Elsevier

Neurobiology of Aging

Volume 35, Issue 12, December 2014, Pages 2858-2869
Neurobiology of Aging

Regular article
Early decrease of type 1 cannabinoid receptor binding and phosphodiesterase 10A activity in vivo in R6/2 Huntington mice

https://doi.org/10.1016/j.neurobiolaging.2014.06.010Get rights and content

Highlights

  • We quantified brain type 1 cannabinoid (CB1) and phosphodiesterase 10A (PDE10A) alterations in Huntington disease in vivo.

  • R6/2 mice were investigated in presymptomatic, early symptomatic and late symptomatic phases.

  • CB1 and PDE10A binding decreased in basal ganglia areas upon disease progression.

  • CB1 and PDE10A binding correlated to anomalies in motor function.

  • [18F]MK-9470 and [18F]JNJ42259152 may be useful as early biomarkers of Huntington disease.

Abstract

Several lines of evidence imply early alterations in endocannabinoid and phosphodiesterase 10A (PDE10A) signaling in Huntington disease (HD). Using [18F]MK-9470 and [18F]JNJ42259152 small-animal positron emission tomography (PET), we investigated for the first time cerebral changes in type 1 cannabinoid (CB1) receptor binding and PDE10A levels in vivo in presymptomatic, early symptomatic, and late symptomatic HD (R6/2) mice, in relation to glucose metabolism ([18F]FDG PET), brain morphology (magnetic resonance imaging) and motor function. Ten R6/2 and 16 wild-type (WT) mice were investigated at 3 different time points between the age of 4 and 13 weeks. Parametric CB1 receptor and PDE10A images were anatomically standardized to Paxinos space and analyzed voxelwise. Volumetric microMRI imaging was performed to assess HD pathology. In R6/2 mice, CB1 receptor binding was decreased in comparison with WT in a cluster comprising the bilateral caudate-putamen, globus pallidus, and thalamic nucleus at week 5 (−8.1% ± 2.6%, p = 1.7 × 10−5). Longitudinal follow-up showed further progressive decline compared with controls in a cluster comprising the bilateral hippocampus, caudate-putamen, globus pallidus, superior colliculus, thalamic nucleus, and cerebellum (late vs. presymptomatic age: −13.7% ± 3.1% for R6/2 and +1.5% ± 4.0% for WT, p = 1.9 × 10−5). In R6/2 mice, PDE10A binding potential also decreased over time to reach significance at early and late symptomatic HD (late vs. presymptomatic age: −79.1% ± 1.9% for R6/2 and +2.1% ± 2.7% for WT, p = 1.5 × 10−4). The observed changes in CB1 receptor and PDE10A binding were correlated to anomalies exhibited by R6/2 animals in motor function, whereas no correlation was found with magnetic resonance imaging-based striatal volume. Our findings point to early regional dysfunctions in endocannabinoid and PDE10A signaling, involving the caudate-putamen and lateral globus pallidus, which may play a role in the progression of the disease in R6/2 animals. PET quantification of in vivo CB1 and/or PDE10A binding may thus be useful early biomarkers for HD. Our results also provide evidence of subtle motor deficits at earlier stages than previously described.

Introduction

Huntington disease (HD) is an autosomal dominant inherited neurodegenerative disease, characterized by motor dysfunctions, behavioral changes, and cognitive decline. The causative mutation is an expanded CAG repeat in exon 1 of the gene encoding the protein huntingtin (htt) (The Huntington's Disease Collaborative Research Group, 1993). Adult onset is usually observed with trinucleotide repeat blocks between 40 and 50 units, whereas more than 60 repeat results in more severe and much less frequently observed juvenile, and even infantile, forms. The most striking pathophysiological feature of HD affected brains is the progressive atrophy of the caudate nucleus and the putamen, accompanied by a secondary enlargement of the lateral ventricles, and cortical degeneration in some patients (Vonsattel et al., 1985). Despite progress in elucidating the molecular pathology of HD, therapeutic benefit for patients in terms of effective pharmacotherapy with either symptomatic or protective effects, has been scarce.

