Elsevier

Neurobiology of Disease

Volume 52, April 2013, Pages 104-116
Neurobiology of Disease

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

https://doi.org/10.1016/j.nbd.2012.11.016Get rights and content

Abstract

In Huntington's disease (HD) mutant huntingtin protein impairs the function of several transcription factors, in particular the cAMP response element-binding protein (CREB). CREB activation can be increased by targeting phosphodiesterases such as phospohodiesterase 4 (PDE4) and phosphodiesterase 10A (PDE10A). Indeed, both PDE4 inhibition (DeMarch et al., 2008) and PDE10A inhibition (Giampà et al., 2010) proved beneficial in the R6/2 mouse model of HD. However, Hebb et al. (2004) reported PDE10A decline in R6/2 mice. These findings raise the issue of how PDE10A inhibition is beneficial in HD if such enzyme is lost.

R6/2 mice and their wild type littermates were treated with the PDE10A inhibitor TP10 (a gift from Pfizer) or saline, sacrificed at 5, 9, and 13 weeks of age, and single and double label immunohistochemistry and western blotting were performed. PDE10A increased dramatically in the spiny neurons of R6/2 compared to the wild type mice. Conversely, in the striatal cholinergic interneurons, PDE10A was lower and it did not change significantly with disease progression. In the other subsets of striatal interneurons (namely, parvalbuminergic, somatostatinergic, and calretininergic interneurons) PDE10A immunoreactivity was higher in the R6/2 compared to the wild-type mice. In the TP10 treated R6/2, PDE10A levels were lower than in the saline treated mice in the medium spiny neurons, whereas they were higher in all subsets of striatal interneurons except for the cholinergic ones. However, in the whole striatum densitometry studies, PDE10A immunoreactivity was lower in the R6/2 compared to the wild-type mice.

Our study demonstrates that PDE10A is increased in the spiny neurons of R6/2 mice striatum. Thus, the accumulation of PDE10A in the striatal projection neurons, by hydrolyzing greater amounts of cyclic nucleotides, is likely to contribute to cell damage in HD. Consequently, the beneficial effect of TP10 in HD models (Giampà et al., 2009, 2010) is explained by the efficiency of such compound in counteracting this phenomenon and therefore increasing the availability of cyclic nucleotides.

Highlights

► PDE10A increases in the striatal projection neurons of R6/2 mice. Such accumulation occurs in the perikarya. ► PDE10 levels decrease in the whole striatum of R6/2 animals as observed in the densitometry studies. ► Accumulation of PDE10A might cause massive hydrolization of cyclic nucleotides damaging the cell. ► PDE10 decrease in the whole striatum is possibly due to its sequestration in the perikarya of projection neurons. ► PDE10A inhibitor TP10 lowers PDE10A levels efficiently in the medium spiny neurons perikarya. ► Cholinergic neurons, resistant to HD, contain moderate amounts of PDE10A, which do not change with TP10 treatment. ► PDE10A is present in the other subsets of striatal interneurons, where it is localized in the neuronal nuclei.

Introduction

Huntington's disease (HD) is an autosomal-dominant inherited neurodegenerative disorder characterized by motor dysfunction, cognitive decline and emotional and psychiatric disturbance (Wilson et al., 1987). The genetic mutation involves the IT15 gene (The Huntington's Disease Collaborative Research Group, 1993) and is characterized by a CAG expansion beyond the normal 10–35 repeat range, resulting in formation of a mutant huntingtin protein with an expanded poly-glutamine repeat region (Albin and Tagle, 1995). Although mutant huntingtin is expressed throughout the brain, the medium spiny neurons of the striatum are particularly vulnerable to the toxicity of this protein (Albin et al., 1992). Indeed, striatal pathology is believed to underlie the motor symptoms that are typical of the disease.

The cause of striatal and cortical neuron loss in HD is under intense investigation. Wild-type huntingtin has a permissive role in the cortical synthesis of brain derived neurotrophic factor (BDNF), and its mutation impairs BDNF production and its transport to the striatum (Zuccato et al., 2001). Moreover, evidence suggests that mutant huntingtin interacts with and impairs the function of a number of other transcription factors (Sugars and Rubinsztein, 2003), in particular the cAMP response element-binding protein (CREB) (Steffan et al., 2000, Sugars and Rubinsztein, 2003, Sugars et al., 2004). CREB-mediated transcription is required for the survival of adult CNS neurons (Hardingham et al., 1998, Mantamadiotis et al., 2002) and induction of CREB signaling has a protective role in several animal models of neurodegeneration (Block et al., 2001, Walton and Dragunow, 2000, Walton et al., 1996). Inhibition of CREB-mediated gene transcription has been hypothesized to contribute to neuronal loss in HD (Jiang et al., 2003, Kazantsev et al., 1999, Nucifora et al., 2001, Steffan et al., 2000, Steffan et al., 2001) and a decreased transcription of CREB-regulated genes was observed in HD transgenic animals (Luthi-Carter et al., 2000, Nucifora et al., 2001, Wyttenbach et al., 2001). Therefore, drugs targeted at counteracting CREB loss of function may be considered as powerful tools to treat HD.

