Abstract
Degeneration of the cholinergic system is considered to be the underlying pathology that results in the cognitive deficit in Alzheimer's disease. This pathology is thought to be linked to a loss of signaling through the cholinergic M1-muscarinic receptor subtype. However, recent studies have cast doubt on whether this is the primary receptor mediating cholinergic-hippocampal learning and memory. The current study offers an alternative mechanism involving the M3-muscarinic receptor that is expressed in numerous brain regions including the hippocampus. We demonstrate here that M3-muscarinic receptor knockout mice show a deficit in fear conditioning learning and memory. The mechanism used by the M3-muscarinic receptor in this process involves receptor phosphorylation because a knockin mouse strain expressing a phosphorylation-deficient receptor mutant also shows a deficit in fear conditioning. Consistent with a role for receptor phosphorylation, we demonstrate that the M3-muscarinic receptor is phosphorylated in the hippocampus following agonist treatment and following fear conditioning training. Importantly, the phosphorylation-deficient M3-muscarinic receptor was coupled normally to Gq/11-signaling but was uncoupled from phosphorylation-dependent processes such as receptor internalization and arrestin recruitment. It can, therefore, be concluded that M3-muscarinic receptor–dependent learning and memory depends, at least in part, on receptor phosphorylation/arrestin signaling. This study opens the potential for biased M3-muscarinic receptor ligands that direct phosphorylation/arrestin-dependent (non-G protein) signaling as being beneficial in cognitive disorders.
Keywords: fear conditioning, G protein-coupled receptor, hippocampus, ligand bias, Alzheimer's disease
Among the multitude of physiological responses regulated by G protein-coupled receptors (GPCRs), one of the most intriguing is the ability of this superfamily of cell-surface receptors to regulate neurological and behavioral processes such as learning and memory (1–4). The members of the muscarinic acetylcholine receptor family are prominent among the GPCR subtypes associated with cognitive function because lesions in cholinergic innervations to the hippocampus and other brain areas are widely thought to underlie the cognitive deficit observed in Alzheimer's disease (5). Whereas the M1-muscarinic receptor subtype has been proposed to be the subtype associated with acetylcholine-mediated cognition (6, 7), recent gene-knockout experiments have cast doubt on the direct role of this receptor subtype in learning and memory (1, 8). This has been reinforced by the discovery of a novel selective M1-muscarinic receptor antagonist that was effective in blocking M1-muscarinic receptor–mediated seizures in vivo but had no effect on hippocampal-based contextual fear conditioning (9). In addition, recent studies using an M1-muscarinic receptor–positive allosteric modulator, BQCA, have suggested that M1-muscarinic receptors can mediate learning and memory through an indirect mechanism by stimulating the prefrontal cortex (10). There is some controversy, however, because the same compound has been used in studies that suggest that M1-muscarinic receptors can directly act on hippocampal-based memory (11).
In light of this uncertainty, we investigate here the possibility that the M3-muscarinic receptor, which is expressed widely in the central nervous system including the hippocampus (12), might play a role in Pavlovian fear conditioning learning and memory. We also probe the contribution that receptor phosphorylation might play by using a knockin mouse strain that expresses a phosphorylation-deficient mutant of the M3-muscarinic receptor. Our data point to an important role for the M3-muscarinic receptor in hippocampal-based learning and memory and to the fact that this process depends on the phosphorylation status of the receptor.
Results
Involvement of the M3-Muscarinic Receptor in Fear Conditioning.
M3-muscarinic receptor knock out mice (M3R-KO) (13) showed a profound deficit in contextual [hippocampal based (14)] fear conditioning response (Fig. 1 A and B). In contrast, the tone fear conditioning response [associated with amygdala based learning and memory (15)] showed a trend toward a deficit, but the overall response was not significantly different from control mice (Fig. 1A). Importantly, there was no significant difference in nociception, as determined by pain thresholds, nor innate anxiety as measured in the elevated plus maze, between wild-type and M3R-KO mice (Fig. 1 C and D). These data indicate an important role for the M3-muscarinic receptor in fear conditioning learning and memory.
Fig. 1.
