Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2008 Aug 4;105(31):10978–10983. doi: 10.1073/pnas.0800567105

Allosteric modulation of the muscarinic M4 receptor as an approach to treating schizophrenia

W Y Chan *, D L McKinzie , S Bose *, S N Mitchell *, J M Witkin , R C Thompson , A Christopoulos §, S Lazareno , N J M Birdsall , F P Bymaster , C C Felder *,†,**
PMCID: PMC2495016  PMID: 18678919

Abstract

Current antipsychotics provide symptomatic relief for patients suffering from schizophrenia and related psychoses; however, their effectiveness is variable and many patients discontinue treatment due to side effects. Although the etiology of schizophrenia is still unclear, a leading hypothesis implicates an imbalanced dopaminergic system. Muscarinic acetylcholine (ACh) receptors regulate dopamine levels in key areas of the brain involved in psychosis, with the M4 subtype emerging as a key regulator of dopaminergic hyperactivity. Unfortunately, no selective small molecule tools exist to provide pharmacological validation of this hypothesis. Here, we describe the discovery of a small molecule modulator, LY2033298, that is highly selective for human M4 receptors by virtue of targeting an allosteric site on this receptor. Pharmacological assays confirmed the selectivity of LY2033298 for the M4 receptor and revealed the highest degree of positive allosteric enhancement of ACh potency thus far identified. Radioligand binding assays also show this compound to directly potentiate agonist binding while having minimal effects on antagonist binding. Mutational analysis identified a key amino acid (D432) in the third extracellular loop of the human M4 receptor to be critical for selectivity and agonist potentiation by LY2033298. Importantly, LY2033298 was active in animal models predictive of clinical antipsychotic drug efficacy indicating its potential use as a first-in-class, selective, allosteric muscarinic antipsychotic agent.

Keywords: cholinergic, GPCR, cooperativity


Schizophrenia is a complex disease presenting a broad spectrum of endophenotypes that can be generalized to symptoms in three major domains: positive (hallucinations, hearing of voices, delusions and disorganized thinking), negative (anhedonia, flat affect), and cognitive (attention and working memory deficits). The etiological basis for the disease is believed to derive predominantly from dysregulation of dopamine and glutamate neurotransmission pathways in mesocortical and mesolimbic brain areas (13). Atypical antipsychotics are the current frontline treatment for schizophrenia, ameliorating the positive symptoms in approximately half of the treated patient population with little efficacy at the negative and cognitive symptoms. Most atypical antipsychotics are broad spectrum G protein-coupled receptor (GPCR) antagonists, and their therapeutic action is mediated primarily through inhibition of dopamine D2, D3 and D4, and serotonin 5HT2A, receptors (4). Their complex pharmacology, however, leads to significant undesirable side effects, including movement disorders and weight gain. Therefore, developing antipsychotics through an alternative mechanism may provide better total symptom control and reduced side effects.

The family of five muscarinic acetylcholine (ACh) receptors plays a prominent role in regulating neurotransmission in the CNS and may provide a mechanism for antipsychotic drug discovery. Studies using muscarinic receptor knock out mice have provided valuable insight into the potential role of these receptors, especially the muscarinic receptor M1 and M4 subtypes, in the pathophysiology of schizophrenia (5, 6). Muscarinic receptors are genetically linked to schizophrenia and cognitive deficits (7, 8). Pharmacologically, relatively non-selective muscarinic antagonists exacerbate, whereas agonists ameliorate, cognitive deficits and psychotic behaviors in animal models and in patients suffering from Alzheimer's disease and schizophrenia (912). In particular, the M1/M4 preferring partial agonist, xanomeline, was found to be efficacious in animal models predictive of antipsychotic behaviors through modulation of both dopamine and glutamate transmission (13). In exploratory clinical trials, xanomeline reduced psychotic behaviors and improved cognitive measures in Alzheimer's patients (9), and significantly improved positive, negative, and cognitive symptoms in a small schizophrenia trial (12). Unfortunately, lack of receptor subtype selectivity led to undesirable side-effects (gastrointestinal disturbances, blood pressure dysregulation) that rendered xanomeline as an unsuitable candidate for further clinical development (14).

