Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 1.
Published in final edited form as: Discov Med. 2012 Dec;14(79):413–420.

Emerging Approaches for Treatment of Schizophrenia: Modulation of Cholinergic Signaling

Daniel J Foster 1, Carrie K Jones 2, P Jeffrey Conn 3
PMCID: PMC3726271  NIHMSID: NIHMS488817  PMID: 23272693

Abstract

Currently available therapeutic agents for treatment of schizophrenia target signaling by monoaminergic neurotransmitters; however, these treatments do not adequately treat the range of symptoms observed in patients. While these therapies treat the positive symptoms, they do not have efficacy in treating the negative symptoms and cognitive deficits that are associated with the disease. Evidence suggests that molecules that modulate signaling by the neurotransmitter acetylcholine (ACh) could provide a more comprehensive treatment of schizophrenia than currently prescribed antipsychotics. Molecules that broadly increase ACh-signaling have been demonstrated to have efficacy in treating numerous symptom clusters in schizophrenia patients. Unfortunately, these compounds induce adverse effects via activation of peripheral receptors that limit their clinical utility. One proposed strategy for retaining the efficacy of cholinergic treatments, without the adverse effects, is to target specific cholinergic receptor subtypes in the brain. Several cholinergic receptors are able to modulate brain circuits that are dysregulated in schizophrenia patients including receptors belonging to both the muscarinic family (i.e., M1, M4, and M5), and the nicotinic family (i.e., α7, α4β2). Recently, great strides have been made in developing small molecules with high specificity for these receptors, and several of these novel molecules have robust efficacy in several preclinical models predictive of both anti-psychotic and pro-cognitive effectiveness. Promising studies suggest that targeting M1 and α7 may be beneficial for pro-cognitive effects, while molecules that target M4 may be ideally suited to address the positive symptoms. Since these receptor subtypes are distinct from those responsible for the adverse effects observed with non-selective cholinergic treatments, there is hope that molecules targeting these receptors could provide novel therapeutics. Further research is needed to examine the utility of such compounds as therapeutics that could be used either alone, or in combination with existing medications, to better treat schizophrenia.

Introduction

Schizophrenia is a devastating and complex psychiatric illness that affects approximately 1% of the population worldwide. Schizophrenia usually manifests with an onset of symptoms in late adolescence/young adulthood and these symptoms can be grouped into three main categories. Positive symptoms include disordered thoughts/speech, delusions, and hallucinations. Negative symptoms are comprised of blunted affect, anhedonia, and social withdrawal; while cognitive impairments include disturbances in attention, working memory, and executive functions (Lewis and Lieberman, 2000). There is strong evidence that genetics plays a key role in schizophrenia; however, it is becoming increasingly evident that this disorder is polygenic in nature and likely involves the interaction between hundreds of genes and environmental factors (Sullivan et al., 2012). Similar to the genetic nature of schizophrenia, the neurobiology underlying this disease is complex. Post-mortem and neuro-imaging studies have demonstrated altered connectivity and function in numerous brain regions including (but not limited to) the prefrontal cortex, hippocampus, striatum, and thalamus (Ross et al., 2006). Until recently the prevailing hypothesis was that the primary pathology of schizophrenia was a dysregulation of signaling by monoamines such as dopamine and serotonin. However, it is becoming increasingly clear that the disease etiology cannot be explained solely as a result of changes in monoamine signaling. Recent research suggests that altered signaling via the N-methyl-D-aspartate (NMDA) receptor subtype of glutamate receptors, GABAA receptors in the prefrontal cortex, and cholinergic muscarinic (mAChR) and nicotinic (nAChR) receptors may contribute to all three symptom clusters of schizophrenia (Lisman et al., 2008; Marin, 2012; Noetzel et al., 2012; Sarter et al., 2012).

Current therapies for schizophrenia include the use of both typical (e.g., haloperidol) and atypical antipsychotics (e.g., clozapine and risperidone) which both exert their primary effects through D2 dopamine receptor antagonism as well as activity at a host of other monoamine receptors (Sawa and Snyder, 2003). These treatments are at least partially effective in treating the positive symptoms of schizophrenia. However, studies have shown that up to 74% of schizophrenia patients discontinue the use of atypical antipsychotics within 18 months due to adverse effects such as parkinsonian-like motor effects and metabolic syndrome which are tied to their mechanism of action (Lieberman et al., 2005). Even for those patients that do not experience pronounced side-effects, the treatments available do not reduce the cognitive and negative symptoms. Given the shortcomings of current therapies it is imperative that novel approaches are developed that can be used to treat all three symptom clusters associated with schizophrenia with fewer adverse effects.

Targeting Muscarinic Receptors for Treatment of Schizophrenia

Multiple lines of evidence suggest that increasing cholinergic signaling via both muscarinic (mAChR) and nicotinic receptors (nAChR) could be beneficial in schizophrenia. Post-mortem studies have demonstrated a decrease in mAChR expression levels in several brain regions of schizophrenic patients including the pre-frontal cortex, striatum, and hippocampus (Crook et al., 2000; Dean et al., 1996; Zavitsanou et al., 2004). Consistent with this finding, administration of nonselective mAChR antagonists can induce psychotic-like symptoms in healthy human volunteers (Osterholm and Camoriano, 1982), and exacerbate existing symptoms when administered to schizophrenic patients (Tandon et al., 1991). Conversely, increasing cholinergic signaling via administration of acetylcholinesterase (AChE) inhibitors can improve cognitive performance (Feldman et al., 2007; Forette et al., 1999), making cholinergic signaling an attractive target to treat a multitude of disorders associated with cognitive impairments. Unfortunately, most agents that target cholinergic signaling have failed in clinical trials due to dose-limiting effects caused by activation of peripheral receptors.

