO TO THE FULL PROTOCOL:
PDF or HTML at Wiley Online Library

Materials

Basic Protocol 1: Measurement of Allosteric Modulation of Radioligand Binding: Saturation Experiments

  Materials
  • Appropriate membrane preparation (see other units of Chapter 1)
  • Assay buffer (e.g., HEPES or Tris‐based buffers)
  • Radioligand
  • Test compound (allosteric modulator)
  • Unlabeled (non‐radioactive) competitive (orthosteric) ligand for determination of nonspecific binding
  • Wash buffer (usually the same as the assay buffer)
  • Scintillation cocktail (e.g., Packard Ultima Gold; Wallac HiSafe)
  • 12 × 75–mm glass or polypropylene culture tubes
  • Shaking water bath
  • Glass fiber filters (e.g., Whatman GF/B)
  • Cell harvester (e.g., Brandell, http://www.brandel.com/, or Skatron)
  • Scintillation counter and vials
  • Additional reagents and equipment for determination of protein concentration ( appendix 3A )

Alternate Protocol 1: Determination of the Affinity Ratio

  Materials
  • Appropriate membrane preparation (see other units of Chapter 1)
  • Assay buffer (e.g., HEPES or Tris‐based buffers)
  • Radioligand
  • Test compound (allosteric modulator)
  • Unlabeled (non‐radioactive) competitive (orthosteric) ligand for determination of nonspecific binding
  • Wash buffer (usually the same as the assay buffer), ice cold
  • Scintillation cocktail (e.g., Packard Ultima Gold; Wallac HiSafe)
  • 12 × 75–mm glass or polypropylene culture tubes
  • Shaking water bath
  • Glass fiber filters (e.g., Whatman GF/B)
  • Cell harvester (e.g., Brandell, http://www.brandel.com/, or Skatron)
  • Scintillation counter and vials
  • Additional reagents and equipment for determination of protein concentration ( appendix 3A )

Basic Protocol 2: Measurement of Allosteric Modulation of Radioligand Binding: Inhibition (or Potentiation) Experiments

  Materials
  • Appropriate membrane preparation (see other units of Chapter 1)
  • Assay buffer (e.g., HEPES or Tris‐based buffers)
  • Radioligand
  • Test compound (allosteric modulator)
  • Unlabeled (non‐radioactive) competitive (orthosteric) ligand for determination of nonspecific binding
  • Wash buffer (usually the same as the assay buffer), ice cold
  • Scintillation cocktail (e.g., Packard Ultima Gold; Wallac HiSafe)
  • 12 × 75–mm glass or polypropylene culture tubes
  • Shaking water bath
  • Glass fiber filters (e.g., Whatman GF/B)
  • Cell harvester (e.g., Brandell, http://www.brandel.com/, or Skatron)
  • Scintillation counter and vials
  • Additional reagents and equipment for determination of protein concentration ( appendix 3A )

Alternate Protocol 2: Measurement of Allosteric Modulation of Radioligand Binding: Inhibition (or Potentiation) Experiments Under Non‐Equilibrium Conditions

  • Sampling manifold (e.g., Millipore 1225) that allows for individual samples to be filtered at different times
  • Flask for holding large volumes of buffer
  • Magnetic stirrer to maintain adequate mixing of the buffer at the desired temperature
GO TO THE FULL PROTOCOL:
PDF or HTML at Wiley Online Library