M4 receptors are most highly expressed in brain regions rich in dopamine and dopamine receptors (15, 16). Experiments with muscarinic knock out mice have implicated the M4 receptor subtype in regulating dopaminergic neurons involved in movement control and cognition (17, 18); therefore, we reasoned that a highly selective M4 muscarinic receptor activator may represent a useful candidate for the development of muscarinic-based antipsychotic agents. Unfortunately, attempts at discovering selective small molecule ligands for the five muscarinic receptor subtypes have largely been unsuccessful when targeted at the receptors' orthosteric site, i.e., the binding site for the endogenous agonist ACh, which is highly conserved across all five muscarinic subtypes (19). This has been particularly true for the ACh-mimetics, such as xanomeline, sabcomeline, and melameline (20). However, it is also known that muscarinic receptors possess allosteric binding sites that have the potential to show greater sequence divergence across subtypes and, hence, may provide an alternative means of attaining receptor subtype selectivity (21, 22). Allosteric modulators also offer the additional potential of maintaining both spatial and temporal neurotransmission through modulation of physiologically relevant receptor-mediated neural regulation, as opposed to direct (continuous) agonism or antagonism (23, 24). Here, we describe the exploitation of this concept to the discovery and molecular characterization of LY2033298 [3-amino-5-chloro-6-methoxy-4-methyl-thieno(2,3-b)pyridine-2-carboxylic acid cyclopropylamide (C13H14ClN3O2S; molecular weight = 311.8)] (Fig. 1A), a highly selective positive allosteric potentiator of ACh actions at the M4 muscarinic receptor that also shows significant efficacy in preclinical models predictive of antipsychotic drug behavior.

Fig. 1.

Fig. 1.

LY2033298 enhances ACh responses selectively at hM4. The effect and selectivity of LY2033298 on ACh were tested by FLIPR (A) and [35S]GTPγS (B) binding, using recombinant cell lines (AV12 Gα15 hM2, or hM4 and CHO hM1, hM3, or hM5) and CHO cell membranes of hM1–M5, respectively. Responses in the presence of LY2033298 were normalized to the control maximal ACh response (100%) and basal response (B; 0%) for each receptor. In both assays, no significant allosteric effect was observed in hM1, hM3, or hM5 cell lines. A very modest allosteric effect on hM2 receptors was observed and a small agonist effect was detected at hM4. (A Inset) Illustrated is the paradigm of functional screening for the concentration-dependent effect of LY2033298 on a submaximal dose of ACh (3 nM), which was obtained by plotting the data points along the dotted line.

Results

LY2033298 Is a Selective Positive Allosteric Modulator of ACh at Human Muscarinic M4 Receptors.

Initial screening quantified intracellular calcium mobilization as a readout of muscarinic receptor activation, using cell lines stably expressing the hM1–M5 receptors coupled to intrinsic or engineered calcium signaling pathways. No agonist activity at any muscarinic receptor subtype was detected when LY2033298 was applied alone. However, upon addition of a submaximal concentration of ACh, an elevated calcium response was readily detected in the hM4 cell lines relative to the effect of the same concentration of ACh in the absence of LY2033298. Subsequent assays constructed complete concentration-response curves to ACh in the presence of increasing concentrations of LY2033298. As shown in Fig. 1A, these experiments revealed a prototypical characteristic of positive allosteric interactions: a robust potentiation of agonist potency that reached a limit at the highest concentrations of allosteric modulator above which no further effect was obtained (24). Application of an allosteric ternary complex model to the data yielded KB = 200 ± 40 nM for LY2033298 at the allosteric site on the unoccupied hM4 receptor, and α = 35 ± 4 for the degree of allosteric enhancement when both orthosteric and allosteric sites are occupied; to our knowledge, this is the highest degree of positive cooperativity reported for any allosteric modulator of a muscarinic receptor. In addition, these experiments also revealed the high selectivity of LY2033298 for the hM4 receptor, because there was no effect at hM1/3/5 receptors at any concentration of modulator and a very small allosteric potentiation at the closest homolog, hM2 (KB = 1.0 ± 0.3 μM; α = 3.7 ± 0.5). The positive allosteric effect of LY2033298 at the hM4 receptor was not unique to ACh, because it also selectively potentiated other full and partial orthosteric agonists, such as carbachol, oxotremorine-M, and McN-A343 (data not shown).

To confirm that the allosteric effect detected in the calcium assay was neither unique to this signaling pathway or a consequence of engineering the coupling of the hM4 receptor to this pathway via the use of promiscuous Gα15 proteins, additional experiments were performed measuring ACh-mediated [35S]guanosine-γ-S–triphosphates (GTPγS) binding to native G proteins in CHO cell membranes (Fig. 1B). As with the calcium assay, selectivity for the hM4 receptor was observed, with no quantifiable effect at the hM2 receptor. Interestingly, a small increase in basal [35S]GTPγS binding was also observed when LY2033298 was applied alone to both hM4 and hM2 receptors, indicating a weak degree of allosteric agonism in addition to any allosteric modulation. However, poor solubility of LY2033298 at high concentrations limited the analysis and characterization of this allosteric agonist effect in this assay. Nonetheless, because the agonist effect of LY2033298 was small, we were still able to apply the simple allosteric ternary complex model to derive apparent estimates of modulator affinity (KB = 870 ± 310 nM) and cooperativity (α = 28 ± 7) at the hM4 receptor.

LY2033298 Directly Enhances the Binding of Agonists but Not the Antagonist N-methyl-scopolamine (NMS).