The above hypothesis was first directly addressed in our studies utilizing the cyclic nucleotide phosphodiesterase type IV (PDE4) inhibitor, rolipram. PDE4 (Houslay and Adams, 2003) is one of the eleven families of phosphodiesterases that regulates through metabolic inactivation cyclic nucleotide signaling throughout the body (Conti and Beavo, 2007), including in brain (Menniti et al., 2006). PDE4 inhibition results in activation of cAMP signaling pathways and, consequently, increased CREB phosphorylation and activation (Hosoi et al., 2003, Jacob et al., 2004, Lee et al., 2004). We have shown that rolipram reduces striatal degeneration in our rat quinolinic acid (QA) model of HD pathology (DeMarch et al., 2007) as well as in a transgenic model of HD, the R6/2 mice (DeMarch et al., 2008). These beneficial effects are hypothesized to result from the increased CREB phosphorylation, and expression of the downstream neurotrophic factor BDNF, that occurred with rolipram treatment in both disease models (DeMarch et al., 2007, DeMarch et al., 2008).

The results of our studies with rolipram provide strong theoretical support for the strategy of targeting CREB signaling to promote striatal and cortical neuron survival in HD. However, these studies do not provide a direct path to a therapy since there are no PDE4 inhibitors currently available for clinical use, as these agents cause severe side effects.

Thus, we investigated another phosphodiesterase, PDE10A, which may be a particularly promising target as a potential treatment for HD (Chappie et al., 2009, Giampà et al., 2009). PDE10A is a cAMP and cAMP-inhibited cGMP PDE that is highly expressed in regions of the brain that are innervated by dopaminergic neurons such as the striatum, nucleus accumbens and olfactory tubercle (Fujishige et al., 1999a, Fujishige et al., 1999b, Loughney et al., 1999; Soderling et al., 1999).

In particular, PDE10A is highly expressed in the striatal medium spiny neurons (Coskran et al., 2006, Seeger et al., 2003; Xie et al., 2011), which are vulnerable in HD. In the striatal neurons, PDE10A is associated primarily with membranes (Kotera et al., 2004; Xie et al., 2011).

In the striatum and accumbens, PDE10A regulates both cAMP and cGMP signaling cascades (Conti and Beavo, 2007, Siuciak et al., 2006a, Siuciak et al., 2006b). Interestingly, PDE10A is the only mammalian PDE in which the GAF domain ligand is cAMP (Gross-Langenhoff et al., 2006). GAF (cGMP-stimulated PDEs, Anabaena adenylyl cyclases and Escherichia coli transcription factor Fhla) domains are regulatory domains found in many PDEs (Beavo, 1995, Conti and Jin, 1999, Soderling and Beavo, 2000) that are thought to bind small molecules (Gross-Langenhoff et al., 2006).

Notably, inhibition of PDE10A with the highly specific inhibitor TP-10 results in a significant increase in CREB phosphorylation in striatum (Schmidt et al., 2008). A recent study published by our group showed that TP-10 treatment in our rat QA model resulted in a significant sparing of striatal neurons, on one hand, and a parallel increase in activated CREB, on the other (Giampà et al., 2009). Indeed, more recently, we have demonstrated that TP-10 treatment resulted in amelioration of HD-like neuropathology and clinical signs also in the R6/2 mouse model of HD (Giampà et al., 2010).

Thus, a decrease in PDE10A levels was proven beneficial in HD models. However, changes in PDE10A during the course of the disease are not fully understood. Hebb and coworkers reported that PDE10A mRNA and protein levels decline very early in the course of disease progression in the R6/2 mice (Hebb et al., 2004, Hu et al., 2004). These findings raise the issue of how the inhibition of PDE10A would be beneficial in a disease where such enzyme is described to be lost.

We proposed to shed light on this issue by investigating, by means of immunohistochemistry, the localization of PDE10A in the striatum of the R6/2 mouse model of HD, with particular attention at the different striatal neuronal subsets. The R6/2 mouse model (Mangiarini et al., 1996) displays a progressive neurological phenotype that mimics many of the features of HD. The age of onset in R6/2 is observed as early as four weeks but more frequently occurs between nine and eleven weeks. Age at death is between 12 and 13 weeks. This line was chosen for our studies because it recapitulates striatal pathology of HD very accurately.

Moreover, we studied changes in PDE10A levels after administration of the PDE10A inhibitor TP-10, in order to shed light on the effects of enzyme activity inhibition on its expression in striatal neurons.