Figures

  •   Figure 1.22.1 Schematic representation of the non‐equilibrium binding assay methodology. Tubes in “Set A” are exposed to prelabeled receptors, whereas tubes in “Set B” are exposed to the receptor and radioligand at the same time.
    View Image
  •   Figure 1.22.2 Analysis of the saturation binding data obtained for the antagonist, [3 H]N‐methylscopolamine ([3 H]NMS), at the cloned human muscarinic M2 receptor in the absence or presence of the indicated concentrations of the allosteric modulator, gallamine. Experimental data are taken from Table . (A ) Constrained, simultaneous nonlinear regression analysis, using the program GraphPad PRISM, according to Equation . (B ) Scatchard (Rosenthal) transformation of the same data. Note the reduction in slope (1/ K App ) with no significant change in B max . (C ) Nonlinear regression analysis of the data using GraphPad PRISM, according to Equation followed by Equation . (D ) Nonlinear regression analysis of the data using GraphPad PRISM, according to Equation followed by Equation . In each instance, the relevant parameter estimates derived from the curve‐fitting procedure are shown.
    View Image
  •   Figure 1.22.3 Example of the affinity‐ratio assay for the interaction of the allosteric modulator N‐benzyl brucine at the human M2 and M3 muscarinic acetylcholine receptors expressed in CHO cells. (A ) Effects of N‐benzyl brucine on the specific binding of [3 H]N‐methylscopolamine ([3 H]NMS (0.4 nM at the M2 receptor and 0.2 nM at the M3 receptor) in the absence or presence of unlabeled acetylcholine (ACh; 2 µM at the M2 receptor and 20 µM at the M3 receptor) in the presence of 0.2 mM GTP, and on [3 H]ACh binding to the M2 receptor measured in the absence of GTP. Incubation was at 30°C. (B ) Conversion of the data shown in A to affinity ratios. The fractional effect of N‐benzyl brucine on the binding of [3 H]ACh at M2 receptors is also shown. Note the ability of the affinity ratio plot to display the direction of the cooperativity, as well as a semi‐quantitative estimate of the magnitude of the cooperativity. Taken from Lazareno et al. ().
    View Image
  •   Figure 1.22.4 Equations that may be utilized to directly fit the ternary complex model to experimental binding data derived from equilibrium binding studies.
    View Image
  •   Figure 1.22.5 Analysis of the potentiation of [3 H]N‐methylscopolamine (0.1 nM) binding (LogKA = –9.52) by the allosteric modulator, alcuronium, at the cloned M2 muscarinic acetylcholine receptor. Incubation was for 5 hr at 37°C. The curve was analyzed using GraphPad PRISM according to Equation .
    View Image
  •   Figure 1.22.6 Analysis of the inhibition of binding of a low (0.12 nM, solid circles) and a high (1 nM, open circles) concentration of [3 H]N‐methylscopolamine (LogKA = –9.52) by the allosteric modulator, gallamine, at the cloned M2 muscarinic acetylcholine receptor. Data are taken from Table . Incubation was for 90 min at 37°C. Analysis was performed using a constrained, simultaneous nonlinear regression of both experimental datasets to Equation using the program GraphPad PRISM. Inset: the same data are shown normalized as percentage of respective control radioligand binding. This better illustrates the limited ability of weak negative allosteric modulators to fully inhibit radioligand binding when the concentration of the latter is increased.
    View Image
  •   Figure 1.22.7 Nonequilibrium assay of the effects of the allosteric modulator, McN‐A‐343, at the human M2 muscarinic acetylcholine receptor expressed in CHO cells. Receptors were either unlabeled or prelabeled with [3 H]N‐methylscopoloamine, as indicated, before the incubation with McN‐A‐343. The final radioligand concentration after dilution was 0.3 nM, and the incubation time was 1 hr and 20 min. The incubation temperature was 37°C. The lines represent the best fit, via constrained, simultaneous nonlinear regression, to Equation .
    View Image
  •   Figure 1.22.8 Effect of the allosteric modulator, heptane‐1,7‐bis‐(dimethyl‐3‐phthalimidopropyl)‐ammonium bromide (C7 /3‐phth), on the dissociation rate of [3 H]N‐methylscopolamine in CHO cell membranes expressing the cloned M2 muscarinic receptor. Membranes were incubated with 0.2 nM [3 H]N‐methylscopolamine at 37°C for 60 min before dissociation was monitored after the addition of 1 µM atropine alone or in combination with C7 /3‐phth at the indicated concentrations. Main figure: normalized curves represent the best fit via constrained, simultaneous nonlinear regression analysis, using GraphPad PRISM, according to Equations and . Inset: nonlinear regression analysis of the same data set using GraphPad PRISM according to Equations and .
    View Image
  •   Figure 1.22.9 Ternary complex model of allosteric interaction. R denotes the receptor, A and B denote the orthosteric and allosteric ligands, respectively; K A and K B denote the equilibrium dissociation constants of AR and BR, respectively. The symbol α denotes the cooperativity factor, and is a quantitative measure of the maximal, reciprocal alteration of the equilibrium dissociation constant of A and B for their respective binding sites, when both ligands bind concomitantly to form the ternary complex ARB.
    View Image
  •   Figure 1.22.10 Flow chart strategy for dealing with compounds identified as potential allosteric modulators.
    View Image