Radioligand binding assays were performed by using the orthosteric agonist [3H]Oxotremorine-M (Oxo-M) and antagonist [3H]NMS to confirm that the allosteric effects of LY2033298 are predominantly manifested directly at the level of orthosteric ligand affinity. Fig. 2A shows that LY2033298 robustly potentiated the specific binding of [3H]Oxo-M in CHO hM4 cell membranes and in rat striatal membranes known to express a high proportion of M4 muscarinic receptor (F.P.B. and C.C.F., unpublished data). Interestingly, although a significant potentiation of [3H]Oxo-M binding was retained in native rat tissue, the potency of LY2033298 at the rat M4 receptor was 5- to 6-fold lower than that at the human M4 receptor, suggesting a possible species difference in the allosteric effect.

Fig. 2.

Fig. 2.

LY2033298 allosterically increases agonist binding to M4 receptors. (A) The specific binding of [3H]Oxo-M to CHO hM4 cell membranes and rat striatal membranes was potentiated by LY2033298 in a dose-dependent manner. (B) The specific binding of [3H]NMS to hM4 receptors in the presence of different concentrations of unlabeled ACh and LY2033298 was measured in the presence of 0.2 mM GTP. The incubation time was 2 h to allow equilibrium to be reached. The low slope factor of the ACh inhibition curve (0.67) was unchanged in the presence of increasing concentrations of LY2033298. The presence of LY2033298 increased ACh potency up to 40-fold. At high concentrations, LY2033298 appears to weakly and negatively modulate [3H]NMS binding at hM4 receptors. The data are from one experiment repeated three times with quantitatively similar results.

To assess the effect of LY2033298 on receptor affinity for the endogenous agonist at hM4 receptors, ACh inhibition of [3H]NMS binding was measured at equilibrium in the presence of 0.2 mM GTP and with increasing concentrations of LY2033298 (Fig. 2B). The inhibition curve in the absence of LY2033298 had a logistic slope factor of 0.67 and, with increasing concentrations of LY2033298, the curves were progressively shifted leftward without change in slope factor. The increase in ACh potency was 40-fold at 3 μM LY2033298, illustrating the very strong positive cooperativity between LY2033298 with ACh for binding to uncoupled hM4 receptors. In contrast, under these incubation conditions, the binding of [3H]NMS was slightly reduced at higher concentrations of LY2033298, compatible with a small negative cooperativity with [3H]NMS (α ≅ 0.5). These assays demonstrated another characteristic of the allosteric interactions of LY2033298, probe-dependency (24). No appreciable radioligand displacement by LY2033298 was detected in orthosteric binding assays for other GPCRs (e.g., dopamine, 5HT, adrenergic receptor families), major ion channels (e.g., nicotinic), signaling molecules, and enzymes (e.g., kinase family) (data not shown). Moreover, LY2033298 enhancement of [3H]Oxo-M binding was lost in M4 knockout and not in M2 knockout or wild-type mouse membranes supporting the selectivity for M4 over the M2 receptor (data not shown).

Molecular Determinants of LY2033298 Selectivity and Allosterism at the M4 Receptor.

The orthosteric ligand binding pocket is highly conserved between muscarinic receptor subtypes and across species (19). Among these receptors, hM2 and hM4 share the closest sequence homology; however, we still observed significant differences in LY2033298 selectivity. Thus, we created a series of hM4 receptor mutants [supporting information (SI) Table S1], in which divergent residues in the extracellular N terminus and loop regions were replaced with those of hM2 receptor at the corresponding sites, and measured ACh-stimulated functional responses. All mutants showed unaltered pharmacology to orthosteric agonists and antagonists compared with control wild-type hM4 receptors (Fig. S1). The most dramatic loss of LY2033298 activity was noted upon progressive mutation of the third extracellular loop (o3) (Fig. 3A). LY2033298-mediated potentiation, both in terms of its potency and magnitude of potentiation, was particularly reduced in mutant hM4-Δc6.1, and a little further in hM4-Δc7.1, indicating residue D432 is critical for this activity. Subsequent point mutations confirmed that the size and charge of this residue are vital to the potency of LY2033298 (Fig. S2).

Fig. 3.

Fig. 3.

Sequential mutations in the o3 loop of muscarinic receptors reveal a critical residue (D432) for LY2033298-mediated potentiation. Receptor mutants were transiently expressed in AV12 Gα15 cells. The effect of LY2033298 on ACh stimulated calcium mobilization was tested upon coapplication of LY2033298 (1 nM to 1 μM) with the EC25 concentration of ACh for each receptor. The results were normalized to the potentiation mediated by LY2033298 on hM4 receptors, where 0% and 100% corresponded to zero and maximum potentiation mediated by LY2033298 on hM4 receptors (see Fig. S4). (A) Mutants hM4-Δ6.1 and hM4-Δ7.1 were significantly less responsive to LY2033298 (*) when compared with hM4 receptors. The data points represents mean ± S.E.M. of more than eight experiments. (B) The enhancement of ACh function by LY2033298 for the humanized rM4 was not significantly different from rM4 receptors, whereas ratinized hM4 was significantly different from hM4 but not from rM4 receptors. The receptor selectivity of LY2033298-mediated potentiation between rM4 and rM2 receptors was sustained. Data shown are mean ± SEM (n ≥ 3).