The type 1 cannabinoid (CB1) receptor and phosphodiesterase 10A (PDE10A) enzyme play a role in changed neurotransmission in HD (Hebb et al., 2004, Katona and Freund, 2008). CB1 receptors presynaptically modulate the release of other neurotransmitters (Goutopoulos and Makriyannis, 2002) and are found at high densities on GABA-ergic striatal projection neurons (Richfield and Herkenham, 1994). PDE10A hydrolyzes the important second messengers cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate and is characterized by a restricted distribution, predominantly in medium spiny neurons of the striatum (Seeger et al., 2003). In HD, loss of CB1 receptor binding and/or signaling from the basal ganglia nuclei is one of the earliest neurochemical alterations observed in humans (Allen et al., 2009, Glass et al., 2000, Richfield and Herkenham, 1994) and experimental models (Blazquez et al., 2011, Centonze et al., 2005, Denovan-Wright and Robertson, 2000, Dowie et al., 2009, Lastres-Becker et al., 2002, Lastres-Becker et al., 2004), preceding the development of cerebral metabolic deficits (Antonini et al., 1996) and striatal volume losses (Glass et al., 2000). Also, decreased levels of PDE10A expression occur before the onset of motor-related HD symptoms in transgenic HD mice (Hebb et al., 2004). In addition, genetic ablation of CB1 receptors showed to accelerate the onset of HD-like symptoms in transgenic R6/2 mice (Blazquez et al., 2011), whereas PDE10A inhibition ameliorated striatal and cortical pathology in experimental HD (Giampa et al., 2010, Giralt et al., 2013, Kleiman et al., 2011). Furthermore, pharmacologic studies demonstrated crosstalk of CB1 and PDE10A with striatal dopaminergic signaling (Chiang et al., 2013, Nishi et al., 2008). In symptomatic HD patients, we found a profound cortical and subcortical loss of CB1 receptor availability in vivo (Van Laere et al., 2010).

So far, only in vitro and ex vivo data exist on the CB1 receptor binding and PDE10A activity in genetic HD models. Ex vivo studies of CB1 receptor changes in transgenic mouse models of HD have focused on the basal ganglia, hippocampus, and motor cortex, but have not assessed other brain regions (Dowie et al., 2009, Dowie et al., 2010 ). Also it remains unknown whether CB1 receptor and PDE10A levels are changed in vivo, and to what spatial and temporal extent. Thanks to the development of selective CB1 ([18F]MK-9470; Burns et al., 2007) and PDE10A radioligands ([18F]JNJ42259152; Celen et al., 2013), in vivo imaging of these proteins became feasible.

Our primary objective was thus to investigate CB1 receptor and PDE10A changes in vivo throughout the disease process of R6/2 transgenic HD mice. As the secondary objective, motor function, brain glucose metabolism, and morphology, which are shown to be altered (early) in patients and animal models of HD (Antonini et al., 1996, Carter et al., 1999, Cowin et al., 2011, Rattray et al., 2013), were investigated in the same animals and correlated to the regional CB1 receptor binding and PDE10A levels.

Section snippets

Animals

Heterozygous male and female mice transgenic for exon 1 of the human HD gene with a greatly expanded CAG repeat (R6/2 mice, C57BL/6 background) (Mangiarini et al., 1996) and wild-type (WT) littermates were obtained from Jackson Laboratories (Bar Harbour, Maine, USA) colony, developed at the Complutense University of Madrid, Spain. In total, 7 transgenic HD (R6/2) female, 3 R6/2 male, and 10 female and 6 male WT mice were bred. All mice were housed per 3 or 4, with the same gender and genotype.

Small-animal [18F]MK-9470, [18F]JNJ42259152, and [18F]FDG PET imaging

Absolute [18F]MK-9470 binding values were not significantly different in the brain of R6/2 mice and WT littermates at the age of 5, 7, and 11 weeks. Also, absolute [18F]MK-9470 values of C57BL/6 mice were consistent in magnitude with those previously reported in Wistar (Casteels et al., 2010a, Casteels et al., 2010b) and Sprague-Dawley rats (Casteels et al., 2011). Mean images of absolute [18F]MK-9470 binding in the mouse brain of 11-week-old R6/2 mice and controls are shown in Fig. 2A. As it

Discussion

In this study, we have for the first time characterized CB1 receptor and PDE10A alterations in vivo in R6/2 transgenic mice of HD using [18F]MK-9470 and [18F]JNJ42259152 small-animal PET. We showed that in early and late stages of the symptomatic phase, both CB1 receptor binding and levels of PDE10A are decreased in basal ganglia regions.