Videos

Literature Cited

   Abbott, A.J. and Nelsestuen, G.L. 1988. The collisional limit: An important consideration for membrane‐associated enzymes and receptors. FASEB J. 2:2858‐2866.
   Avlani, V.A., May, L.T., Sexton, P.M., and Christopoulos, A. 2004. Application of a kinetic model to the apparently complex behavior of negative and positive allosteric modulators. J. Pharmacol. Exp. Ther. 308:1062‐1072.
   Avlani, V.A., McLoughlin, D.J., Sexton, P.M., and Christopoulos, A. 2008. The impact of orthosteric radioligand depletion on the quantification of allosteric modulator interactions. J. Pharmacol. Exp. Ther. 325:927‐934.
   Birdsall, N.J., Lazareno, S., and Matsui, H. 1996. Allosteric regulation of muscarinic receptors. Prog. Brain Res. 109:147‐151.
   Bruns, R.F. and Fergus, J.H. 1990. Allosteric enhancement of adenosine A1 receptor binding and function by 2‐amino‐3‐benzoylthiophenes. Mol. Pharmacol. 38:939‐949.
   Changeux, J.‐P. 1966. Responses of acetylcholinesterase from Torpedo marmorata to salts and curarizing drugs. Mol. Pharmacol. 2:369‐392.
   Changeux, J.‐P. 1993. Allosteric proteins: From enzymes to receptors. Bioessays 15:625‐634.
   Christopoulos, A. 1998. Assessing the distribution of parameters in models of ligand‐receptor interaction: To log or not to log. Trends Pharmacol. Sci. 19:351‐357.
   Christopoulos, A., Lanzafame, A., and Mitchelson, F. 1998. Allosteric interactions at muscarinic cholinoceptors. Clin. Exp. Pharmacol. Physiol. 25:184‐194.
   Christopoulos, A. and Kenakin, T. 2002. G protein‐coupled receptor allosterism and complexing. Pharmacol. Rev. 54:323‐374.
   Colquhoun, D. 1973. The relation between classical and cooperative models for drug action. In Drug Receptors (H.P. Rang, ed.) pp. 149‐182. London: Macmillan Press.
   Conn, P.J., Christopoulos, A., and Lindsley, C.W. 2009. Allosteric modulators of GPCRs: A novel approach for the treatment of CNS disorders. Nat. Rev. Drug Discov. 8:41‐54.
   De Lean, A., Stadel, J.M., and Lefkowitz, R.J. 1980. A ternary complex model explains the agonist‐specific binding properties of the adenylate cyclase‐coupled β‐adrenergic receptor. J. Biol. Chem. 255:7108‐7117.
   Ehlert, F.J. 1988. Estimation of the affinities of allosteric ligands using radioligand binding and pharmacological null methods. Mol. Pharmacol. 33:187‐194.
   Ehlert, F.J., Roeske, W.R., Gee, K.W., and Yamamura, H.I. 1983. An allosteric model for benzodiazepine receptor function. Biochem. Pharmacol. 32:2375‐2383.
   Ellis, J. 1997. Allosteric binding sites on muscarinic receptors. Drug Dev. Res. 40:193‐204.
   Gerlach, L.O., Skerlj, R.T., Bridger, G.J., and Schwartz, T.W. 2001. Molecular interactions of cyclam and bicyclam non‐peptide antagonists with the CXCR4 chemokine receptor. J. Biol. Chem. 276:14153‐14160.
   Gregory, K.J., Sexton, P.M., and Christopoulos, A. 2007. Allosteric modulation of muscarinic acetylcholine receptors. Curr. Neuropharmacol. 5:157‐167.
   Hoare, S.R.J. and Strange, P.G. 1996. Regulation of D2 dopamine receptors by amiloride and amiloride analogs. Mol. Pharmacol. 50:1295‐1308.
   Holzgrabe, U. and Mohr, K. 1998. Allosteric modulators of ligand binding to muscarinic acetylcholine receptors. Drug Disc. Today 3:214‐222.
   Jensen, A.A. and Brauner‐Osborne, H. 2007. Allosteric modulation of the calcium‐sensing receptor. Curr. Neuropharmacol. 5:180‐186.
   Karlin, A. 1967. On the application of “a Plausible Model” of allosteric proteins to the receptor for acetylcholine. J. Theor. Biol. 16:306‐320.
   Kaumann, A.J. and Frenken, M. 1985. A paradox: The 5‐HT2‐receptor antagonist ketanserin restores the 5‐HT‐induced contraction depressed by methysergide in large coronary arteries of calf. Allosteric regulation of 5‐HT2‐receptors. Naunyn‐Schmied. Arch. Pharmacol. 328:295‐300.
   Kenakin, T.P. 1997. Pharmacologic Analysis of Drug‐Receptor Interaction, 3rd ed. Lippincott‐Raven, Philadelphia, Pa.
   Koshland, D.E., Némethy, G., and Filmer, D. 1966. Comparison of experimental binding data and theoretical models in proteins containing subunits. Biochemistry 5:365‐385.