Comparison of rat and human M4 receptor sequences at this domain also identified two nonconserved residues in the o3 loop, one of which corresponded to D432 in the hM4 receptor. In agreement with the results of the [3H]Oxo-M binding assays, the rM4 receptor demonstrated a decreased potency of LY2033298 and a reduced potentiation of ACh-mediated calcium mobilization compared with the hM4 receptor, although the higher selectivity of LY2033298 for the M4 subtype relative to the M2 subtype was retained (Fig. 3A). When the two divergent residues within the third extracellular loop of the hM4 receptor were converted to their rM4 counterparts (“ratinized hM4”), there was a significant loss LY2033298 activity, albeit not to the degree observed at the wild-type rM4. However, when the reciprocal mutations were made in the rM4 receptor (“humanized rM4”), there was no gain of function (Fig. 3B). This discrepancy suggests that the species difference observed likely involves a complex interplay between the o3 loop and other domains/residues on the rM4 receptor.

LY2033298 Displays in Vivo Efficacy in Preclinical Animal Models Predictive of Antispsychotic Drug Effects.

When LY2033298 was administered alone to rats in conditioned avoidance responding (CAR), and prepulse inhibition (PPI) models and when performing microdialysis sampling of brain mono-amines, we did not observe any effect- consistent with lower activity of LY2033298 at the rodent vs. human M4 receptor. However, when coadministered with a subeffective single dose of oxotremorine, LY2033298 was active in attenuation of CAR and reversal of apomorphine-disrupted PPI in a dose-dependent manner (Fig. 4 A and B), indicating that the compound is indeed effective in vivo through a muscarinic mechanism. Similar results were observed in microdialysis experiments where LY2033298 positively modulated the dopaminergic system in the prefrontal cortex in the presence of an inactive dose of oxotremorine (Fig. S3), suggesting it had reached and affected the desired target pathway. The effectiveness of LY2033298 in rodent models predictive of antipsychotic efficacy provides compelling proof-of-concept that allosteric potentiation of the M4 muscarinic receptor is a viable approach toward the development of muscarinic-based antipsychotic agents. Because of its significantly higher activity at the human receptor, this and related compounds may have efficacy when given alone in human subjects.

Fig. 4.

Fig. 4.

LY2033298-mediated potentiation is effective in rat CAR and PPI psychosis models. (A) LY2033298 reduced conditioned avoidance responding (CAR) in trained male HSD rats in a dose-dependent manner in the presence of an inactive dose of oxotremorine (*, P < 0.05 versus vehicle). (B) Apomorphine-induced suppression of the acoustic startle response was reversed by LY2033298 in a dose-dependent manner in the presence of a subeffective dose of Oxo. Data are mean percentage prepulse inhibition (PPI) values (V, vehicle; Apo, apomorphine; Oxo, oxotremorine; LY, 2033298) (#, P < 0.05 versus V/V/Apo; *, P < 0.05 versus V/V/V).

Discussion

This study has identified a functionally potent and selective allosteric potentiator of muscarinic receptors. The characteristics of this allosteric modulator LY2033298 include: (i) selectivity for modulating ACh actions at the M4 receptor, attributable to epitopes on the receptor topographically distinct from the highly conserved orthosteric site; (ii) saturability in the enhancement of ACh potency as a direct consequence of cooperativity between orthosteric and allosteric sites; (iii) probe dependence in the allosteric effect, because LY2033298 selectively potentiates the actions of orthosteric agonists at M4 receptors but interacts very weakly with the orthosteric antagonist, [3H]NMS; and (iv) signal transduction pathway independence, because the effects were observed in [35S]GTPγS binding and calcium mobilization assays.

Xanomeline is pharmacologically characterized as an M1/M4 > M2/M3 > M5 partial agonist with modest interaction at serotonin receptors (6, 25). The clinical efficacy detailed in the Introduction raises the question as to which muscarinic receptor(s) to focus on for the indication of schizophrenia. In the absence of selective pharmacology, muscarinic receptor knockout mice have provided evidence encouraging a focus on the M4 receptor as a logical target for modulation of dopamine neurotransmission (17) in areas thought to be disrupted in schizophrenia (18). BuTAC and PTAC, potent molecules with M2/M4 agonist, M1/M3/M5 antagonist activity, were active in models predictive of antipsychotic efficacy (26, 27). Together with our data on LY2033298, these findings support a role for M4 as potentially sufficient to treat symptoms of schizophrenia. Because M1 receptor involvement in cognitive processes has been well documented (28, 29), a combination of M1 and M4 agonistic activity may offer some additional efficacy in the clinic.