Most mAChR agonists that have entered clinical testing were originally developed for treatment of Alzheimer’s disease (AD). Of the mAChR agonists, the M1/M4 preferring agonist xanomeline progressed the furthest in the clinical development for treatment of cognitive deficits associated with Alzheimer’s disease. While xanomeline showed a trend towards improving cognitive function in AD patients, the cognition-enhancing effects observed in a phase III trial failed to reach statistical significance. However, a surprising finding from this study was that this M1/M4 preferring agonist had robust therapeutic effects on psychotic symptoms and behavioral disturbances associated with AD. In particular, xanomeline produced robust, dose-dependent reductions in delusions, hallucinations, vocal outbursts, suspiciousness, and agitation while improving blunted affect and other behavioral disturbances (Bodick et al., 1997a; 1997b). More recently, a four-week, double-blind, placebo-controlled outcome trial in subjects with schizophrenia (n=20) was performed to further evaluate the antipsychotic efficacy of xanomeline. This study found robust improvements in both the positive and negative symptoms of schizophrenic patients compared to the placebo group. Both the magnitude and the time-course of xanomeline efficacy were superior to those observed with typical antipsychotics. Therapeutic effects of xanomeline were statistically significant after one week of treatment as opposed to the multi-week delay observed with typical antipsychotics. In terms of treating cognitive impairments, xanomeline produced statistically significant improvements in some measures of cognitive function including verbal learning and short-term memory (Shekhar et al., 2008). Unfortunately, adverse effects, particularly gastrointestinal-related, were observed with xanomeline and dose-limitations have removed it from consideration for long-term clinical use. However, these clinical studies provide strong validation for mAChR agonists as potential therapeutics to treat psychosis and behavioral disturbances in a wide spectrum of diseases including schizophrenia and neurodegenerative diseases.

Allosteric Modulators of Muscarinic Receptors

While results of clinical studies with mAChR agonists such as xanomeline are encouraging, the clinical utility of nonselective muscarinic agonists and AChE inhibitors is limited, primarily due to the dose-limiting side effects including brachycardia, GI distress, excessive salivation, and sweating. The mAChRs are members of the family A class of G-protein coupled receptors and include five subtypes named M1-M5. Evidence suggests that the most prominent adverse effects of AChE inhibitors and other nonselective cholinergic agents are mediated by peripheral M2 and M3 receptors (Bymaster et al., 2003). Accordingly, efforts were made to specifically target M1, M4, or M5 receptors with the goal of retaining the therapeutic efficacy of non-selective cholinergic agents without the adverse side-effects. However, the acetylcholine (ACh) binding site (also called the orthosteric site) is highly conserved across the five subtypes, making it difficult to develop subtype-selective ACh-site ligands. To circumvent this problem, an approach of targeting binding sites that are separate from the ACh-site and are referred to as allosteric sites has been adopted. These allosteric, non-ACh binding pockets are less well conserved between receptor subtypes, and targeting these sites has been highly successful for multiple G-protein coupled receptors including muscarinic receptors (Conn et al., 2009a; May et al., 2007). Allosteric activators can function mechanistically as either allosteric agonists, which in the course of binding to the allosteric site directly induce receptor activation, or as positive allosteric modulators (PAMs) which do not activate the receptor directly but potentiate the ability of the endogenous agonist ACh to induce receptor activation. These mechanisms are not exclusive in nature and in some cases a single molecule can have both allosteric potentiator and allosteric agonist activity (Conn et al., 2009b). As discussed below, the discovery of subtype-specific allosteric modulators has greatly advanced our understanding of the physiological role of various muscarinic receptor subtypes in the brain and have emphasized the potential utility of mAChR subtypes as targets for the development of novel antipsychotic treatments.

M4 Muscarinic Receptor

While the receptor subtypes that mediate the clinical antipsychotic efficacy of xanomeline have not been definitively established, several lines of evidence suggest that M1 and/or M4 are likely responsible. The M4 receptor is expressed in many brain regions including the striatum, cortex, and hippocampus (Hersch et al., 1994; Levey et al., 1995; 1991). This receptor canonically signals via activation of the Gai/Gao G-proteins with concomitant inhibition of adenylyl cyclase and cAMP production. M4 knock-out mice show an overall hyper-dopaminergic phenotype with increased locomotor activity, and increased basal- and psychostimulant-induced accumbal dopamine levels (Gomeza et al., 1999; Tzavara et al., 2004). M4 knock-out mice, unlike wild-type mice, are not susceptible to muscarinic agonist-induced reductions in striatal and accumbal dopamine (Threlfell et al., 2010), suggesting that M4 negatively regulates dopaminergic tone in these areas. M4 receptors have been found to co-localize with D1 dopamine receptors in the striatum (Ince et al., 1997), and selective deletion of M4 from D1-expressing cells is sufficient to enhance hyperlocomotor activity and increase behavioral sensitization to psychostimulants (Jeon et al., 2010). Further evidence of the importance of this subpopulation of M4 receptors to antipsychotic efficacy comes from the finding that these mice are no longer responsive to the antipsychotic effects of xanomeline as assessed by amphetamine-induced locomotion (Dencker et al., 2011).

The above studies suggest that M4-specific modulators could represent a novel therapeutic approach for the treatment of schizophrenia. A major breakthrough in M4 PAMs occurred in 2008 with the independent discoveries of two structurally distinct M4 PAMs, VU0010010 and LY2033298. The M4 modulator VU0010010 demonstrated robust and potent (EC50 = 400nM) activity at M4 and was void of activity at all the other mAChR subtypes. Extensive in vitro characterization demonstrated that VU0010010 bound to an allosteric site on M4 and that binding did not cause activation of the receptor directly, but instead increased the affinity of the receptor for ACh. In brain slice preparations VU010010 potentiated M4-mediated depression of glutamatergic, but not GABAergic, afferents onto CA1 pyramidal cells of the hippocampus indicating a key regulatory role for M4 in hippocampal function (Shirey et al., 2008). Chemical optimization of VU0010010 led to the discovery of two related PAMs, VU0152100 and VU0152099, demonstrating M4 subtype-specificity and possessing improved physiochemical properties making them suitable for in vivo administration. Importantly, both VU0152100 and VU0152099 have demonstrated efficacy in reversing amphetamine-induced hyperlocomotion in rats (Brady et al., 2008). LY2033298 represents another structurally-distinct M4-selective PAM that has specificity for the M4 receptor and can be administered to rodents in vivo. Systemic administration of LY2033298 produces robust effects in many preclinical models of psychosis including conditioned avoidance responding, and amphetamine-induced locomotion (Chan et al., 2008; Leach et al., 2010; Suratman et al., 2011). The collective data from these subtype-selective PAMs demonstrate that activation of M4 can be beneficial in numerous preclinical models predictive of anti-psychotic-like activity. These findings support the hypothesis that M4 is partially responsible for the clinical efficacy of xanomeline and highlight the potential of M4 PAMs as novel therapeutics for schizophrenia.