Fear conditioning response in wild-type and M3R-KO mice. Wild-type (WT) or M3-muscarinic receptor knockout mice (KO) were subjected to fear conditioning training. 24 h after training mice were analyzed for contextual fear conditioning and 24 h after this the animals were analyzed for cued fear conditioning. (A) Cumulative fear conditioning responses in WT (n = 10) and KO (n = 10) mice. (B) Example of a single movement trace from WT and a KO mouse undergoing a test for contextual fear conditioning. Freezing is defined when movement amplitude falls below the white line. Length and frequency of freezing is also represented by the yellow bars on the bar graph. (C) Measure for anxiety was conducted on WT (n = 10) and KO (n = 10) mice using the elevated plus maze. (D) Pain thresholds determined in WT (n = 10) and KO (n = 10) mice. Data represents the means ± SE. *, P < 0.01 (t test).
Generation of a Knockin Mouse Strain Expressing a Phosphorylation-Deficient M3-Muscarinic Receptor.
Nearly all GPCRs are rapidly phosphorylated in response to agonist stimulation at multiple sites on the intracellular loops and C-terminal tail (16). The M3-muscarinic receptor is no exception, being phosphorylated by a number of protein kinases at serine clusters contained within the third intracellular loop (17, 18). We tested the possibility that M3-muscarinic receptor phosphorylation was important in the mechanism of action of this receptor in physiological responses. Toward this goal, we generated mutant mice in which the wild-type M3-muscarinic receptor coding sequence had been replaced (by homologous recombination) with a mutant version of the receptor containing 15 point mutations in serine phospho-acceptor sites within the third intracellular loop of the receptor (Fig. S1 A–C). Mice homozygous for the knockin gene targeting event (designated M3R-KI) developed into adult animals with no gross physiological defects and showed normal Mendelian breeding.
Ligand binding analysis of cerebellar granule cell (CG) neurons using the hydrophilic antagonist [H3]-N-methyl scopolamine that labels cell surface receptors demonstrated no significance difference in the expression of the mutant receptor in neurons derived from M3R-KI mice compared with wild-type controls [wild-type Bmax= 167 ± 1 fmol/mg protein, M3R-KI Bmax = 143 ± 3 fmol/mg protein (n = 3 ± SE)]. Furthermore, immunoprecipitation of the M3-muscarinic receptor from extracts of CG neurons or from adult hippocampal extracts followed by Western blotting for the M3-muscarinic receptor revealed that the receptor was expressed similarly in both M3R-KI and wild-type neurons (Fig. S1 D and E).
As anticipated, the mutant M3-muscarinic receptor showed reduced levels of agonist-mediated phosphorylation as determined by [32P]orthophosphate labeling of CG neurons derived from wild-type and M3R-KI mice. The mutant M3-muscarinic receptor showed a 63 ± 1% reduction in agonist-mediated phosphorylation compared with wild-type receptor controls (Fig. 2 A and B).
Fig. 2.
Characterization of the phosphorylation-deficient M3-muscarinic receptor. (A) Phosphorylation of the M3-muscarinic receptor was determined in CG neurons derived from wild-type (WT) or M3R-KI (KI) mice metabolically labeled with [32P]orthophosphate. CG neurons were stimulated with or without methacholine (Meth, 100 μM) for 5 min before solubilization and immunoprecipitation of the M3-muscarinic receptor. Shown is an example autoradiograph (showing long and short exposures) and the Western blot loading control (M3-Receptor Western). (B) Quantification of the receptor phosphorylation data shown in A. The data represents the mean ± SE from four independent experiments. (C) Coupling of the M3-muscarinic receptor to the phosphoinositide pathway was determined in CG neurons derived from wild-type (WT) or M3R-KI (KI) mice transfected with the phosphoinositide biosensor (eGFP-PHPLCδ1). CG neurons were stimulated with methacholine (0.1 mM; downward arrow) for the indicated time (the bar represents 30 s). Agonist was then removed (upward arrow). The translocation of the biosensor from the plasma membrane to the cytoplasm was monitored on an inverted epifluorescence microscope. Shown are images of a single neuron before and during stimulation with methacholine. On the left is a representative time course of fluorescence change in the cytoplasm expressed as a self ratio (F/F0). (D) Internalization of M3-muscarinic receptors expressed in CG neurons derived from wild-type (WT) and M3R-KI (KI) mice determined by stimulating cultures at 37 °C in the absence or presence of methacholine (0.1 mM) for 30 min. Shown is the mean percentage receptor internalization ± SE (n = 5). (E and F) β-Arrestin recruitment to the M3-muscarinic receptor determined using PathHunter CHO-K1 cells expressing either the wild-type human M3-muscarinic receptor (WT) or the mutated human M3-muscarinic receptor lacking phospho-acceptor sites (H-M3phos-neg) (18). Shown are concentration response curves to the full muscarinic receptor agonists methacholine and acetylcholine. The data are the means ± SE of three independent experiments.