Previous investigations identified subtype-selective muscarinic allosteric enhancers (23, 3032). However, these molecules displayed much lower affinity and cooperativity with ACh. A particularly striking finding from our functional assays was the high degree of positive cooperativity between the modulator and ACh at the hM4 receptor (α≈30), which is the highest for muscarinic receptors reported to date and is sufficient to impart behavioral responses in animal models predictive of antipsychotic drug effects at reasonable therapeutic doses. Radioligand binding experiments using recombinant hM4 and native rM4 membrane preparations confirmed that the allosteric effects of LY2033298 are mediated predominantly by enhancement of agonist binding while having close to neutral effects on antagonist binding. [35S]GTPγS binding experiments indicate a low intrinsic efficacy of LY2033298 at the hM2 and hM4 receptors in the absence of added ACh consistent with an allosteric partial agonist of very low efficacy (33, 34).

To date, limited mutagenesis studies of muscarinic receptors have focused on a common allosteric binding site on M1, M2, and M5 receptor subtypes (33, 35) and M3 (36, 37) and M4 receptors (38) used by prototypical negative allosteric modulators, such as gallamine, alcuronium, and C7/3-phth (21). Functional screening of a wide range of receptor mutants systematically pointed to the acidic residue D432 of the hM4 receptor as being critical to the potentiation of ACh by LY2033298 (Fig. 3A). Additional studies on the rM4 receptor, which shows a lower cooperativity with LY2033298, confirmed the importance of the o3 region of the receptor in the actions of the modulator (Fig. 3B). However, a humanized rM4 mutant did not show a gain-of-function in enhancing the actions of LY2033298, suggesting that a more complex network of interactions with other residues must also be involved. In general, the finding that D432 was critical to the cooperativity between LY2033298 and ACh at the hM4 receptor contrasts with residues important for the action of less robust allosteric modulators, which, for the hM2 receptors, have been localized to the o2 loop and the top of transmembrane domain 7 (38, 39). However, an unequivocal demonstration that LY2033298 interacts with a second allosteric site that is topographically distinct from that used by prototypical muscarinic modulators requires further pharmacological and structural characterization.

A key finding of our study was the demonstration of in vivo efficacy of a muscarinic allosteric potentiator in rodent preclinical models predictive of antipsychotic drug effects. Allosteric sites are not expected to have evolved to accommodate a common endogenous neurotransmitter or hormone; they are more likely to show higher sequence divergence across receptor subtypes (which represents a therapeutic targeting advantage) but may also show variation between species, which represents a disadvantage in terms of target validation and/or preclinical efficacy and safety studies in animal models. Transgenic approaches to knock-in humanized receptor proteins into mice may be helpful for progressing allosteric drug leads showing high human receptor selectivity. Alternatively, allosteric compounds may be coadministered with inactive doses of orthosteric agonists.

Arguably, little innovation has been made in the treatment of schizophrenia for the past several decades. Frontline treatments are generally compounds with complex broad GPCR antagonist pharmacology with highest affinity as antagonists for D2-type dopamine receptors and 5HT2 serotonin receptors (4). GPCR agonist mechanisms have largely failed or, at best, show temporary efficacy because of issues of receptor desensitization or down regulation (40). Aripiprazole may be an exception because of its partial agonist activity at D2 receptors, although the exact mechanism of action of this compound has been controversial (41). More recent preliminary studies suggest antipsychotic efficacy through agonism of type 2 and 3 metabotropic glutamate receptors (42). Muscarinic selective agonism with xanomeline (although not completely muscarinic receptor subtype selective) has shown interesting promise in managing Alzheimer's-related psychosis and schizophrenia symptoms, including positive, negative, and cognitive domains (6, 9). An M4-selective drug that provides modest or self-limiting agonism may be preferable to a full orthosteric agonist, although it is not clear how much receptor agonism (or modulator affinity and cooperativity) will be required in patients to achieve maximal therapeutic efficacy and tolerable side effects. Studies to date indicate that M4 receptors are largely unchanged in schizophrenic patients with the exception of a potential modest decrease in hippocampus, suggesting that M4 agonist-based therapies would remain relevant during disease progression (43, 44). Our rodent studies, although preliminary, provide support for a highly M4 receptor-selective allosteric potentiator to control symptoms without inducing sedation or catalepsy observed with atypical antagonist antipsychotics such as clozapine. The allosteric mechanism inherent in LY2033298 may appropriately modulate imbalanced dopamine and glutamate neurotransmission, thus representing a potential first-in-class muscarinic antipsychotic therapy that does not involve direct targeting of dopamine and serotonin receptors.

Materials and Methods

Reagents.

3-Amino-5-chloro-6-methoxy-4-methyl-thieno[2,3-b]pyridine-2-carboxylic acid cyclopropylamide (LY2033298; C13H14ClN3O2S; molecular weight = 311.8) (Fig. 1A) was synthesized by Eli Lilly & Co.. LY2033298 is soluble in buffer or in 100% DMSO up to 4 mM and is stable as a dry powder at room temperature or as a frozen DMSO stock solution. The human muscarinic receptors were stably expressed in cell lines as described in SI Methods.