M1 Muscarinic Receptor

The M1 receptor is the predominant mAChR subtype in the CNS and is found in many brain regions including high levels of expression in the striatum, cortex, and hippocampus (Levey et al., 1995; 1991). This receptor canonically signals via activation of the Gaq G-protein with concomitant activation of phospholipase C leading to phosphoinositide hydrolysis and calcium mobilization. M1 knock-out mice are deficient in tasks requiring the medial prefrontal cortex (mPFC), and exhibit enhanced amphetamine-induced hyperactivity and hyperactive dopamine transmission (Anagnostaras et al., 2003; Gerber et al., 2001; Miyakawa et al., 2001). The high levels of M1 receptor expression in brain regions that are dysregulated in schizophrenia, combined with the clinical efficacy of the M1/M4 preferring agonist xanomeline, suggests that M1-selective compounds could be useful therapeutic agents for treating schizophrenia.

Over the past several years, numerous M1 allosteric agonists and PAMs have been discovered and have greatly furthered our understanding of M1 function in the brain. A major breakthrough in the discovery of M1 PAMs came with the discovery of BQCA, which is a highly M1-selective compound that interacts via an allosteric region of the receptor and has favorable pharmacokinetics and CNS exposure making it useful for in vivo studies (Ma et al., 2009). In wild-type, but not M1 knock-out mice, BQCA potentiates mAChR agonist-induced increases in inward currents, and the frequency of spontaneous EPSCs in mPFC pyramidal cells. In addition, BQCA increases the firing rates of these cells in vivo and enhances prefrontal cortical-dependent forms of cognitive function, providing strong collective evidence that M1 is an important regulator of prefrontal cortical function (Shirey et al., 2009). BQCA is also efficacious in reversing deficits in the acquisition of contextual fear induced by the administration of a non-selective mAChR antagonist, suggesting an important role for M1 in hippocampal function. Systemic administration of BQCA was also found to reverse amphetamine-induced hyperlocomotion (Ma et al., 2009) in a manner similar to that observed with M4 PAMs or the atypical antipsychotic clozapine. However, further studies with other M1-selective agents have not demonstrated robust effects on amphetamine-induced hyperlocomotor activity, suggesting that allosteric activators of M1 may not all have efficacy in this model that is often used to predict efficacy in treating the positive symptoms of schizophrenia. Together, these findings suggest that M1 PAMs may have greater efficacy in treating cognitive disturbances in schizophrenia patients, whereas M4 PAMs may be preferable for treating positive symptoms. However, future clinical studies will be required to establish the potential efficacy of M1- and M4-selective PAMs in reducing different symptom clusters in schizophrenia patients.

In addition to M1 PAMs, a number of systemically-active M1 agonists have been reported that appear to act as allosteric agonists and demonstrate high selectivity for activation of M1 relative to other mAChR subtypes. The mechanism of action for these allosteric agonists including 77-LH-28-1, VU0364572, and others, is complex and current data suggests that these molecules may bind in a bitopic manner involving interactions with both allosteric and orthosteric sites on the receptor (Avlani et al., 2010; Digby et al., 2012b). Several lines of evidence suggest that M1 activation via 77-LH-28-1, or VU0364752 and related compounds, can regulate hippocampal function and plasticity at the CA1-Schaffer collateral synapse via modulation of NMDA receptor currents in a context-dependent manner (Buchanan et al., 2010; Digby et al., 2012a; Jo et al., 2010; Jones et al., 2008; Langmead et al., 2008; Lebois et al., 2011). Unfortunately, while these M1 allosteric agonists can selectively activate M1 relative to other mAChRs, this selectivity is based on functional selectivity rather than selective high affinity binding to M1 (Digby et al., 2012b). In addition these compounds can differentially induce M1-mediated signaling to various pathways including Ca2+ mobilization and β-arrestin activation and can have differential effects that depend on varying levels of receptor reserve in different neuronal populations (Digby et al., 2012a). These issues, along with the bitopic nature of binding of most M1 allosteric agonists, complicate the activity of these compounds relative to the highly selective PAMs, such as BQCA. Based on this, M1 PAMs may represent a more viable approach to treatment of cognitive disturbances in schizophrenia than would allosteric agonists.

M5 Muscarinic Receptor

The M5 subtype of muscarinic receptor accounts for <2% of the total mAChRs expressed in the CNS (Yasuda et al., 1993). However, the expression pattern is highly-restricted to the cerebrovasculature as well as midbrain dopaminergic neurons, where it is the only detectable mAChR subtype (Weiner et al., 1990; Yamada et al., 2001). Similar to M1, the M5 receptor canonically signals via activation of the Gaq G-protein. Consistent with its localization to dopamine neurons, M5 knock-out mice display altered striatal dopamine release and blunted responses to drugs that modulate the dopaminergic system such as cocaine and opiates (Basile et al., 2002; Bendor et al., 2010; Thomsen et al., 2005). These mice also display deficits in prepulse inhibition (Thomsen et al., 2007) and exhibit reductions in amphetamine-induced hyperlocomotion (Wang et al., 2004). In addition to the knock-out studies in mice, genetic studies in humans have found that the M5 mAChR gene (CHRM5) along with the α7 nAChR gene (CHRNA7) are linked to schizophrenia susceptibility (De Luca et al., 2004).

The restricted pattern of M5 expression in midbrain dopamine neurons, along with the genetic evidence detailed above, suggest that M5 represents another compelling target for the development of novel antipsychotics. However, the lack of subtype selective ligands has limited our understanding of the function of this mAChR in circuitry relevant to schizophrenia. Recently, great strides have been made in developing M5-selective compounds, ultimately leading to the discovery of the first M5-selective PAM VU0238429. This compound interacts with the receptor via an allosteric binding site and robustly potentiates responses at M5 without activity at other mAChR subtypes (Bridges et al., 2009). Studies in brain slices have demonstrated that VU0238429 can potentiate both inward currents and Ca2+ mobilization induced by a non-selective mAChR agonist in dopamine neurons of the substantia nigra (Foster et al., 2012), demonstrating that activation of M5 can robustly modulate the dopaminergic system. Unfortunately, VU0238429 does not possess physiochemical properties amenable to in vivo dosing. Further optimization of M5-selective modulators is in progress (Bridges et al., 2010a; 2010b), and future in vivo studies will be necessary to fully elucidate the potential utility of M5- modulators as antipsychotics.