Phosphorylation-Deficient M3-Muscarinic Receptor Shows “G Protein Bias” in Neurons.
To determine whether the phosphorylation-deficient M3-muscarinic receptor was functionally coupled to Gq/11 proteins in CG neurons, a biosensor for phosphoinositide signaling was used. This biosensor consisted of the pleckstrin homology (PH) domain of phospholipase Cδ1 fused to eGFP (eGFP-PHPLCδ1), which binds phosphoinositide 4,5-bisphosphate (PIP2) in the membrane under basal conditions but translocates to the cytoplasm following receptor-mediated PIP2 hydrolysis (19). Using this biosensor, we found that the coupling of the mutant receptor expressed in M3R-KI mice to the phosphoinositide pathway was not significantly different from that observed for the wild-type receptor (Fig. 2C and Fig. S2). These data were confirmed in recombinant CHO cells expressing the phosphorylation-deficient M3-muscarinic receptor where stimulation of inositol phosphate-mediated calcium mobilization was not significantly different from the wild-type receptor (Fig. S3 A–C).
In contrast, there was a significant deficit in the coupling of the phosphorylation-deficient M3-muscarinic receptor to phosphorylation/arrestin-dependent processes. GPCR internalization is well known to depend on the phosphorylation of the receptor and the subsequent recruitment of arrestin (20). Here, we show that internalization of the M3-muscarinic receptor in CG neurons was significantly reduced in neurons derived from the M3R-KI mice, indicating that there was a deficit in phosphorylation/arrestin signaling (Fig. 2D). We next tested whether removal of the phospho-acceptor sites on the M3-muscarinic receptor affected the ability of the receptor to recruit arrestin. These experiments were conducted on the human M3-muscarinic receptor where we have shown previously that mutation of the phospho-acceptor sites (comparable to those of the mouse receptor) resulted in a reduction in both agonist-mediated receptor phosphorylation and receptor internalization (18). In an assay for the recruitment of β-arrestin, the phosphorylation-deficient M3-muscarinic receptor showed a significant reduction in both the potency and efficacy of arrestin-recruitment in response to the full agonists methacholine and acetylcholine (Fig. 2 E and F) [EC50 for arrestin recruitment to the wild-type receptor with methacholine and acetylcholine was −5.35 ± 0.1 M and −5.57 ± 0.1 M (log10), respectively, compared with −4.95 ± 0.1 M and −5.14 ± 0.1 M (log10) for the phosphorylation-deficient receptor (n = 5 ± SE)]. Consistent with the data from the mouse M3-muscarinic receptor, mutation of the phosphorylation sites in the human receptor did not affect coupling to the Gq/11/calcium mobilization pathway (Fig. S4 A and B).
Involvement of M3-Muscarinic Receptor Phosphorylation in Fear Conditioning.
The M3R-KI mice were tested for fear conditioning learning and memory. In these experiments, the wild-type mice spent 49.2 ± 8.9% of the time frozen in the contextual fear conditioning test and 63.9 ± 7.2% of the time frozen in the tone-associated fear conditioning test (Fig. 3 A and B). These responses were significantly reduced in M3R-KI animals, where the contextual fear conditioning response was 28.6 ± 3.2% and the tone-associated fear conditioning response was 47.5 ± 3.4% (Fig. 3 A and B). No significant difference was observed in controls for nociception or innate anxiety (Fig. 3 C and D).
Fig. 3.
Fear conditioning response in wild-type and M3R-KI mice. Wild-type (WT) or M3R-KI (KI) mice were subjected to fear conditioning training. Twenty-four hours after training, mice were analyzed for contextual fear conditioning, and 24 h later the animals were analyzed for cued fear conditioning. (A) Cumulative fear conditioning responses in WT (n = 10) and M3R-KI (n = 10) mice. (B) Example of a single movement trace from a WT and a M3R-KI (KI) mouse undergoing a test for contextual fear conditioning. Freezing is defined when movement amplitude falls below the white line. Length and frequency of freezing are also represented by the yellow bars on the bar graph. (C) Measure for anxiety was conducted on WT (n = 10) and M3R-KI (n = 10) mice using the elevated plus maze. (D) Pain thresholds were determined in WT (n = 10) and M3R-KI (n = 10) mice. Data represents means ± SE. *, P < 0.05 (t test).