Methods.

Signal transduction and radioligand binding assays and data analyses were performed as described in refs. 24, 45, and 46 with modifications as detailed in SI Methods. Rat behavioral assays, conditioned avoidance responding (CAR), and prepulse inhibition of the acoustic startle reflex (PPI) were performed as described in ref. 47 with modifications as detailed in SI Methods. Mutagenesis and receptor expression methods are described in SI Methods.

Supplementary Material

Supporting Information

Acknowledgments.

We thank George Nomikos and Angela Popham for their key contributions to this manuscript. A.C. is a Senior Research Fellow of the National Health and Medical Research Council of Australia. This work was supported by Eli Lilly & Co. and the Medical Research Council, United Kingdom (N.J.M.B. and S.L.).

Footnotes

Conflict of interest statement: W.Y.C., D.L.M., S.B., S.N.M., J.M.W., R.C.T., F.P.B., and C.C.F. are or were employed by Eli Lilly & Co.

This article is a PNAS Direct Submission.

This article contains supporting information online at www.pnas.org/cgi/content/full/0800567105/DCSupplemental.

References

  • 1.Jablensky A. Subtyping schizophrenia: Implications for genetic research. Mol Psychiatry. 2006;11:815–836. doi: 10.1038/sj.mp.4001857. [DOI] [PubMed] [Google Scholar]
  • 2.Lindsley CW, et al. Progress towards validating the NMDA receptor hypofunction hypothesis of schizophrenia. Curr Top Med Chem. 2006;6(8):771–785. doi: 10.2174/156802606777057599. [DOI] [PubMed] [Google Scholar]
  • 3.Tamminga CA. The neurobiology of cognition in schizophrenia. J Clin Psychiatry. 2006;67(Suppl. 9):9–13. [PubMed] [Google Scholar]
  • 4.Roth BL, Sheffler DJ, Kroeze WK. Magic shotguns versus magic bullets: Selectively non-selective drugs for mood disorders and schizophrenia. Nat Rev Drug Discov. 2004;3:353–359. doi: 10.1038/nrd1346. [DOI] [PubMed] [Google Scholar]
  • 5.Felder CC, Bymaster FP, Ward J, DeLapp N. Therapeutic opportunities for muscarinic receptors in the central nervous system. J Med Chem. 2000;43(23):4333–4353. doi: 10.1021/jm990607u. [DOI] [PubMed] [Google Scholar]
  • 6.Raedler TJ, Bymaster FP, Tandon R, Copolov D, Dean B. Towards a muscarinic hypothesis of schizophrenia. Mol Psychiatry. 2007;12:232–246. doi: 10.1038/sj.mp.4001924. [DOI] [PubMed] [Google Scholar]
  • 7.De Luca V, et al. Linkage of M5 muscarinic and alpha7-nicotinic receptor genes on 15q13 to schizophrenia. Neuropsychobiology. 2004;50:124–127. doi: 10.1159/000079102. [DOI] [PubMed] [Google Scholar]
  • 8.Liao D-L, et al. Association of muscarinic M1 receptor genetic polymorphisms with psychiatric symptoms and cognitive function in schizophrenic patients. Neuropsychobiology. 2003;48:72–76. doi: 10.1159/000072880. [DOI] [PubMed] [Google Scholar]
  • 9.Bodick NC, et al. Effects of xanomeline, a selective muscarinic receptor agonist, on cognitive function and behavioral symptoms in Alzheimer disease. Arch Neurol. 1997;54:465–473. doi: 10.1001/archneur.1997.00550160091022. [DOI] [PubMed] [Google Scholar]
  • 10.Ellis JR, et al. Muscarinic and nicotinic receptors synergistically modulate working memory and attention in humans. Int J Neuropsychopharmacology. 2006;9:175–189. doi: 10.1017/S1461145705005407. [DOI] [PubMed] [Google Scholar]
  • 11.Minzenberg MJ, Poole JH, Benton C, Vinogradov S. Association of anticholinergic load with impairment of complex attention and memory in schizophrenia. Am J Psychiatry. 2004;161:116–124. doi: 10.1176/appi.ajp.161.1.116. [DOI] [PubMed] [Google Scholar]
  • 12.Mirza NR, Peters D, Sparks RG. Xanomeline and the antipsychotic potential of muscarinic receptor subtype selective agonists. CNS Drug Rev. 2003;9:159–186. doi: 10.1111/j.1527-3458.2003.tb00247.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Raedler TJ, Tandon R. Cholinergic mechanisms in schizophrenia: Current concepts. Curr Psychosis Therapeutics Rep. 2006;4(1):20–26. [Google Scholar]