Targeting Nicotinic Receptors for Treatment of Schizophrenia

Numerous clinical and preclinical findings suggest that modulating nicotinic receptors (nAChRs) could provide therapeutic efficacy in treating schizophrenia. In healthy humans, administration of nAChR antagonists can induce schizophrenic symptoms including psychosis and amnesia. Interestingly, schizophrenics have a drastically higher incidence of cigarette smoking (>80%) compared to both the general population and to individuals with other psychiatric disorders (Miwa et al., 2011). While the cause of this is not completely understood, it is clear that acute nicotine exposure in schizophrenic patients can improve cognitive processes, including sensory information processing. Unfortunately, the effects observed with nicotine disappear quickly after administration (Adler et al., 1993). The transient efficacy and the associated cardiovascular and abuse liability limit the utility of nicotine as a therapeutic for patients with schizophrenia. However, novel subtype-selective agonists and PAMs of nicotinic receptors may provide positive cognitive outcomes that are sustained over time with fewer adverse effects.

Nicotinic receptors are pentameric ligand-gated ion channels that are comprised of β and/or a subunits and can be either homomeric or heteromeric in nature. Differences in subunit composition can alter receptor function by changing the receptors ligand-sensitivity, Ca2+ permeability, and desensitization kinetics. The α4β2 and α7 nAChR subtypes are the most prominently expressed subtypes in the brain and are located in several circuits shown to be imbalanced in schizophrenia (Fabian-Fine et al., 2001; Perry et al., 2002). α4β2 agonists have been found to be effective in modulating attention and memory in rodents and non-human primates (Buccafusco et al., 1995; Hahn et al., 2003). While there are currently no clinical reports regarding the efficacy of these molecules on schizophrenic patients, it has been found that the α4β2 agonist TC-1734 was well-tolerated and robustly improved age-associated memory impairments (Dunbar et al., 2011). Further studies are needed to determine the utility of such α4β2 agonists in treating schizophrenia.

The expression level of the α7 nAChR receptor is decreased in schizophrenic patients (Court et al., 1999) and the gene for this receptor has been linked to schizophrenia vulnerability (De Luca et al., 2004; Leonard et al., 2002). Among the compounds targeting nAChRs for the treatment of schizophrenia, the α7 agonist DXMB-A has made it the farthest in clinical development showing pro-cognitive efficacy in both a phase I study (Kitagawa et al., 2003), and a phase II study examining the effects of single-day administration of DMXB-A to schizophrenic patients (Olincy et al., 2006). Unfortunately, a follow-up phase II study examining the effects of DXMB-A after 4 weeks of daily dosing found no significant changes in cognitive improvements compared to placebo (Freedman et al., 2008). However, improvements were detected in the Scale for the Assessment of Negative Symptoms (SANS), particularly in the anhedonia and alogia sub-scales. It is difficult to definitively conclude if the absence of pro-cognitive efficacy after 4 weeks of DXMB-A administration is due to drug-induced desensitization, or whether the efficacy is masked by a significant practice-effect that was observed across all groups during test repetition. Recently, numerous α7 PAMs have been discovered, which due to their mechanism of action may be more refractory to causing desensitization. These PAMs (including PNU-120596 and A-867744) and several novel α7 agonists have been demonstrated to be efficacious in several preclinical models of schizophrenia (see Jones et al., 2012 for a comprehensive review), and represent exciting new avenues for treating the cognitive symptoms of schizophrenia.

Summary

Schizophrenia is a complex disease that is epitomized by symptoms spanning numerous clusters including cognitive, behavioral, and emotional disturbances. Current therapies are only partially effective at treating the behavioral symptoms and do not alleviate the cognitive or emotional deficits. Several preclinical and clinical studies suggest that cholinergic receptors represent novel targets with the potential to provide comprehensive and efficacious treatment of schizophrenia. While broad-spectrum modulators of muscarinic and nicotinic receptors have demonstrated clinical efficacy they have ultimately failed due to peripheral side-effects and a loss of efficacy over time. However, the discovery of receptor subtype-specific modulators has raised hopes that these molecules can provide long-lasting clinical efficacy with fewer adverse-effects. The coming years will be instrumental in elucidating the feasibility of modulating the M1, M4, M5, α4β2, and α7 cholinergic receptors as therapeutic treatments for schizophrenia.

Footnotes

Disclosure

P.J.C. is a consultant for Seaside Therapeutic and Karuna Pharmaceuticals and receives research support from Bristol-Myers Squibb. C.K.J. receives research support from Bristol-Myers Squibb. P.J.C. and C.K.J. are inventors on multiple composition of matter patents protecting allosteric modulators of mGlus and other GPCRs.

Contributor Information

Daniel J. Foster, Department of Pharmacology and Vanderbilt Center for Neuroscience, Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37202, USA

Carrie K. Jones, Department of Pharmacology and Vanderbilt Center for Neuroscience, Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37202, USA

P. Jeffrey Conn, Department of Pharmacology and Vanderbilt Center for Neuroscience, Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37202, USA