These data firstly point to an important role for receptor phosphorylation in the mechanism of M3-muscarinic receptor–mediated fear conditioning learning and memory. Furthermore, due to the G protein bias of the receptor, the data indicate that coupling of the receptor to heterotrimeric G proteins is not the primary mechanism of mediating the fear conditioning response but that rather it is via a G protein-independent process that involves receptor phosphorylation.
The importance of receptor phosphorylation in the fear conditioning response was further confirmed in studies of neuronal activity as measured by changes in the expression of the early/intermediate gene c-Fos. Here, c-Fos expression was rapidly induced in the hippocampus and amygdala of wild-type mice following fear conditioning (Fig. 4). An observation consistent with other studies (21). However, the c-Fos expression after fear conditioning in M3R-KI mice was significantly reduced in the hippocampus when compared with controls (Fig. 4 A and B). Hence, in wild-type animals c-Fos expression in the dentate gyrus increased by 359% and 219%, 30 and 120 min after fear conditioning, respectively. In contrast, c-Fos immunostaining in the dentate gyrus of the M3R-KI mice increased by only 167% and 68%, 30 and 120 min after fear conditioning, respectively (Fig. 4A). Similarly, the induction of c-Fos expression was significantly attenuated in the CA3 region of the hippocampus in M3R-KI animals when compared with wild-type controls (Fig. 4B). Importantly, despite the fact that c-Fos expression in amygdala was substantially increased following fear conditioning, there was no significant difference in c-Fos expression in this brain region observed following fear conditioning in the wild-type and M3R-KI mice (Fig. 4C).
Fig. 4.
c-Fos induction in the hippocampus and amygdala following fear conditioning depends on M3-muscarinic receptor phosphorylation. Wild-type (WT) or M3R-KI (KI) mice were subjected to fear conditioning training, and the animals were killed by cardiac perfusion of fixative solution 30 or 120 min after training. The brain was then dissected and sectioned before staining for c-Fos expression. Basal conditions represent animals that had not undergone fear conditioning training. (A) Immunofluorescent staining for c-Fos expression in the dentate gyrus of the hippocampus. (B) Immunofluorescent staining for c-Fos expression in the CA3 region of the hippocampus. (C) Immunofluorescent staining for c-Fos expression in the amygdala. Graphs represent quantification of the immunofluorescent staining showing means ± SE (n = 4). *, P < 0.05 (t test). c-Fos appears as the red label and nuclei are stained blue using DAPI.
Interestingly, previous studies had determined that the M3-muscarinic receptor can directly up-regulate c-Fos expression (22). We investigated here whether this process depended on β-arrestin. siRNA knockdown of β-arrerstin1/2 in CHO cells stably expressing the M3-muscarinic receptor resulted in a marked reduction in M3-muscarinic receptor–mediated c-Fos up-regulation (Fig. S5). These data raise the possibility that those neurons in the hippocampus that express the M3-muscarinic receptor may show reduced c-Fos up-regulation in M3R-KI mice where receptor signaling via β-arrestin is disrupted.
M3-Muscarinic Receptor Phosphorylation in the Hippocampus.
Because our data pointed to a role for M3-muscarinic receptor phosphorylation in hippocampal-based fear conditioning learning and memory, we investigated directly the phosphorylation status of the receptor. Mass spectrometry of the recombinant M3-muscarinic receptor expressed in Chinese hamster ovary cells demonstrated that the receptor was phosphorylated on serine384 in the third intracellular loop (Fig. S6A). A phospho-specific antibody against this site was generated and confirmed to be phospho-specific on the basis that it did not recognize the nonphosphorylated epitope present in a bacterial fusion protein containing the third intracellular loop of the receptor (Fig. S6B). Furthermore, the antibody did identify the intact M3-muscarinic receptor immunoprecipitated from transfected CHO cells (Fig. S6C) but not following treatment with calf intestinal phosphatase which de-phosphorylated the receptor (Fig. S6C).