  • 14.Medina A, Bodick N, Goldberger AL, Mac Mahon M, Lipsitz LA. Effects of central muscarinic-1 receptor stimulation on blood pressure regulation. Hypertension. 1997;29:828–834. doi: 10.1161/01.hyp.29.3.828. [DOI] [PubMed] [Google Scholar]
  • 15.Yasuda RP, et al. Development of antisera selective for m4 and m5 muscarinic cholinergic receptors: Distribution of m4 and m5 receptors in rat brain. Mol Pharmacol. 1993;43:149–157. [PubMed] [Google Scholar]
  • 16.Wirtshafter D, Osborn CV. The distribution of m4 muscarinic acetylcholine receptors in the islands of Calleja and striatum of rats and cynomolgus monkeys. J Chem Neuroanatomy. 28:107–116. doi: 10.1016/j.jchemneu.2004.05.007. and erratum (2005) 29(3):227. [DOI] [PubMed] [Google Scholar]
  • 17.Bymaster FP, McKinzie DL, Felder CC, Wess J. Use of M-1-M-5 muscarinic receptor knockout mice as novel tools to delineate the physiological roles of the muscarinic cholinergic system. Neurochem Res. 2003;28:437–442. doi: 10.1023/a:1022844517200. [DOI] [PubMed] [Google Scholar]
  • 18.Tzavara ET, et al. M4 muscarinic receptors regulate the dynamics of cholinergic and dopaminergic neurotransmission: Relevance to the pathophysiology and treatment of related CNS pathologies. FASEB J. 2004;18:1410–1412. doi: 10.1096/fj.04-1575fje. [DOI] [PubMed] [Google Scholar]
  • 19.Lu ZL, Saldanha JW, Hulme EC. Seven-transmembrane receptors: Crystals clarify. Trends Pharmacol Sci. 2002;23:140–146. doi: 10.1016/S0165-6147(00)01973-8. [DOI] [PubMed] [Google Scholar]
  • 20.Eglen RM. Muscarinic receptor subtype pharmacology and physiology. Progress Med Chem. 2005;43:105–136. doi: 10.1016/S0079-6468(05)43004-0. [DOI] [PubMed] [Google Scholar]
  • 21.Birdsall NJ, Lazareno S. Allosterism at muscarinic receptors: Ligands and mechanisms. Mini Rev Med Chem. 2005;5:523–543. doi: 10.2174/1389557054023251. [DOI] [PubMed] [Google Scholar]
  • 22.Christopoulos A, Lanzafame A, Mitchelson F. Allosteric interactions at muscarinic cholinoceptors. Clin Exp Pharmacol Physiol. 1998;25:185–194. doi: 10.1111/j.1440-1681.1998.t01-4-.x. [DOI] [PubMed] [Google Scholar]
  • 23.Birdsall NJ, et al. Subtype-selective positive cooperative interactions between brucine analogs and acetylcholine at muscarinic receptors: Functional studies. Mol Pharmacol. 1999;55:778–786. [PubMed] [Google Scholar]
  • 24.Christopoulos A, Kenakin T. G protein-coupled receptor allosterism and complexing. Pharmacol Rev. 2002;54:323–374. doi: 10.1124/pr.54.2.323. [DOI] [PubMed] [Google Scholar]
  • 25.Watson J, et al. Functional effects of the muscarinic receptor agonist, xanomeline, at 5-HT1 and 5-HT2 receptors. Br J Pharmacol. 1998;125:1413–1420. doi: 10.1038/sj.bjp.0702201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rasmussen T, et al. The muscarinic receptor agonist BuTAC, a novel potential antipsychotic, does not impair learning and memory in mouse passive avoidance. Schizophrenia Res. 2001;49:193–201. doi: 10.1016/s0920-9964(00)00129-8. [DOI] [PubMed] [Google Scholar]
  • 27.Bymaster FP, et al. Unexpected antipsychotic-like activity with the muscarinic receptor ligand (5R,6R)6-(3-propylthio-1,2,5-thiadiazol-4-yl)-1-azabicyclo[3.2.1]octane. Eur J Pharmacol. 1998;356:109–119. doi: 10.1016/s0014-2999(98)00487-7. [DOI] [PubMed] [Google Scholar]
  • 28.Ferreira AR, et al. Role of hippocampal M1 and M4 muscarinic receptor subtypes in memory consolidation in the rat. Pharmacol Biochem Behav. 2003;74:411–415. doi: 10.1016/s0091-3057(02)01007-9. [DOI] [PubMed] [Google Scholar]
  • 29.McKinney M. Brain cholinergic vulnerability: Relevance to behavior and disease. Biochem Pharmacol. 2005;70(8):1115–1124. doi: 10.1016/j.bcp.2005.05.019. [DOI] [PubMed] [Google Scholar]
  • 30.Lazareno S, et al. Allosteric effects of four stereoisomers of a fused indole ring system with 3H-N-methylscopolamine and acetylcholine at M1–M4 muscarinic receptors. Life Sci. 1999;64:519–526. doi: 10.1016/s0024-3205(98)00596-7. [DOI] [PubMed] [Google Scholar]
  • 31.Lazareno S, Dolezal V, Popham A, Birdsall NJ. Thiochrome enhances acetylcholine affinity at muscarinic M4 receptors: Receptor subtype selectivity via cooperativity rather than affinity. Mol Pharmacol. 2004;65:257–266. doi: 10.1124/mol.65.1.257. [DOI] [PubMed] [Google Scholar]