References

  1. Adler LE, Hoffer LD, Wiser A, Freedman R. Normalization of auditory physiology by cigarette smoking in schizophrenic patients. Am J Psychiatry. 1993;150(12):1856–1861. doi: 10.1176/ajp.150.12.1856. [DOI] [PubMed] [Google Scholar]
  2. Anagnostaras SG, Murphy GG, Hamilton SE, Mitchell SL, Rahnama NP, Nathanson NM, Silva AJ. Selective cognitive dysfunction in acetylcholine M1 muscarinic receptor mutant mice. Nat Neurosci. 2003;6(1):51–58. doi: 10.1038/nn992. [DOI] [PubMed] [Google Scholar]
  3. Avlani VA, Langmead CJ, Guida E, Wood MD, Tehan BG, Herdon HJ, Watson JM, Sexton PM, Christopoulos A. Orthosteric and allosteric modes of interaction of novel selective agonists of the M1 muscarinic acetylcholine receptor. Mol Pharmacol. 2010;78(1):94–104. doi: 10.1124/mol.110.064345. [DOI] [PubMed] [Google Scholar]
  4. Basile AS, Fedorova I, Zapata A, Liu X, Shippenberg T, Duttaroy A, Yamada M, Wess J. Deletion of the M5 muscarinic acetylcholine receptor attenuates morphine reinforcement and withdrawal but not morphine analgesia. Proc Natl Acad Sci U S A. 2002;99(17):11452–11457. doi: 10.1073/pnas.162371899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bendor J, Lizardi-Ortiz JE, Westphalen RI, Brandstetter M, Hemmings HC, Jr, Sulzer D, Flajolet M, Greengard P. AGAP1/AP-3-dependent endocytic recycling of M5 muscarinic receptors promotes dopamine release. EMBO J. 2010;29(16):2813–2826. doi: 10.1038/emboj.2010.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bodick NC, Offen WW, Levey AI, Cutler NR, Gauthier SG, Satlin A, Shannon HE, Tollefson GD, Rasmussen K, Bymaster FP, Hurley DJ, Potter WZ, Paul SM. Effects of xanomeline, a selective muscarinic receptor agonist, on cognitive function and behavioral symptoms in Alzheimer disease. Arch Neurol. 1997a;54(4):465–473. doi: 10.1001/archneur.1997.00550160091022. [DOI] [PubMed] [Google Scholar]
  7. Bodick NC, Offen WW, Shannon HE, Satterwhite J, Lucas R, Van Lier R, Paul SM. The selective muscarinic agonist xanomeline improves both the cognitive deficits and behavioral symptoms of Alzheimer disease. Alzheimer Dis Assoc Disord. 1997b;11(Suppl 4):S16–S22. [PubMed] [Google Scholar]
  8. Brady AE, Jones CK, Bridges TM, Kennedy JP, Thompson AD, Heiman JU, Breininger ML, Gentry PR, Yin H, Jadhav SB, Shirey JK, Conn PJ, Lindsley CW. Centrally active allosteric potentiators of the M4 muscarinic acetylcholine receptor reverse amphetamine-induced hyperlocomotor activity in rats. J Pharmacol Exp Ther. 2008;327(3):941–953. doi: 10.1124/jpet.108.140350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bridges TM, Kennedy JP, Cho HP, Breininger ML, Gentry PR, Hopkins CR, Conn PJ, Lindsley CW. Chemical lead optimization of a pan G(q) mAChR M(1), M(3), M(5) positive allosteric modulator (PAM) lead. Part I: Development of the first highly selective M(5) PAM. Bioorg Med Chem Lett. 2010a;20(2):558–562. doi: 10.1016/j.bmcl.2009.11.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bridges TM, Kennedy JP, Hopkins CR, Conn PJ, Lindsley CW. Heterobiaryl and heterobiaryl ether derived M5 positive allosteric modulators. Bioorg Med Chem Lett. 2010b;20(19):5617–5622. doi: 10.1016/j.bmcl.2010.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bridges TM, Marlo JE, Niswender CM, Jones CK, Jadhav SB, Gentry PR, Plumley HC, Weaver CD, Conn PJ, Lindsley CW. Discovery of the first highly M5-preferring muscarinic acetylcholine receptor ligand, an M5 positive allosteric modulator derived from a series of 5-trifluoromethoxy N-benzyl isatins. J Med Chem. 2009;52(11):3445–3448. doi: 10.1021/jm900286j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Buccafusco JJ, Jackson WJ, Terry AV, Jr, Marsh KC, Decker MW, Arneric SP. Improvement in performance of a delayed matching-to-sample task by monkeys following ABT-418: a novel cholinergic channel activator for memory enhancement. Psychopharmacology (Berl) 1995;120(3):256–266. doi: 10.1007/BF02311172. [DOI] [PubMed] [Google Scholar]
  13. Buchanan KA, Petrovic MM, Chamberlain SE, Marrion NV, Mellor JR. Facilitation of long-term potentiation by muscarinic M(1) receptors is mediated by inhibition of SK channels. Neuron. 2010;68(5):948–963. doi: 10.1016/j.neuron.2010.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bymaster FP, Carter PA, Yamada M, Gomeza J, Wess J, Hamilton SE, Nathanson NM, McKinzie DL, Felder CC. Role of specific muscarinic receptor subtypes in cholinergic parasympathomimetic responses, in vivo phosphoinositide hydrolysis, and pilocarpine-induced seizure activity. Eur J Neurosci. 2003;17(7):1403–1410. doi: 10.1046/j.1460-9568.2003.02588.x. [DOI] [PubMed] [Google Scholar]
  15. Chan WY, McKinzie DL, Bose S, Mitchell SN, Witkin JM, Thompson RC, Christopoulos A, Lazareno S, Birdsall NJ, Bymaster FP, Felder CC. Allosteric modulation of the muscarinic M4 receptor as an approach to treating schizophrenia. Proc Natl Acad Sci U S A. 2008;105(31):10978–10983. doi: 10.1073/pnas.0800567105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Conn PJ, Christopoulos A, Lindsley CW. Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nat Rev Drug Discov. 2009a;8(1):41–54. doi: 10.1038/nrd2760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Conn PJ, Jones CK, Lindsley CW. Subtype-selective allosteric modulators of muscarinic receptors for the treatment of CNS disorders. Trends Pharmacol Sci. 2009b;30(3):148–155. doi: 10.1016/j.tips.2008.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Court J, Spurden D, Lloyd S, Mckeith I, Ballard C, Cairns N, Kerwin R, Perry R, Perry E. Neuronal nicotinic receptors in dementia with Lewy bodies and schizophrenia: alpha-bungarotoxin and nicotine binding in the thalamus. J Neurochem. 1999;73(4):1590–1597. doi: 10.1046/j.1471-4159.1999.0731590.x. [DOI] [PubMed] [Google Scholar]