Section snippets

Methods

Heterozygous transgenic R6/2 males of the CBAXC57BL/6 strain were obtained from Jackson Laboratories (Bar Harbor, ME, USA) and maintained by backcrossing carrier males with CBAXC57BL/6 _F1 females. The offspring were genotyped by polymerase chain reaction assay of DNA obtained from tail tissue. Mice were weaned and, after genotyping, treatments began at 5 weeks of age (n = 24/group) and lasted until the animals were killed at 9 and 13 weeks of age. The study groups included: R6/2 mice that were

Single label study

In the wild type animals, the single label immunohistochemical study showed PDE10A immunoreactivity in most neuronal perikarya of the caudate-putamen with a cytoplasmic localization. In the globus pallidus and in the substantia nigra pars reticulata PDE10A immunoreactive nerve fibers were observed. In the cortex, cerebellum and in the hippocampus, scattered PDE10A immunoreactive perikarya were observed (Giorgi et al., 2011).

In the R6/2 most of the striatal neurons that had the morphological

Discussion

Cyclic nucleotides play an important role in intracellular signaling. Indeed, alterations in cyclic nucleotide availability can alter neuronal cell function, and therefore precipitate or worsen motor, cognitive and psychiatric disturbances that are typical of HD. The concentration of cAMP and cGMP depends on the rate of synthesis and also on their degradation by PDEs.

PDE10A is a member of one of the newer PDE gene families. PDE10A regulates the excitability of medium spiny neurons (Siuciak et

Acknowledgement

Dr. F. S. Menniti is thanked for his help with the discussion.

References (71)

  • M. Gross-Langenhoff et al.

    cAMP is a ligand for the tandem GAF domain of human phosphodiesterase 10 and cGMP for the tandem GAF domain of phosphodiesterase 11

    J. Biol. Chem.

    (2006)
  • G.E. Hardingham et al.

    Mechanisms controlling gene expression by nuclear calcium signals

    Cell Calcium

    (1998)
  • 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)
  • M.D. Houslay et al.

    Tailoring cAMP-signalling responses through isoform multiplicity

    Trends Biochem. Sci.

    (1997)
  • Y. Kawaguchi et al.

    Striatal interneurones: chemical, physiological and morphological characterization

    Trends Neurosci.

    (1995)
  • J. Kotera et al.

    Subcellular localization of cyclic nucleotide phosphodiesterase type 10A

    J. Biol. Chem.

    (2004)
  • K. Loughney et al.

    Isolation and characterization of PDE10A, a novel human 3′, 5′-cyclic nucleotide phosphodiesterase

    Gene

    (1999)
  • 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)
  • A. Nishi et al.

    Advanced research on dopamine signaling to develop drugs for the treatment of mental disorders: biochemical and behavioral profiles of phosphodiesterase inhibition in dopaminergic neurotransmission

    J. Pharmacol. Sci.

    (2010)
  • T.F. Seeger et al.

    Immunohistochemical localization of PDE10A in the rat brain

    Brain Res.

    (2003)
  • J.A. Siuciak et al.

    Inhibition of the striatum-enriched phosphodiesterase PDE10A: a novel approach to the treatment of psychosis

    Neuropharmacology

    (2006)
  • J.A. Siuciak et al.

    Genetic deletion of the striatum-enriched phosphodiesterase PDE10A: evidence for altered striatal function

    Neuropharmacology

    (2006)
  • S.H. Soderling et al.

    Regulation of cAMP and cGMP signaling, new phosphodiesterases and new functions

    Curr. Opin. Cell Biol.

    (2000)
  • C.A. Strick et al.

    Alterations in gene regulation following inhibition of the striatum-enriched phosphodiesterase, PDE10A

    Neuropharmacology

    (2010)
  • K.L. Sugars et al.

    Transcriptional abnormalities in Huntington disease

    TIG

    (2003)
  • K.L. Sugars et al.

    Decreased cAMP response element-mediated transcription: an early event in exon 1 and full-length cell models of Huntington's disease that contributes to polyglutamine pathogenesis

    J. Biol. Chem.

    (2004)
  • Z. Sun et al.

    The differential vulnerability of striatal projection neurons in 3-nitropropionic acid-treated rats does not match that typical of adult-onset Huntington's disease

    Exp. Neurol.

    (2002)
  • Z. Sun et al.

    Differential changes in striatal projection neurons in R6/2 transgenic mice for Huntington's disease

    Neurobiol. Dis.

    (2002)
  • M.R. Walton et al.

    Is CREB a key to neuronal survival?

    TINS

    (2000)
  • M. Walton et al.

    The role of the cyclic AMP-responsive element binding protein (CREB) in hypoxic-ischemic brain damage and repair

    Brain Res. Mol. Brain Res.