  • 32.Jakubik J, Bacakova L, El-Fakahany EE, Tucek S. Positive cooperativity of acetylcholine and other agonists with allosteric ligands on muscarinic acetylcholine receptors. Mol Pharmacol. 1997;52:172–179. doi: 10.1124/mol.52.1.172. [DOI] [PubMed] [Google Scholar]
  • 33.May LT, Leach K, Sexton PM, Christopoulos A. Allosteric modulation of G protein-coupled receptors. Annu Rev Pharmacol Toxicol. 2007;47:1–51. doi: 10.1146/annurev.pharmtox.47.120505.105159. [DOI] [PubMed] [Google Scholar]
  • 34.Leach K, Sexton PM, Christopoulos A. Allosteric GPCR modulators: Taking advantage of permissive receptor pharmacology. Trends Pharmacol Sci. 2007;28:382–389. doi: 10.1016/j.tips.2007.06.004. [DOI] [PubMed] [Google Scholar]
  • 35.Wess J. Allosteric binding sites on muscarinic acetylcholine receptors. Mol Pharmacol. 2005;68:1506–1509. doi: 10.1124/mol.105.019141. [DOI] [PubMed] [Google Scholar]
  • 36.Jakubik J, Krejci A, Dolezal V. Asparagine, valine, and threonine in the third extracellular loop of muscarinic receptor have essential roles in the positive cooperativity of strychnine-like allosteric modulators. J Pharmacol Exp Ther. 2005;313:688–696. doi: 10.1124/jpet.104.080358. [DOI] [PubMed] [Google Scholar]
  • 37.Krejci A, Tucek S. Changes of cooperativity between N-methylscopolamine and allosteric modulators alcuronium and gallamine induced by mutations of external loops of muscarinic M(3) receptors. Mol Pharmacol. 2001;60:761–767. [PubMed] [Google Scholar]
  • 38.Gnagey AL, Seidenberg M, Ellis J. Site-directed mutagenesis reveals two epitopes involved in the subtype selectivity of the allosteric interactions of gallamine at muscarinic acetylcholine receptors. Mol Pharmacol. 1999;56:1245–1253. doi: 10.1124/mol.56.6.1245. [DOI] [PubMed] [Google Scholar]
  • 39.Huang XP, Prilla S, Mohr K, Ellis J. 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. Mol Pharmacol. 2005;68:769–778. doi: 10.1124/mol.105.014043. [DOI] [PubMed] [Google Scholar]
  • 40.Sawa A, Snyder SH. Schizophrenia: Diverse approaches to a complex disease. Science. 2002;296:692–695. doi: 10.1126/science.1070532. [DOI] [PubMed] [Google Scholar]
  • 41.DeLeon A, Patel NC, Crismon ML. Aripiprazole: A comprehensive review of its pharmacology, clinical efficacy, and tolerability. Clin Ther. 2004;26:649–666. doi: 10.1016/s0149-2918(04)90066-5. [DOI] [PubMed] [Google Scholar]
  • 42.Patil ST, et al. Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: A randomized Phase 2 clinical trial. Nat Med. 2007;13:1102–1107. doi: 10.1038/nm1632. [DOI] [PubMed] [Google Scholar]
  • 43.Matsumoto I, Inoue Y, Iwazaki T, Pavey G, Dean B. 5-HT2A and muscarinic receptors in schizophrenia: A postmortem study. Neurosci Lett. 2005;379(3):64–168. doi: 10.1016/j.neulet.2004.12.059. [DOI] [PubMed] [Google Scholar]
  • 44.Scarr E, Sundram S, Keriakous D, Dean B. Altered hippocampal muscarinic M4, but not M1, receptor expression from subjects with schizophrenia. Biol Psychiatry. 2007;61:1161–1170. doi: 10.1016/j.biopsych.2006.08.050. [DOI] [PubMed] [Google Scholar]
  • 45.Lazareno S, Birdsall NJ. 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. 1995;48:362–378. [PubMed] [Google Scholar]
  • 46.DeLapp NW, et al. Determination of [S-35]guanosine-5′-O-(3-thio)triphosphate binding mediated by cholinergic muscarinic receptors in membranes from Chinese hamster ovary cells and rat striatum using an anti-G protein scintillation proximity assay. J Pharmacol Exp Ther. 1999;289:946–955. [PubMed] [Google Scholar]
  • 47.Rorick-Kehn LM, et al. In vivo pharmacological characterization of the structurally novel, potent, selective mGlu2/3 receptor agonist LY404039 in animal models of psychiatric disorders. Psychopharmacology. 2007;193:121–136. doi: 10.1007/s00213-007-0758-3. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting Information

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES