Elsevier

Neuropharmacology

Volume 44, Issue 5, April 2003, Pages 653-661
Neuropharmacology

Regional brain uptake of the muscarinic ligand, [18F]FP-TZTP, is greatly decreased in M2 receptor knockout mice but not in M1, M3 and M4 receptor knockout mice

https://doi.org/10.1016/S0028-3908(03)00050-9Get rights and content

Abstract

A muscarinic receptor radioligand, 3-(3-(3-fluoropropyl)thio) -1,2,5,thiadiazol-4-yl)-1,2,5,6-tetrahydro-1-methylpyridine (fP-TZTP) radiolabeled with the positron emitting radionuclide 18F ([18F]FP-TZTP) displayed regional brain distribution consistent with M2 receptor densities in rat brain. The purpose of the present study is to further elucidate the subtype selectivity of [18F]FP-TZTP using genetically engineered mice which lacked functional M1, M2, M3, or M4 muscarinic receptors. Using ex vivo autoradiography, the regional brain localization of [18F]FP-TZTP in M2 knockout (M2 KO) was significantly decreased (51.3 to 61.4%; P<0.01) when compared to the wild-type (WT) mice in amygdala, brain stem, caudate putamen, cerebellum, cortex, hippocampus, hypothalamus, superior colliculus, and thalamus. In similar studies with M1KO, M3KO and M4KO compared to their WT mice, [18F]FP-TZTP uptakes in the same brain regions were not significantly decreased at P<0.01. However, in amygdala and hippocampus small decreases of 19.5% and 22.7%, respectively, were observed for M1KO vs WT mice at P<0.05. Given the fact that large decreases in [18F]FP-TZTP brain uptakes were seen only in M2 KO vs. WT mice, we conclude that [18F]FP-TZTP preferentially labels M2 receptors in vivo.

Introduction

Our goal is to develop receptor-specific ligands labeled with a positron emitting radionuclide in order to follow biochemical changes as a function of disease by external imaging using positron emission tomography (PET). Postmortem quantitation of muscarinic receptor subtypes in Alzheimer’s patients indicate a selective loss of M2 subtype in cortical regions while the M1 subtype was preserved (Aubert et al., 1992a, Quirion et al., 1989, Rodriguez-Puertas et al., 1997). Therefore, we sought to develop an M2 selective ligand labeled with a positron-emitting radionuclide since that would be most sensitive to M2 receptor subtype density changes in the living human brain. This, in turn, would allow the study of the progression of Alzheimer’s dementia, early diagnosis, and monitoring of drug therapies using non-invasive external imaging.

We have developed a radioligand that is a fluorinated analog of a compound that showed M2 selectivity in biological assays (Sauerberg et al., 1992). This compound, 3-(3-(3-fluoropropyl)thio) -1,2,5,thiadiazol-4-yl)-1,2,5,6-tetrahydro-1-methyl-pyridine (FP-TZTP) has been radiolabeled with F-18, a positron emitting radionuclide with a half life of 109.7 min (Kiesewetter et al., 1995). Ex vivo autoradiography of rat brain using no carrier added [18F]FP-TZTP confirmed a distribution of radioactivity in gray matter which was characteristic of M2 receptor density (Kiesewetter et al., 1999). This distribution of radioactivity is in good agreement with the distribution as described in immunocytochemical localization studies of M2 receptors (Levey, 1993). Intravenous co-injection of P-TZTP [3-(3-(propyl)thio)-1,2,5,thiadiazol-4-yl)-1,2,5,6-tetrahydro-1-methyl-pyridine] at 5, 50 and 500 nmol per rat inhibited [18F]FP-TZTP uptake in a dose-dependent manner in all gray matter regions which contain M2 receptors. Rats co-injected with 50 nmol P-TZTP and [18F]FP-TZTP showed 55% inhibition of uptake in the heart when sacrificed 5 min after injection compared to the uptake in the heart of [18F]FP-TZTP alone. The brain distribution of the agonist [18F]FP-TZTP was only weakly inhibited by coinjection of antagonists such as RS FMeQNB (Kiesewetter et al., 1995). At present, there are no subtype selective muscarinic agonists with high enough affinity to compete with the high affinity radioligand, [18F]FP-TZTP. The inability of known antagonists to block agonist binding may be a result of different binding characteristics and pharmacokinetics (Herschberg et al., 1995).

Although these data were consistent with FP-TZTP binding to the M2 receptor subtype, the degree to which other muscarinic receptor subtypes are labeled by FP-TZTP remains uncertain. Unfortunately, no highly subtype-selective muscarinic agonists or antagonists that freely cross the blood–brain barrier have been identified. As a result, selective pharmacological blockade of specific muscarinic receptor subtypes in vivo is not possible at present. However, the recent development of mutant mouse strains in which specific muscarinic receptor genes are inactivated by gene targeting techniques has offered new perspectives for studying the selectivity of muscarinic agonists and antagonists in vivo (Gomeza et al., 1999a, Gomeza et al., 1999b, Hamilton et al., 1997). In the present study, we have used homozygous M1−/− (M1 KO), M2−/− (M2 KO), M3−/− (M3 KO) and M4−/− (M4 KO) receptor mutant mice (Fisahn et al., 2002, Gomeza et al., 1999a, Gomeza et al., 1999b, Miyakawa et al., 2001, Yamada et al., 2001) to further examine the receptor subtype selectivity of the radiotracer, [18F]FP-TZTP, in vivo. Immunoprecipitation studies with receptor subtype-selective antibodies showed that the lack of an individual muscarinic receptor subtype had no significant effect on the expression levels of the remaining muscarinic receptor subtypes (Fisahn et al., 2002, Gomeza et al., 1999a, Gomeza et al., 1999b, Miyakawa et al., 2001, Yamada et al., 2001).