Specifically for [18F]MK-9470, at all studied ages, CB1 receptor binding was reduced in the caudate-putamen and globus pallidus of transgenic mice, where cell

Conclusions

In vivo cerebral mapping of presymptomatic, early symptomatic, and late symptomatic mice transgenic for HD using [18F]MK-9470 and [18F]JNJ42259152 small-animal PET points to early regional dysfunctions in endocannabinoid and PDE10A signaling, incorporating the caudate-putamen. In vivo CB1 receptor and PDE10A enzyme measurements using [18F]MK-9470 and [18F]JNJ42259152 may thus be early biomarkers for HD. Translational studies in premanifest human carriers of the HD mutation using the same

Disclosure statement

The authors declare that they have no conflict of interest.

Acknowledgements

The authors acknowledge Merck & Co, Inc for the availability of the [18F]MK-9470 precursor and Janssen Research and Development for the one of [18F]JNJ42259152. They also thank Ann Van Santvoort and Julie Cornelis for their assistance in data acquisition, as well as the Leuven PET radiopharmacy team for tracer preparations. Financial support of the Fund for Scientific Research, Flanders, Belgium (FWO/G.0972.13), the KU Leuven In Vivo Molecular Imaging (IMIR) Consortium (KUL PF/10/017), and the

References (68)

  • M.J. Dowie et al.

    Behavioural and molecular consequences of chronic cannabinoid treatment in Huntington's disease transgenic mice

    Neuroscience

    (2010)
  • S. Gines et al.

    Enhanced Akt signaling is an early pro-survival response that reflects N-methyl-D-aspartate receptor activation in Huntington's disease knock-in striatal cells

    J. Biol. Chem.

    (2003)
  • M. Glass et al.

    The pattern of neurodegeneration in Huntington's disease: a comparative study of cannabinoid, dopamine, adenosine and GABA(A) receptor alterations in the human basal ganglia in Huntington's disease

    Neuroscience

    (2000)
  • A. Goutopoulos et al.

    From cannabis to cannabinergics: new therapeutic opportunities

    Pharmacol. Ther.

    (2002)
  • A.L. Hebb et al.

    Striatal phosphodiesterase mRNA and protein levels are reduced in Huntington's disease transgenic mice prior to the onset of motor symptoms

    Neuroscience

    (2004)
  • A. Leuti et al.

    Phosphodiesterase 10A (PDE10A) localization in the R6/2 mouse model of Huntington's disease

    Neurobiol. Dis.

    (2013)
  • H.G. Luesse et al.

    Evaluation of R6/2 HD transgenic mice for therapeutic studies in Huntington's disease: behavioral testing and impact of diabetes mellitus

    Behav. Brain Res.

    (2001)
  • L. Mangiarini et al.

    Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice

    Cell

    (1996)
  • G.A. Metz et al.

    Drug-induced rotation intensity in unilateral dopamine-depleted rats is not correlated with end point or qualitative measures of forelimb or hindlimb motor performance

    Neuroscience

    (2002)
  • G.A. Metz et al.

    Cortical and subcortical lesions impair skilled walking in the ladder rung walking test: a new task to evaluate fore- and hindlimb stepping, placing, and co-ordination

    J. Neurosci. Methods

    (2002)
  • G. Poisnel et al.

    Increased regional cerebral glucose uptake in an APP/PS1 model of Alzheimer's disease

    Neurobiol. Aging

    (2012)
  • C.A. Ross et al.

    Huntington's disease: from molecular pathogenesis to clinical treatment

    Lancet Neurol.

    (2011)
  • S.J. Sawiak et al.

    Voxel-based morphometry in the R6/2 transgenic mouse reveals differences between genotypes not seen with manual 2D morphometry

    Neurobiol. Dis.

    (2009)
  • T.F. Seeger et al.

    Immunohistochemical localization of PDE10A in the rat brain

    Brain Res.

    (2003)
  • K.J. Van Laere et al.

    Experimental performance assessment of SPM for SPECT neuroactivation studies using a subresolution sandwich phantom design

    Neuroimage

    (2002)
  • C.M. Wooley et al.

    Age, experience and genetic background influence treadmill walking in mice

    Physiol. Behav.

    (2009)
  • J.I. Andres et al.

    Synthesis, in vivo occupancy, and radiolabeling of potent phosphodiesterase subtype-10 inhibitors as candidates for positron emission tomography imaging

    J. Med. Chem.

    (2011)
  • A. Antonini et al.