Although it was clear from our previous studies that the M3-muscarinic receptor was phosphorylated at multiple sites (18), not just serine384, we nevertheless investigated the phosphorylation at this single site as an exemplar of receptor phosphorylation. Immunoprecipitation of the M3-muscarinic receptor from wild-type hippocampus followed by Western blotting using the anti-phosphoserine384 antibody revealed that the receptor was phosphorylated at serine384 in the basal state in the mouse hippocampus (Fig. 5A). The phosphorylation status of the receptor at serine384 increased by 75% following the incubation of the dissected hippocampus with the muscarinic receptor agonist methacholine (Fig. 5A). In these experiments, the specificity of the anti-phosphoserine384 antibody was further confirmed by the lack of a band associated with the M3-muscarinic receptor in hippocampal preparations from M3R-KI mice expressing the phosphorylation-deficient M3-muscarinic receptor mutant, which includes the Ser384 to Ala mutation (Fig. 5A).
Fig. 5.
Phosphorylation of serine384 on the M3-muscarinic receptor in the hippocampus. (A) Hippocampi dissected from wild-type (WT) or M3R-KI (KI) mice were exposed to vehicle or methacholine (Meth, 1 mM) for 10 min. Membranes were then prepared and solubilized. The M3-muscarinic receptor was then immunoprecipitated using an anti-M3-muscarinic receptor monoclonal antibody and the immunoprecipitate probed in a Western blot using anti-phosphoserine384 antibody. The blot was then stripped and reprobed with a rabbit polyclonal anti-receptor antibody as a loading control. A typical experiment is shown together with the quantification from three independent experiments. (B) Wild-type mice were subjected to fear conditioning training, and the animals were killed either immediately after training (time point zero) or 5 min after training. The hippocampi were then dissected and membranes were prepared. Phosphorylation of the M3-muscarinic receptor using anti-phosphoserine384 antibody was then determined as in A. As a control for the stress response to the footshock, animals were subjected to immediate footshock with no fear conditioning training. Under these conditions there was no significant change in the phosphorylation status of serine 384 (Right). Typical experiments are shown together with the quantification from three independent experiments. The graphical data represent the means ± SE of three independent experiments. *, P < 0.05 (t test).
These data established that M3-muscarinic receptor phosphorylation at serine384 is regulated by agonist in the hippocampus. We next examined whether physiological stimuli, such as fear conditioning, could mediate changes in the phosphorylation status of the receptor. To test this possibility, mice were killed either immediately after fear conditioning training (time point zero) or 30 min after training. The receptor was then immunoprecipitated from hippocampal lysates and probed with the anti-phosphoserine384 antibody. The phosphorylation status of the receptor increased by 30% and 39% immediately after training (time point zero) and 30 min after training, respectively (Fig. 5B), demonstrating that the phosphorylation status of the M3-muscarinic receptor did change in response to fear conditioning training. Importantly, applying an immediate footshock with no fear conditioning training did not result in a significant change in the phosphorylation status of serine384 (Fig. 5B).
Discussion
We show here that M3R-KO mice displayed a deficit in fear conditioning learning and memory, indicating that this receptor subtype is important in the mechanism of cholinergic-mediated learning and memory. This finding may have significant clinical implications because stimulation of cholinergic transmission by the use of acetyl cholinesterase inhibitors is the front line treatment for the cognitive deficit in Alzheimer's disease (23). The target for the enhanced cognition in response to this treatment was thought to be M1-muscarinic receptor signaling. This notion is based on the fact that the M1-muscarinic receptor is the most highly expressed muscarinic receptor subtype in the cortex and hippocampus (12) and that pharmacological studies point to this receptor subtype as being the primary effector of cholinergic-mediated cognition (6, 24, 25). However, recent studies using M1-muscarinic receptor knockout mice (1) together with studies using more selective pharmacological tools (9, 10) have suggested that the M1-muscarinic receptor does not mediate hippocampal cognition directly. Hence, our finding that the M3-muscarinic receptor is important in hippocampal-based learning and memory offers an alternative explanation for the positive effects of promoting cholinergic transmission in patients with a cognitive deficiency.
Interestingly, earlier studies on mice where the M3-muscarinic receptor had been deleted had not identified a deficit in fear conditioning learning and memory (13). These studies did, however, use mice on a different genetic background (C57BL/J6 × 129SvEv) as well as a different experimental setup (13). Furthermore, the primary focus of these earlier studies was not learning and memory.