The goal of this study is to use mice that are deficient in muscarinic subtypes to show unequivocally that the distribution of [18F]FP-TZTP in vivo is controlled by the M2 subtype. If [18F]FP-TZTP can be validated as a probe of the M2 receptor using muscarinic receptor subtype KO mice, then this probe will be the first available to study a specific muscarinic subtype as a function of treatment in such diseases as Alzheimer’s disease by external imaging.

Section snippets

Radiochemical synthesis

A radiochemical synthesis for [18F]FP-TZTP has been previously published (Kiesewetter et al., 1995); however, for the present study a modified synthesis that requires only one radiochemical step was used. The radiochemical yield with this synthesis was 27.3±4.6% (n=69) and 39.5±6.5% (n=100), in a time of 58±4 min with a specific activity of 4377±2011 Ci/mmol at EOB for a 100 batches.

Solutions of K222 (4.5 mg/100 μL acetonitrile) and substrate [3-methanesulfonyloxypropyl-TZTP methanesulfonate

Kinetics and metabolism of [18F]FP-PTZTP in M1, M2, M3, and M4 receptor knockout and wild-type control mice

The regional brain uptake of [18F]FP-TZTP was determined at 5, 15, 30 and 60 min in 129 SvEv mice (Fig. 1). After 60 min the radioactivity was decreased 78 to 85% in all the brain regions examined when compared to 5 min while after 30 min only 48% to 60% of the radioactivity had cleared from the brain regions. In these studies a sample of blood and brain were taken at each point and extracted in acetonitrile to determine the percentage of parent present by TLC analysis (Table 1). The

Discussion

The major finding is that [18F]FP-TZTP binding is reduced in the M2 KO mice, but not in M1, M3 and M4 KO mice compared to their respective WT mice at a significance level of P<0.01. In vitro studies with mouse brain and heart membrane homogenates showed significant reduction of [3H]QNB binding in M2 KO compared to WT mice in heart, olfactory bulb, cerebellum, cortex, and brain stem regions, whereas a significant reduction in binding of [3H]QNB was not observed in the hippocampus and striatum (

Acknowledgements

Part of this work was supported financially through a CRADA between the NIDDK and the Eli Lilly Research Laboratories. We thank Ms. J. Gan for expert technical assistance and acknowledge Dr. Alokesh Duttaroy for advice and support. We acknowledge the cyclotron group in the NIH Positron Emission Tomography Department for isotope production. We thank Josie Divel of the Positron Emission Tomography Department’s Quality Control section for specific activity determinations of [18F]FP-TZTP.

References (20)

There are more references available in the full text version of this article.

Cited by (34)

  • Unexpected decrease in in vivo binding of [<sup>3</sup>H]QNB in the mouse cerebral cortex in the developing brain - A comparison with [<sup>11</sup>C]NMPB

    2018, Nuclear Medicine and Biology
    Citation Excerpt :

    Bakker et al. developed 123I-iododexetimide as a M1 selective SPECT ligand [30]. [ 18F]FP-TZTPI was evaluated as a M2 selective PET probe in subtype knockout mouse [31]. These subtype-selective ligands might be of value to verify the validity of the above hypothesis.

  • Imaging of the muscarinic acetylcholine neuroreceptor in rats with the M2 selective agonist [ <sup>18</sup>F]FP-TZTP

    2012, Nuclear Medicine and Biology
    Citation Excerpt :

    The slices are ca. 6–8 mm from the tip of the brain, and the positioning of the animal was identical whether imaged in vivo or ex vivo. [18F]FP-TZTP is a radiotracer that has in vivo affinity for the M2 muscarinic subtype receptor [5]. To calculate the kinetics of this tracer in rats, we performed in vivo and ex vivo scans using the ATLAS small animal PET after the animals received a bolus injection of [18F]FP-TZTP.

  • Decreased muscarinic receptor binding in the frontal cortex of bipolar disorder and major depressive disorder subjects

    2009, Journal of Affective Disorders
    Citation Excerpt :

    PET studies have reported decreased binding of [18F]FP-TZTP, a CHRM2 selective agonist, within the anterior cingulate cortex of individuals with bipolar disorder, however, no change in [18F]FP-TZTP binding was found in individuals with major depressive disorder (Cannon et al., 2006). As [18F]FP-TZTP binds predominantly to the high affinity binding state of CHRM2 (Jagoda et al., 2003), the apparent decrease in the functional receptors binding sites may represent elevated intrasynaptic acetylcholine concentrations and/or a shift of the CHRM2 pool into the low affinity binding state, possibly in response to elevated acetylcholine levels. Thus, the decreased expression of CHRM2 in bipolar and major depressive disorder and CHRM3 in bipolar disorder within discrete regions of the frontal cortex suggested by our data may reflect a regionally specific down regulation of CHRM2 and CHRM3 in response to elevated acetylcholine associated with affective disorders.

  • 18F-Labeled PET-Tracers for Cardiological Imaging

    2008, Fluorine and Health: Molecular Imaging, Biomedical Materials and Pharmaceuticals
View all citing articles on Scopus
View full text