    Striatal glucose metabolism and dopamine D2 receptor binding in asymptomatic gene carriers and patients with Huntington's disease

    Brain

    (1996)
  • C. Blazquez et al.

    Loss of striatal type 1 cannabinoid receptors is a key pathogenic factor in Huntington's disease

    Brain

    (2011)
  • H.D. Burns et al.

    [18F]MK-9470, a positron emission tomography (PET) tracer for in vivo human PET brain imaging of the cannabinoid-1 receptor

    Proc. Natl. Acad. Sci. U.S.A

    (2007)
  • R.J. Carter et al.

    Characterization of progressive motor deficits in mice transgenic for the human Huntington's disease mutation

    J. Neurosci.

    (1999)
  • C. Casteels et al.

    Preclinical evaluation and quantification of [(18)F]MK-9470 as a radioligand for PET imaging of the type 1 cannabinoid receptor in rat brain

    Eur. J. Nucl. Med. Mol. Imaging

    (2012)
  • C. Casteels et al.

    Type 1 cannabinoid receptor mapping with [(18)F]MK-9470 PET in the rat brain after quinolinic acid lesion: a comparison to dopamine receptors and glucose metabolism

    Eur. J. Nucl. Med. Mol. Imaging

    (2010)
  • C. Casteels et al.

    Construction and evaluation of quantitative small-animal PET probabilistic atlases for [(1)(8)F]FDG and [(1)(8)F]FECT functional mapping of the mouse brain

    PLoS One

    (2013)
  • Cited by (28)

    • Involvement of the Endocannabinoid System in the pathophysiology and therapeutics of movement disorders

      2022, Neurology Perspectives
      Citation Excerpt :

      In models induced by the administration of 3-NP, where there is a neuronal loss of the STR, an additional loss of CB1 receptors is observed.76 PET studies in transgenic models of HD rats and mice have also confirmed reduced binding of the CB1 receptor in the STR and GPe at all stages and alterations in the cortex, hippocampus, thalamic nuclei, or the cerebellum in more advanced stages of the disease.77 Therefore, ES activity is markedly reduced in BG in HD, which would fit with the hyperkinetic profile of this disease and with the possibility that those compounds capable of increasing the activity of this system, acting preferentially through the CB1 receptor, they could serve to alleviate some of the most prominent symptoms of the disease, such as choreic movements.78

    • The globus pallidus as a target for neuropeptides and endocannabinoids participating in central activities

      2020, Peptides
      Citation Excerpt :

      Cannabinoid CB1 receptors have been demonstrated to be involved in several neurological disorders including Huntington's disease, Parkinson's disease, schizophrenia and depression. Previous studies have revealed a significant loss of CB1 receptors in all basal ganglia regions, including the globus pallidus, in Huntington's disease [36–42]. The dramatic loss of CB1 receptors in the globus pallidus appears in the very early stages of Huntington's disease in human post-mortem tissue [37], as well as in the early symptomatic phase of Huntington disease in a transgenic rat model [43].

    • Cannabinoid pharmacology/therapeutics in chronic degenerative disorders affecting the central nervous system

      2018, Biochemical Pharmacology
      Citation Excerpt :

      Numerous changes in the endocannabinoid elements, affecting mainly CB1 and CB2 receptors and the hydrolyzing enzymes, have been described in brain samples from patients with HD and animal models. A key observation in postmortem tissues derived from patients with HD [197] and in different transgenic animal models [198–202] was the early defects in striatal CB1 receptor signaling with a progressive loss of these receptors, which was evident even before neuronal death and the occurrence of choreic symptoms. An early stimulation of the CB1 receptors located in striatal projecting neurons dampened their impairment, thus improving their capacity to inhibit the excitotoxic events that initiate the damage and to regulate growth factor generation that promotes striatal neuron survival [203].

    • High-speed video gait analysis reveals early and characteristic locomotor phenotypes in mouse models of neurodegenerative movement disorders

      2016, Behavioural Brain Research
      Citation Excerpt :

      In the R6/2 mouse model of HD ventral plane videography results have been inconclusive. Tg mice show reduced stride lengths, decreased swing speed, increased print area, and longer duration of paw contact during unforced locomotion, on the Catwalk™ system [45]. However, on a treadmill setup (Digigait™), they surprisingly do not show major gait abnormalities, even if the footprint test displays clear disturbances already at much younger ages in the same mice [46].

    View all citing articles on Scopus
    View full text