  19. Crook JM, Tomaskovic-Crook E, Copolov DL, Dean B. Decreased muscarinic receptor binding in subjects with schizophrenia: a study of the human hippocampal formation. Biol Psychiatry. 2000;48(5):381–388. doi: 10.1016/s0006-3223(00)00918-5. [DOI] [PubMed] [Google Scholar]
  20. De Luca V, Wang H, Squassina A, Wong GW, Yeomans J, Kennedy JL. Linkage of M5 muscarinic and alpha7-nicotinic receptor genes on 15q13 to schizophrenia. Neuropsychobiology. 2004;50(2):124–127. doi: 10.1159/000079102. [DOI] [PubMed] [Google Scholar]
  21. Dean B, Crook JM, Opeskin K, Hill C, Keks N, Copolov DL. The density of muscarinic M1 receptors is decreased in the caudateputamen of subjects with schizophrenia. Mol Psychiatry. 1996;1(1):54–58. [PubMed] [Google Scholar]
  22. Dencker D, Wortwein G, Weikop P, Jeon J, Thomsen M, Sager TN, Mork A, Woldbye DP, Wess J, Fink-Jensen A. Involvement of a subpopulation of neuronal M4 muscarinic acetylcholine receptors in the antipsychotic-like effects of the M1/M4 preferring muscarinic receptor agonist xanomeline. J Neurosci. 2011;31(16):5905–5908. doi: 10.1523/JNEUROSCI.0370-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Digby GJ, Noetzel MJ, Bubser M, Utley TJ, Walker AG, Byun NE, Lebois EP, Xiang Z, Sheffler DJ, Cho HP, Davis AA, Nemirovsky NE, Mennenga SE, Camp BW, Bimonte-Nelson HA, Bode J, Italiano K, Morrison R, Daniels JS, Niswender CM, et al. Novel allosteric agonists of M1 muscarinic acetylcholine receptors induce brain region-specific responses that correspond with behavioral effects in animal models. J Neurosci. 2012a;32(25):8532–8544. doi: 10.1523/JNEUROSCI.0337-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Digby GJ, Utley TJ, Lamsal A, Sevel C, Sheffler DJ, Lebois EP, Bridges TM, Wood MR, Niswender CM, Lindsley CW, Conn PJ. Chemical modification of the M1 agonist VU0364572 reveals molecular switches in pharmacology and a bitopic binding mode. ACS Chem Neurosci. 2012b;3(12):1025–1036. doi: 10.1021/cn300103e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Dunbar GC, Kuchibhatla RV, Lee G. A randomized double-blind study comparing 25 and 50 mg TC-1734 (AZD3480) with placebo, in older subjects with age-associated memory impairment. J Psychopharmacol. 2011;25(8):1020–1029. doi: 10.1177/0269881110367727. [DOI] [PubMed] [Google Scholar]
  26. Fabian-Fine R, Skehel P, Errington ML, Davies HA, Sher E, Stewart MG, Fine A. Ultrastructural distribution of the alpha7 nicotinic acetylcholine receptor subunit in rat hippocampus. J Neurosci. 2001;21(20):7993–8003. doi: 10.1523/JNEUROSCI.21-20-07993.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Feldman HH, Ferris S, Winblad B, Sfikas N, Mancione L, He Y, Tekin S, Burns A, Cummings J, Del Ser T, Inzitari D, Orgogozo JM, Sauer H, Scheltens P, Scarpini E, Herrmann N, Farlow M, Potkin S, Charles HC, Fox NC, et al. Effect of rivastigmine on delay to diagnosis of Alzheimer’s disease from mild cognitive impairment: the InDDEx study. Lancet Neurol. 2007;6(6):501–512. doi: 10.1016/S1474-4422(07)70109-6. [DOI] [PubMed] [Google Scholar]
  28. Forette F, Anand R, Gharabawi G. A phase II study in patients with Alzheimer’s disease to assess the preliminary efficacy and maximum tolerated dose of rivastigmine (Exelon) Eur J Neurol. 1999;6(4):423–429. doi: 10.1046/j.1468-1331.1999.640423.x. [DOI] [PubMed] [Google Scholar]
  29. Foster DJ, Gentry PR, Bridges TM, Xiang Z, Lindsley CW, Conn PJ. Examination of M5-muscarinic receptor neurophysiology in substantia nigra pars compacta neurons using the allosteric modulator VU0238429. Society for Neuroscience Meeting; New Orleans, LA. 2012. Abstr #38.05. [Google Scholar]
  30. Freedman R, Olincy A, Buchanan RW, Harris JG, Gold JM, Johnson L, Allensworth D, Guzman-Bonilla A, Clement B, Ball MP, Kutnick J, Pender V, Martin LF, Stevens KE, Wagner BD, Zerbe GO, Soti F, Kem WR. Initial phase 2 trial of a nicotinic agonist in schizophrenia. Am J Psychiatry. 2008;165(8):1040–1047. doi: 10.1176/appi.ajp.2008.07071135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Gerber DJ, Sotnikova TD, Gainetdinov RR, Huang SY, Caron MG, Tonegawa S. Hyperactivity, elevated dopaminergic transmission, and response to amphetamine in M1 muscarinic acetylcholine receptor-deficient mice. Proc Natl Acad Sci U S A. 2001;98(26):15312–15317. doi: 10.1073/pnas.261583798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Gomeza J, Zhang L, Kostenis E, Felder C, Bymaster F, Brodkin J, Shannon H, Xia B, Deng C, Wess J. Enhancement of D1 dopamine receptor-mediated locomotor stimulation in M(4) muscarinic acetylcholine receptor knockout mice. Proc Natl Acad Sci U S A. 1999;96(18):10483–10488. doi: 10.1073/pnas.96.18.10483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Hahn B, Sharples CG, Wonnacott S, Shoaib M, Stolerman IP. Attentional effects of nicotinic agonists in rats. Neuropharmacology. 2003;44(8):1054–1067. doi: 10.1016/s0028-3908(03)00099-6. [DOI] [PubMed] [Google Scholar]
  34. Hersch SM, Gutekunst CA, Rees HD, Heilman CJ, Levey AI. Distribution of m1–m4 muscarinic receptor proteins in the rat striatum: light and electron microscopic immunocytochemistry using subtype-specific antibodies. J Neurosci. 1994;14(5 Pt 2):3351–3363. doi: 10.1523/JNEUROSCI.14-05-03351.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Ince E, Ciliax BJ, Levey AI. Differential expression of D1 and D2 dopamine and m4 muscarinic acetylcholine receptor proteins in identified striatonigral neurons. Synapse. 1997;27(4):357–366. doi: 10.1002/(SICI)1098-2396(199712)27:4<357::AID-SYN9>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
  36. Jeon J, Dencker D, Wortwein G, Woldbye DP, Cui Y, Davis AA, Levey AI, Schutz G, Sager TN, Mork A, Li C, Deng CX, Fink-Jensen A, Wess J. A subpopulation of neuronal M4 muscarinic acetylcholine receptors plays a critical role in modulating dopamine-dependent behaviors. J Neurosci. 2010;30(6):2396–2405. doi: 10.1523/JNEUROSCI.3843-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Jo J, Son GH, Winters BL, Kim MJ, Whitcomb DJ, Dickinson BA, Lee YB, Futai K, Amici M, Sheng M, Collingridge GL, Cho K. Muscarinic receptors induce LTD of NMDAR EPSCs via a mechanism involving hippocalcin, AP2 and PSD-95. Nat Neurosci. 2010;13(10):1216–1224. doi: 10.1038/nn.2636. [DOI] [PubMed] [Google Scholar]