   Lanzafame, A., Christopoulos, A., and Mitchelson, F. 1996. Interactions of agonists with an allosteric antagonist at muscarinic acetylcholine M2 receptors. Eur. J. Pharmacol. 316:27‐32.
   Lanzafame, A.A., Sexton, P.M., and Christopoulos, A. 2006. Interaction studies of multiple binding sites on m4 muscarinic acetylcholine receptors. Mol. Pharmacol. 70:736‐746.
   Lazareno, S. and Birdsall, N.J.M. 1995. Detection, quantitation, and verification of allosteric interactions of agents with labeled and unlabeled ligands at G protein‐coupled receptors: Interactions of strychnine and acetylcholine at muscarinic receptors. Mol. Pharmacol. 48:362‐378.
   Lazareno, S., Gharagozloo, P., Kuonen, D., Popham, A., and Birdsall, N.J.M. 1998. Subtype‐selective positive cooperative interactions between brucine analogues and acetylcholine at muscarinic receptors: Radioligand binding studies. Mol. Pharmacol. 53:573‐589.
   Lee, N.H. and El‐Fakahany, E.E. 1991. Allosteric antagonists of the muscarinic acetylcholine receptor. Biochem. Pharmacol. 42:199‐205.
   Leppik, R.A., Lazareno, S., Mynett, A., and Birdsall, N.J. 1998. Characterization of the allosteric interactions between antagonists and amiloride analogues at the human a2A‐adrenergic receptor. Mol. Pharmacol. 53:916‐925.
   Limbird, L.E. 1996. Cell Surface Receptors: A Short Course on Theory and Methods, 2nd ed. Kluwer Academic Publishers, Boston.
   Maillet, E.L., Pellegrini, N., Valant, C., Bucher, B., Hibert, M., Bourguignon, J.J., and Galzi, J.L. 2007. A novel, conformation‐specific allosteric inhibitor of the tachykinin NK2 receptor (NK2R) with functionally selective properties. FASEB J. 21:2124‐2134.
   Marino, M.J. and Conn, P.J. 2006. Glutamate‐based therapeutic approaches: allosteric modulators of metabotropic glutamate receptors. Curr. Opin. Pharmacol. 6:98‐102.
   May, L.T., Leach, K., Sexton, P.M., and Christopoulos, A. 2007. Allosteric modulation of G protein‐coupled receptors. Annu. Rev. Pharmacol. Toxicol. 47:1‐51.
   Monod, J., Changeux, J.‐P., and Jacob, F. 1963. Allosteric proteins and cellular control systems. J. Mol. Biol. 6:306‐329.
   Monod, J., Wyman, J., and Changeux, J.‐P. 1965. On the nature of allosteric transitions: A plausible model. J. Mol. Biol. 12:88‐118.
   Price, M.R., Baillie, G.L., Thomas, A., Stevenson, L.A., Easson, M., Goodwin, R., McLean, A., McIntosh, L., Goodwin, G., Walker, G., Westwood, P., Marrs, J., Thomson, F., Cowley, P., Christopoulos, A., Pertwee, R.G., and Ross, R.A. 2005. Allosteric modulation of the cannabinoid CB1 receptor. Mol. Pharmacol. 68:1484‐1495.
   Schetz, J.A. and Sibley, D.R. 1997. Zinc allosterically modulates antagonist binding to cloned D1 and D2 dopamine recpetors. J. Neurochem. 68:1990‐1997.
   Thomas, E.A., Carson, M.J., Neal, M.J., and Sutcliffe, J.G. 1997. Unique allosteric regulation of 5‐hydroxytryptamine receptor‐mediated signal transduction by oleamide. Proc. Natl. Acad. Sci. U.S.A. 94:14115‐14119.
   Thron, C.D. 1973. On the analysis of pharmacological experiments in terms of an allosteric receptor model. Mol. Pharmacol. 9:1‐9.
   Tucek, S. and Proska, J. 1995. Allosteric modulation of muscarinic acetylcholine receptors. Trends Pharmacol. Sci. 16:205‐212.
   Watson, C., Jenkinson, S., Kazmierski, W., and Kenakin, T. 2005. The CCR5 receptor‐based mechanism of action of 873140, a potent allosteric noncompetitive HIV entry inhibitor. Mol. Pharmacol. 67:1268‐1282.
   Wregget, K.A. and Wells, J.W. 1995. Cooperativity manifest in the binding properties of purified cardiac muscarinic receptors. J. Biol. Chem. 270:22488‐22499.
Key References
   Ehlert, F.J. 1988. See above.
   These citations cover the detection and quantification of allosteric interactions at GPCRs in great detail.
   Lazareno, S. and Birdsall, N.J.M. 1995. See above.
GO TO THE FULL PROTOCOL:
PDF or HTML at Wiley Online Library
 
提问
扫一扫
丁香实验小程序二维码
实验小助手
丁香实验公众号二维码
关注公众号
反馈
TOP
打开小程序