The mechanism by which the M3-muscarinic receptor mediates fear conditioning depends on the phosphorylation status of the receptor. This was revealed by using a knockin mouse strain that expressed a phosphorylation-deficient mutant of the M3-muscarinic receptor. This mutant mouse was shown to have a deficit in fear conditioning learning and memory similar to that observed in the receptor knockout mouse. Furthermore, the M3-muscarinic receptor was phosphorylated (at least on serine384) in the hippocampus in an agonist-dependent fashion and phosphorylation at serine384 was also up-regulated in the hippocampus following fear conditioning. These data point to the possibility that agonist-mediated phosphorylation of the M3-muscarinic receptor at the hippocampus is a regulatory step in fear conditioning learning and memory.
Importantly, mutation of the phospho-acceptor sites on the M3-muscarinic receptor did not affect cell surface or tissue expression of the receptor. The mutations also did not significantly affect the ability of the receptor to couple to Gq/11/calcium signaling. Interestingly, signaling of the phosphorylation-deficient receptor to Gq/11 in neurons and transfected cells did not show signs of altered desensitization. Because previous studies had demonstrated that M3-muscarinic receptor Gq/11-responses can be subject to phosphorylation-dependent desensitization (26, 27), it is possible that the phosphorylation-deficient mutant receptor, in vivo, might have different rates of desensitization that might impact on learning and memory.
The phosphorylation-deficient receptor was, however, defective in phosphorylation-dependent signaling such as receptor internalization and the recruitment of arrestin. In this way the mutant M3-muscarinic receptor could be described as showing G protein bias (28) where signaling is biased toward Gq/11-signaling pathways as opposed to phosphorylation/arrestin-dependent signaling pathways. Because mice expressing this receptor mutant showed a defect in hippocampal learning and memory, it would seem that the mechanism used by the M3-muscarinic receptor in mediating learning and memory is, at least in part, via phosphorylation and arrestin-dependent signaling. This conclusion is important in the context of recently described “biased ligands” that are able to direct signaling of GPCRs selectively through phosphorylation/arrestin-dependent pathways (28). Such biased ligands have been described for a number of GPCRs (e.g., ref. 29) and raises the possibility that a biased ligand to the M3-muscarinic receptor that directed the receptor toward phosphorylation and arrestin-dependent (non-G protein) signaling would promote cholinergic-mediated learning and memory.
The M3-muscarinic receptor can be phosphorylated by a number of protein kinases including members of the GRK family (26, 27) as well as protein kinase CK2 (18) and CK1α (17). In this respect M3-muscarinic receptors are similar to many other GPCRs where phosphorylation at multiple sites by more than one protein kinase has been observed (30). This has led to the notion that tissue specific GPCR signaling could be mediated, in part, by the employment of a subset of the receptor kinases resulting in a tissue specific phosphorylation profile (16). Whereas it was not possible in the current study to define the receptor kinases used in the phosphorylation of the M3-muscarinic receptor in the hippocampus, it would seem plausible that one or more of the protein kinases identified from in vitro studies are used to impart a phosphorylation profile on the receptor that allows for the regulation of learning and memory.
In conclusion, the deficit in fear conditioning shown by the M3R-KO mice implicates this receptor in cholinergic-mediated learning and memory. The fact that the mutant mice expressing a phosphorylation-deficient M3-muscarinic receptor showed a similar fear conditioning deficit to the M3R-KO mice demonstrates the importance of receptor phosphorylation, and potentially arrestin recruitment, in the mechanism of action of this receptor in learning and memory. In light of these findings it would seem plausible that biased ligands that direct the signaling of the M3-muscarinic receptor through phosphorylation/arrestin pathways might be of clinical benefit in the treatment of cognitive disorders.
Materials and Methods
Generation of the Muscarinic Receptor Mutant Strain M3R-KI Mice.
The mouse M3-muscarinic receptor coding sequence was subjected to consecutive rounds of mutagenesis (QuikChange; Stratagene) to generate a receptor, termed M3Rphos-neg with serine to alanine mutations at the following positions: 285, 286, 288, 290, 291, 302, 303, 331, 332, 333, 335, 374, 379, 384, and 385 (Fig. S1A). This receptor was used to replace (knock in) the coding sequence of the M3-muscarinic receptor using homologous recombination. A detailed description can be found in SI Materials and Methods.
Fear Conditioning.