    (1996)
  • R.L. Albin et al.

    Preferential loss of striato-external pallidal projection neurons in presymptomatic Huntington's disease

    Ann. Neurol.

    (1992)
  • H.S. Bateup et al.

    Cell type-specific regulation of DARPP-32 phosphorylation by psychostimulant and antipsychotic drugs

    Nat. Neurosci.

    (2008)
  • J. Beavo

    Cyclic nucleotide phosphodiesterase: functional implications of multiple isoforms

    Physiol. Rev.

    (1995)
  • F. Block et al.

    Rolipram reduces excitotoxic neuronal damage

    Neuroreport

    (2001)
  • T.A. Chappie et al.

    PDE10A inhibitors: an assessment of the current CNS drug discovery landscape

    Curr. Opin. Investig. Drugs

    (2009)
  • Cited by (46)

    • Phosphodiesterase 10A deactivation induces long-term neurological recovery, Peri-infarct remodeling and pyramidal tract plasticity after transient focal cerebral ischemia in mice

      2022, Experimental Neurology
      Citation Excerpt :

      By activating the PKA signal transduction pathway, cAMP increases phosphorylation of cAMP-response element-binding protein (CREB) and extracellular receptor kinase (ERK)-1/2, which regulate many transcription processes in neurons that control neuronal survival and plasticity (Giralt et al., 2013; Kong et al., 2019). In recent years, PDE10A has gained interest in neurodegenerative conditions including Huntington's disease (Niccolini et al., 2015), Parkinson's disease (Lee et al., 2019) and ischemic stroke (Beker et al., 2022; Birjandi et al., 2021) due to its high abundance in the striatum specifically in medium-sized spiny neurons (Leuti et al., 2013; Seeger et al., 2003). In ischemic stroke, a previous study found that besides Adora2a (adenosine receptor A2A) and Drd2 (dopamine receptor D2), Pde10a belongs to a set of genes that might decisively impede stroke recovery (Ito et al., 2018).

    • Principles and Practice of Movement Disorders

      2021, Principles and Practice of Movement Disorders
    • Alterations in cyclic nucleotide signaling are implicated in healthy aging and age-related pathologies of the brain

      2021, Vitamins and Hormones
      Citation Excerpt :

      HD mouse models have shown reduced levels of cGMP in the hippocampus, potentially due to lower sGC activity stimulated by NO (Saavedra, Giralt, Arumi, Alberch, & Perez-Navarro, 2013). PDE10A and PDE1B (Ahmad et al., 2014; Beaumont et al., 2016; Haggkvist et al., 2017; Hebb et al., 2004; Hu, McCaw, Hebb, Gomez, & Denovan-Wright, 2004; Leuti et al., 2013; Miller et al., 2014; Niewiadomska-Cimicka et al., 2017; Russell et al., 2014; Wilson et al., 2016), but not PDE5A or PDE9A (Saavedra et al., 2013), expression appears to be downregulated in HD. In fact, a recent study suggests PDE10A is the earliest imaging biomarker capable of detecting neural changes associated with HD, though these changes may be secondary to cAMP-mediated dopaminergic signaling (Fazio et al., 2020).

    • Huntingtin protein: A new option for fixing the Huntington's disease countdown clock

      2018, Neuropharmacology
      Citation Excerpt :

      Analogy comparisons with other known structures of HEAT/HEAT-like proteins (Perry and Kleckner, 2003) support these findings, which are further backed up by the extensive interaction network HTT is involved in (Shirasaki et al., 2012; Tourette et al., 2014). This network includes many cellular pathways and functions like vesicles trafficking, endocytosis, protein turnover, and cell morphology (Caviston and Holzbaur, 2009; Caviston et al., 2007; Godin et al., 2010; Harjes and Wanker, 2003; Hoffner et al., 2002; Hong et al., 2017; Ismailoglu et al., 2014; Leuti et al., 2013; Mackenzie et al., 2017; Melone et al., 2013; Nuzzo et al., 2017; Silva et al., 2017; Wanker, 2005; Zheng and Joinnides, 2009). An in-depth discussion of the HTT functions is outside the scope of this review since researchers have detailed them in previous reports (Harjes and Wanker, 2003; Jimenez-Sanchez et al., 2017; Marques Sousa and Humbert, 2013; Wanker, 2005; Zheng and Joinnides, 2009).

    View all citing articles on Scopus
    1

    These authors equally contributed to the manuscript.

    2

    Current address: Pharmacology Lab, School of Medicine, University of Western Sydney, Campbelltown Campus, Locked Bag 1797, Penrith, NSW 2751, Australia.

    3

    Current address: Department of Neuroscience, Division of Pharmacology, School of Medicine, “Federico II”, University of Naples, Building 19, Floor 17, Via Pansini, 5 80131 Napoli, Italy.

    View full text