  38. Jones CK, Brady AE, Davis AA, Xiang Z, Bubser M, Tantawy MN, Kane AS, Bridges TM, Kennedy JP, Bradley SR, Peterson TE, Ansari MS, Baldwin RM, Kessler RM, Deutch AY, Lah JJ, Levey AI, Lindsley CW, Conn PJ. Novel selective allosteric activator of the M1 muscarinic acetylcholine receptor regulates amyloid processing and produces antipsychotic-like activity in rats. J Neurosci. 2008;28(41):10422–10433. doi: 10.1523/JNEUROSCI.1850-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Jones CK, Byun N, Bubser M. Muscarinic and nicotinic acetylcholine receptor agonists and allosteric modulators for the treatment of schizophrenia. Neuropsychopharmacology. 2012;37(1):16–42. doi: 10.1038/npp.2011.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Kitagawa H, Takenouchi T, Azuma R, Wesnes KA, Kramer WG, Clody DE, Burnett AL. Safety, pharmacokinetics, and effects on cognitive function of multiple doses of GTS-21 in healthy, male volunteers. Neuropsychopharmacology. 2003;28(3):542–551. doi: 10.1038/sj.npp.1300028. [DOI] [PubMed] [Google Scholar]
  41. Langmead CJ, Austin NE, Branch CL, Brown JT, Buchanan KA, Davies CH, Forbes IT, Fry VA, Hagan JJ, Herdon HJ, Jones GA, Jeggo R, Kew JN, Mazzali A, Melarange R, Patel N, Pardoe J, Randall AD, Roberts C, Roopun A, et al. Characterization of a CNS penetrant, selective M1 muscarinic receptor agonist, 77-LH-28–1. Br J Pharmacol. 2008;154(5):1104–1115. doi: 10.1038/bjp.2008.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Leach K, Loiacono RE, Felder CC, Mckinzie DL, Mogg A, Shaw DB, Sexton PM, Christopoulos A. Molecular mechanisms of action and in vivo validation of an M4 muscarinic acetylcholine receptor allosteric modulator with potential antipsychotic properties. Neuropsychopharmacology. 2010;35(4):855–869. doi: 10.1038/npp.2009.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Lebois EP, Digby GJ, Sheffler DJ, Melancon BJ, Tarr JC, Cho HP, Miller NR, Morrison R, Bridges TM, Xiang Z, Daniels JS, Wood MR, Conn PJ, Lindsley CW. Development of a highly selective, orally bioavailable and CNS penetrant M1 agonist derived from the MLPCN probe ML071. Bioorg Med Chem Lett. 2011;21(21):6451–6455. doi: 10.1016/j.bmcl.2011.08.084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Leonard S, Gault J, Hopkins J, Logel J, Vianzon R, Short M, Drebing C, Berger R, Venn D, Sirota P, Zerbe G, Olincy A, Ross RG, Adler LE, Freedman R. Association of promoter variants in the alpha7 nicotinic acetylcholine receptor subunit gene with an inhibitory deficit found in schizophrenia. Arch Gen Psychiatry. 2002;59(12):1085–1096. doi: 10.1001/archpsyc.59.12.1085. [DOI] [PubMed] [Google Scholar]
  45. Levey AI, Edmunds SM, Koliatsos V, Wiley RG, Heilman CJ. Expression of m1–m4 muscarinic acetylcholine receptor proteins in rat hippocampus and regulation by cholinergic innervation. J Neurosci. 1995;15(5 Pt 2):4077–4092. doi: 10.1523/JNEUROSCI.15-05-04077.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Levey AI, Kitt CA, Simonds WF, Price DL, Brann MR. Identification and localization of muscarinic acetylcholine receptor proteins in brain with subtype-specific antibodies. J Neurosci. 1991;11(10):3218–3226. doi: 10.1523/JNEUROSCI.11-10-03218.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Lewis DA, Lieberman JA. Catching up on schizophrenia: natural history and neurobiology. Neuron. 2000;28(2):325–334. doi: 10.1016/s0896-6273(00)00111-2. [DOI] [PubMed] [Google Scholar]
  48. Lieberman JA, Stroup TS, Mcevoy JP, Swartz MS, Rosenheck RA, Perkins DO, Keefe RS, Davis SM, Davis CE, Lebowitz BD, Severe J, Hsiao JK. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med. 2005;353(12):1209–1223. doi: 10.1056/NEJMoa051688. [DOI] [PubMed] [Google Scholar]
  49. Lisman JE, Coyle JT, Green RW, Javitt DC, Benes FM, Heckers S, Grace AA. Circuit-based framework for understanding neurotransmitter and risk gene interactions in schizophrenia. Trends Neurosci. 2008;31(5):234–242. doi: 10.1016/j.tins.2008.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Ma L, Seager MA, Wittmann M, Jacobson M, Bickel D, Burno M, Jones K, Graufelds VK, Xu G, Pearson M, Mccampbell A, Gaspar R, Shughrue P, Danziger A, Regan C, Flick R, Pascarella D, Garson S, Doran S, Kreatsoulas C, et al. Selective activation of the M1 muscarinic acetylcholine receptor achieved by allosteric potentiation. Proc Natl Acad Sci U S A. 2009;106(37):15950–15955. doi: 10.1073/pnas.0900903106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Marin O. Interneuron dysfunction in psychiatric disorders. Nat Rev Neurosci. 2012;13(2):107–120. doi: 10.1038/nrn3155. [DOI] [PubMed] [Google Scholar]
  52. 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]
  53. Miwa JM, Freedman R, Lester HA. Neural systems governed by nicotinic acetylcholine receptors: emerging hypotheses. Neuron. 2011;70(1):20–33. doi: 10.1016/j.neuron.2011.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Miyakawa T, Yamada M, Duttaroy A, Wess J. Hyperactivity and intact hippocampus-dependent learning in mice lacking the M1 muscarinic acetylcholine receptor. J Neurosci. 2001;21(14):5239–5250. doi: 10.1523/JNEUROSCI.21-14-05239.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Noetzel MJ, Jones CK, Conn PJ. Emerging approaches for treatment of schizophrenia: modulation of glutamatergic signaling. Discov Med. 2012;14(78):335–343. [PMC free article] [PubMed] [Google Scholar]
  56. Olincy A, Harris JG, Johnson LL, Pender V, Kongs S, Allensworth D, Ellis J, Zerbe GO, Leonard S, Stevens KE, Stevens JO, Martin L, Adler LE, Soti F, Kem WR, Freedman R. Proof-of-concept trial of an alpha7 nicotinic agonist in schizophrenia. Arch Gen Psychiatry. 2006;63(6):630–638. doi: 10.1001/archpsyc.63.6.630. [DOI] [PubMed] [Google Scholar]