For all behavioral tests, 8- to 15-week-old mice were used. For fear conditioning, mice were placed in the conditioning chamber (Coulbourn Instruments) and after a 2-min adaptation period received three tone/footshock pairings where the footshock [unconditioned stimulus (US); 2 s, 0.4 mA) always co-terminated with a tone [conditioned stimulus (CS); 30 s, 2.8 kHz, 85 dB]. The next day mice were placed back in the conditioning chamber and percent of time spent freezing was recorded for 3 min to assess context-dependent learning. Cued-conditioning was evaluated 48 h after training. For immunohistochemistry, mice were killed either 30 min or 2 h after the training session.
See SI Materials and Methods for a detailed explanation of the methods used.
Supplementary Material
Acknowledgments
We thank Prof. Ian Forsythe and Martin Haustein for help and support. A.B.T. is supported by the Wellcome Trust (047600). R.P. is supported by Marie Curie Excellence Grant MEXT-CT-2006-042265 from the European Commission.
Footnotes
The authors declare no conflict of interest.
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.0914801107/-/DCSupplemental.
References
- 1.Anagnostaras SG, et al. Selective cognitive dysfunction in acetylcholine M1 muscarinic receptor mutant mice. Nat Neurosci. 2003;6:51–58. doi: 10.1038/nn992. [DOI] [PubMed] [Google Scholar]
- 2.Catapano LA, Manji HK. G protein-coupled receptors in major psychiatric disorders. Biochim Biophys Acta. 2007;1768:976–993. doi: 10.1016/j.bbamem.2006.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Langmead CJ, Watson J, Reavill C. Muscarinic acetylcholine receptors as CNS drug targets. Pharmacol Ther. 2008;117:232–243. doi: 10.1016/j.pharmthera.2007.09.009. [DOI] [PubMed] [Google Scholar]
- 4.Wess J, Eglen RM, Gautam D. Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development. Nat Rev Drug Discov. 2007;6:721–733. doi: 10.1038/nrd2379. [DOI] [PubMed] [Google Scholar]
- 5.McKinney M, Jacksonville MC. Brain cholinergic vulnerability: relevance to behavior and disease. Biochem Pharmacol. 2005;70:1115–1124. doi: 10.1016/j.bcp.2005.05.019. [DOI] [PubMed] [Google Scholar]
- 6.Caccamo A, et al. M1 receptors play a central role in modulating AD-like pathology in transgenic mice. Neuron. 2006;49:671–682. doi: 10.1016/j.neuron.2006.01.020. [DOI] [PubMed] [Google Scholar]
- 7.Fisher A, et al. M1 muscarinic agonists can modulate some of the hallmarks in Alzheimer's disease: implications in future therapy. J Mol Neurosci. 2003;20:349–356. doi: 10.1385/JMN:20:3:349. [DOI] [PubMed] [Google Scholar]
- 8.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;21:5239–5250. doi: 10.1523/JNEUROSCI.21-14-05239.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sheffler DJ, et al. A novel selective muscarinic acetylcholine receptor subtype 1 antagonist reduces seizures without impairing hippocampus-dependent learning. Mol Pharmacol. 2009;76:356–368. doi: 10.1124/mol.109.056531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Shirey JK, et al. A selective allosteric potentiator of the M1 muscarinic acetylcholine receptor increases activity of medial prefrontal cortical neurons and restores impairments in reversal learning. J Neurosci. 2009;29:14271–14286. doi: 10.1523/JNEUROSCI.3930-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ma L, et al. Selective activation of the M1 muscarinic acetylcholine receptor achieved by allosteric potentiation. Proc Natl Acad Sci USA. 2009;106:15950–15955. doi: 10.1073/pnas.0900903106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.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;15:4077–4092. doi: 10.1523/JNEUROSCI.15-05-04077.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yamada M, et al. Mice lacking the M3 muscarinic acetylcholine receptor are hypophagic and lean. Nature. 2001;410:207–212. doi: 10.1038/35065604. [DOI] [PubMed] [Google Scholar]
- 14.Phillips RG, LeDoux JE. Differential contribution of amygdala and hippocampus to cued and contextual fear conditioning. Behav Neurosci. 1992;106:274–285. doi: 10.1037//0735-7044.106.2.274. [DOI] [PubMed] [Google Scholar]
- 15.Maren S, Aharonov G, Fanselow MS. Retrograde abolition of conditional fear after excitotoxic lesions in the basolateral amygdala of rats: absence of a temporal gradient. Behav Neurosci. 1996;110:718–726. doi: 10.1037//0735-7044.110.4.718. [DOI] [PubMed] [Google Scholar]
- 16.Tobin AB, Butcher AJ, Kong KC. Location, location, location…site-specific GPCR phosphorylation offers a mechanism for cell-type-specific signalling. Trends Pharmacol Sci. 2008;29:413–420. doi: 10.1016/j.tips.2008.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Budd DC, McDonald JE, Tobin AB. Phosphorylation and regulation of a Gq/11-coupled receptor by casein kinase 1alpha. J Biol Chem. 2000;275:19667–19675. doi: 10.1074/jbc.M000492200. [DOI] [PubMed] [Google Scholar]
- 18.Torrecilla I, et al. Phosphorylation and regulation of a G protein-coupled receptor by protein kinase CK2. J Cell Biol. 2007;177:127–137. doi: 10.1083/jcb.200610018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hirose K, Kadowaki S, Tanabe M, Takeshima H, Iino M. Spatiotemporal dynamics of inositol 1,4,5-trisphosphate that underlies complex Ca2+ mobilization patterns. Science. 1999;284:1527–1530. doi: 10.1126/science.284.5419.1527. [DOI] [PubMed] [Google Scholar]
- 20.Lefkowitz RJ, Rajagopal K, Whalen EJ. New roles for beta-arrestins in cell signaling: not just for seven-transmembrane receptors. Mol Cell. 2006;24:643–652. doi: 10.1016/j.molcel.2006.11.007. [DOI] [PubMed] [Google Scholar]
- 21.Beck CH, Fibiger HC. Conditioned fear-induced changes in behavior and in the expression of the immediate early gene c-fos: with and without diazepam pretreatment. J Neurosci. 1995;15:709–720. doi: 10.1523/JNEUROSCI.15-01-00709.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Rosethorne EM, Nahorski SR, Challiss RA. Regulation of cyclic AMP response-element binding-protein (CREB) by Gq/11-protein-coupled receptors in human SH-SY5Y neuroblastoma cells. Biochem Pharmacol. 2008;75:942–955. doi: 10.1016/j.bcp.2007.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lleó A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer's disease. Annu Rev Med. 2006;57:513–533. doi: 10.1146/annurev.med.57.121304.131442. [DOI] [PubMed] [Google Scholar]
- 24.Fornari RV, Moreira KM, Oliveira MG. Effects of the selective M1 muscarinic receptor antagonist dicyclomine on emotional memory. Learn Mem. 2000;7:287–292. doi: 10.1101/lm.34900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Soares JC, Fornari RV, Oliveira MG. Role of muscarinic M1 receptors in inhibitory avoidance and contextual fear conditioning. Neurobiol Learn Mem. 2006;86:188–196. doi: 10.1016/j.nlm.2006.02.006. [DOI] [PubMed] [Google Scholar]
- 26.Luo J, Busillo JM, Benovic JL. M3 muscarinic acetylcholine receptor-mediated signaling is regulated by distinct mechanisms. Mol Pharmacol. 2008;74:338–347. doi: 10.1124/mol.107.044750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Willets JM, Challiss RA, Nahorski SR. Endogenous G protein-coupled receptor kinase 6 Regulates M3 muscarinic acetylcholine receptor phosphorylation and desensitization in human SH-SY5Y neuroblastoma cells. J Biol Chem. 2002;277:15523–15529. doi: 10.1074/jbc.M111217200. [DOI] [PubMed] [Google Scholar]
- 28.Violin JD, Lefkowitz RJ. Beta-arrestin-biased ligands at seven-transmembrane receptors. Trends Pharmacol Sci. 2007;28:416–422. doi: 10.1016/j.tips.2007.06.006. [DOI] [PubMed] [Google Scholar]
- 29.Wisler JW, et al. A unique mechanism of beta-blocker action: carvedilol stimulates beta-arrestin signaling. Proc Natl Acad Sci USA. 2007;104:16657–16662. doi: 10.1073/pnas.0707936104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tobin AB. G-protein-coupled receptor phosphorylation: where, when and by whom. Br J Pharmacol. 2008;153(Suppl 1):S167–S176. doi: 10.1038/sj.bjp.0707662. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