  57. Osterholm RK, Camoriano JK. Transdermal scopolamine psychosis. JAMA. 1982;247(22):3081. [PubMed] [Google Scholar]
  58. Perry DC, Xiao Y, Nguyen HN, Musachio JL, Davila-Garcia MI, Kellar KJ. Measuring nicotinic receptors with characteristics of alpha4beta2, alpha3beta2 and alpha3beta4 subtypes in rat tissues by autoradiography. J Neurochem. 2002;82(3):468–481. doi: 10.1046/j.1471-4159.2002.00951.x. [DOI] [PubMed] [Google Scholar]
  59. Ross CA, Margolis RL, Reading SA, Pletnikov M, Coyle JT. Neurobiology of schizophrenia. Neuron. 2006;52(1):139–153. doi: 10.1016/j.neuron.2006.09.015. [DOI] [PubMed] [Google Scholar]
  60. Sarter M, Lustig C, Taylor SF. Cholinergic contributions to the cognitive symptoms of schizophrenia and the viability of cholinergic treatments. Neuropharmacology. 2012;62(3):1544–1553. doi: 10.1016/j.neuropharm.2010.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Sawa A, Snyder SH. Schizophrenia: neural mechanisms for novel therapies. Mol Med. 2003;9(1–2):3–9. [PMC free article] [PubMed] [Google Scholar]
  62. Shekhar A, Potter WZ, Lightfoot J, Lienemann J, Dube S, Mallinckrodt C, Bymaster FP, McKinzie DL, Felder CC. Selective muscarinic receptor agonist xanomeline as a novel treatment approach for schizophrenia. Am J Psychiatry. 2008;165(8):1033–1039. doi: 10.1176/appi.ajp.2008.06091591. [DOI] [PubMed] [Google Scholar]
  63. Shirey JK, Brady AE, Jones PJ, Davis AA, Bridges TM, Kennedy JP, Jadhav SB, Menon UN, Xiang Z, Watson ML, Christian EP, Doherty JJ, Quirk MC, Snyder DH, Lah JJ, Levey AI, Nicolle MM, Lindsley CW, Conn PJ. A selective allosteric potentiator of the M1 muscarinic acetylcholine receptor increases activity of medial prefrontal cortical neurons and restores impairments in reversal learning. J Neurosci. 2009;29(45):14271–14286. doi: 10.1523/JNEUROSCI.3930-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Shirey JK, Xiang Z, Orton D, Brady AE, Johnson KA, Williams R, Ayala JE, Rodriguez AL, Wess J, Weaver D, Niswender CM, Conn PJ. An allosteric potentiator of M4 mAChR modulates hippocampal synaptic transmission. Nat Chem Biol. 2008;4(1):42–50. doi: 10.1038/nchembio.2007.55. [DOI] [PubMed] [Google Scholar]
  65. Sullivan PF, Daly MJ, O’Donovan M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet. 2012;13(8):537–551. doi: 10.1038/nrg3240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Suratman S, Leach K, Sexton P, Felder C, Loiacono R, Christopoulos A. Impact of species variability and ‘probe-dependence’ on the detection and in vivo validation of allosteric modulation at the M4 muscarinic acetylcholine receptor. Br J Pharmacol. 2011;162(7):1659–1670. doi: 10.1111/j.1476-5381.2010.01184.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Tandon R, Shipley JE, Greden JF, Mann NA, Eisner WH, Goodson JA. Muscarinic cholinergic hyperactivity in schizophrenia. Relationship to positive and negative symptoms. Schizophr Res. 1991;4(1):23–30. doi: 10.1016/0920-9964(91)90006-d. [DOI] [PubMed] [Google Scholar]
  68. Thomsen M, Woldbye DP, Wortwein G, Fink-Jensen A, Wess J, Caine SB. Reduced cocaine self-administration in muscarinic M5 acetylcholine receptor-deficient mice. J Neurosci. 2005;25(36):8141–8149. doi: 10.1523/JNEUROSCI.2077-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Thomsen M, Wortwein G, Fink-Jensen A, Woldbye DP, Wess J, Caine SB. Decreased prepulse inhibition and increased sensitivity to muscarinic, but not dopaminergic drugs in M5 muscarinic acetylcholine receptor knockout mice. Psychopharmacology (Berl) 2007;192(1):97–110. doi: 10.1007/s00213-006-0682-y. [DOI] [PubMed] [Google Scholar]
  70. Threlfell S, Clements MA, Khodai T, Pienaar IS, Exley R, Wess J, Cragg SJ. Striatal muscarinic receptors promote activity dependence of dopamine transmission via distinct receptor subtypes on cholinergic interneurons in ventral versus dorsal striatum. J Neurosci. 2010;30(9):3398–3408. doi: 10.1523/JNEUROSCI.5620-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Tzavara ET, Bymaster FP, Davis RJ, Wade MR, Perry KW, Wess J, Mckinzie DL, Felder C, Nomikos GG. 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(12):1410–1412. doi: 10.1096/fj.04-1575fje. [DOI] [PubMed] [Google Scholar]
  72. Wang H, Ng K, Hayes D, Gao X, Forster G, Blaha C, Yeomans J. Decreased amphetamine-induced locomotion and improved latent inhibition in mice mutant for the M5 muscarinic receptor gene found in the human 15q schizophrenia region. Neuropsychopharmacology. 2004;29(12):2126–2139. doi: 10.1038/sj.npp.1300502. [DOI] [PubMed] [Google Scholar]
  73. Weiner DM, Levey AI, Brann MR. Expression of muscarinic acetylcholine and dopamine receptor mRNAs in rat basal ganglia. Proc Natl Acad Sci U S A. 1990;87(18):7050–7054. doi: 10.1073/pnas.87.18.7050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Yamada M, Lamping KG, Duttaroy A, Zhang W, Cui Y, Bymaster FP, McKinzie DL, Felder CC, Deng CX, Faraci FM, Wess J. Cholinergic dilation of cerebral blood vessels is abolished in M(5) muscarinic acetylcholine receptor knockout mice. Proc Natl Acad Sci U S A. 2001;98(24):14096–14101. doi: 10.1073/pnas.251542998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Yasuda RP, Ciesla W, Flores LR, Wall SJ, Li M, Satkus SA, Weisstein JS, Spagnola BV, Wolfe BB. Development of antisera selective for m4 and m5 muscarinic cholinergic receptors: distribution of m4 and m5 receptors in rat brain. Mol Pharmacol. 1993;43(2):149–157. [PubMed] [Google Scholar]
  76. Zavitsanou K, Katsifis A, Mattner F, Huang XF. Investigation of m1/m4 muscarinic receptors in the anterior cingulate cortex in schizophrenia, bipolar disorder, and major depression disorder. Neuropsychopharmacology. 2004;29(3):619–625. doi: 10.1038/sj.npp.1300367. [DOI] [PubMed] [Google Scholar]

RESOURCES