Multiple Behavior Phenotypes of the Fragile-X Syndrome Mouse Model Respond to Chronic Inhibition of Phosphodiesterase-4D (PDE4D)

Mark E Gurney, Patricia Cogram, Robert M Deacon, Christopher Rex, Michael Tranfaglia, Mark E Gurney, Patricia Cogram, Robert M Deacon, Christopher Rex, Michael Tranfaglia

Abstract

Fragile-X syndrome (FXS) patients display intellectual disability and autism spectrum disorder due to silencing of the X-linked, fragile-X mental retardation-1 (FMR1) gene. Dysregulation of cAMP metabolism is a consistent finding in patients and in the mouse and fly FXS models. We therefore explored if BPN14770, a prototypic phosphodiesterase-4D negative allosteric modulator (PDE4D-NAM) in early human clinical trials, might provide therapeutic benefit in the mouse FXS model. Daily treatment of adult male fmr1 C57Bl6 knock-out mice with BPN14770 for 14 days reduced hyperarousal, improved social interaction, and improved natural behaviors such as nesting and marble burying as well as dendritic spine morphology. There was no decrement in behavioral scores in control C57Bl6 treated with BPN14770. The behavioral benefit of BPN14770 persisted two weeks after washout of the drug. Thus, BPN14770 may be useful for the treatment of fragile-X syndrome and other disorders with decreased cAMP signaling.

Conflict of interest statement

M.E.G is an employee of Tetra. Tetra owns BPN14770. All other authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Schematic Diagram Depicting Treatment Groups and Schedule of Events. Treatment groups were wild-type C57Bl6 mice exposed to vehicle (WT-Vehicle), wild-type C57Bl6 mice exposed to BPN14770 (WT-BPN14770), fmr1 KO mice exposed to vehicle, or fmr1 KO mice exposed to BPN14770. Three experiments were performed. In each experiment, mice were treated daily for 14 days by oral gavage with either vehicle or BPN14770. In the first experiment (Expt 1), mice were profiled for open field activity, social interaction, and natural behaviors (nesting & marble burying) on the 14th day of treatment. In the second experiment (Expt 2), mice were treated daily for 14 days, profiled for open field activity on the 14th day of treatment, then killed and prepared for spine morphometry. In the third experiment (Expt 3), mice were treated daily for 14 days, treatment was then stopped, and 14 days after cessation of treatment the mice were profiled for open field activity, social interaction, nesting & marble burying. Each treatment group contained 10 adult male mice.
Figure 2
Figure 2
BPN14770 Improved Behavioral Phenotypes of fmr1 KO Mice (A) Wild-type (WT) or fmr1 KO (KO) adult male mice were treated with vehicle or BPN14770 daily for 14 days (Expt 1) and then profiled for behavior. From left-to-right, data are shown for hyperarousal (Number of Squares crossed in the open field test), social interaction (Duration of Sniffing), nesting (Nesting Score), and marble burying (Marbles Buried). (B) Mice were treated daily for 14 days with vehicle or BPN14770 and then drug was withdrawn for 14 days (Expt 3). Mice were profiled for hyperarousal, social interaction, nesting and marble burying on the 14th day after drug withdrawal. Data were analyzed by Two-way ANOVA followed by Tukey’s post-test corrected for multiple comparisons. P-values shown are: n.s. not significant, *p < 0.05, **p < 0.01, ***p < 0.001 (N = 10 mice per group).
Figure 3
Figure 3
Representative Laser Scanning Confocal Micrographs Of Dendritic Segments And Dendritic Spines For Pyramidal Neurons In Medial Prefrontal Cortex. Dendritic spine morphometry was performed on dendritic segments of layer 3 and layer 5 pyramidal cells in medial prefrontal cortex (schematic). The sampling region for layer 3 pyramidal cells (L3P) was a segment of the primary apical dendrite extending from the soma to a distance of 100–150 µM. Layer 5 pyramidal cells were sampled from the soma to a distance of 200–250 µM of the primary apical dendrite and from the branch point of the apical tuft for a distance of 50 µM. High magnification insets for each segment sampled show the presence of thin-necked, immature spines (arrow) as well as mushroom (arrowhead only) and stubby spine (double arrowhead) types.
Figure 4
Figure 4
Dendritic Spine Multi-Dimensional Feature Distribution Plots. Multi-dimensional spine length x head diameter plots are shown for (A) the apical dendrite of Layer 3 pyramidal cells (N = 3,502 spines, 70 cells vehicle; N = 3,322 spines, 69 cells treated), (B) the apical dendrite of Layer 5 pyramidal cells (N = 3,792 spines, 70 cells vehicle; N = 3,443 spines, 69 cells treated), and (C) the apical dendrite tufts of Layer 5 pyramidal cells (N = 2,626 spines, 65 cells vehicle; N = 3,026 spines, 68 cells treated). The first two panels compare spines from fmr1 KO mice exposed to vehicle or BPN14770. The third panel shows the difference plot. The difference map (vehicle subtracted from BPN14770) shows maturation (shortening) of spines on the apical dendrites of layer 3 pyramidal neurons. There was no change in dendritic spine maturation on layer 5 pyramidal neurons.
Figure 5
Figure 5
Effect of BPN14770 on Dendritic Spine Length. The distribution of spine length is shown for the apical dendrite of Layer 3 pyramidal cells (L3P apical), the apical dendrite of Layer 5 pyramidal cells (L5P apical), and the apical dendrite tufts of Layer 5 pyramidal cells (L5P tufts). Dendritic spines were sampled for length, head dimeter and neck width. Data are plotted for vehicle (red circles) and BPN14770-treated (blue squares) fmr1 KO mice. Data were analyzed by the 2-sample Komolgorov-Smirnov test. P-values shown are ***p < 0.0001.

References

    1. Verkerk AJ, et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell. 1991;65:905–914. doi: 10.1016/0092-8674(91)90397-H.
    1. Hagerman RJ, et al. Advances in the treatment of fragile X syndrome. Pediatrics. 2009;123:378–390. doi: 10.1542/peds.2008-0317.
    1. Lozano R, Azarang A, Wilaisakditipakorn T, Hagerman RJ. Fragile X syndrome: A review of clinical management. Intractable Rare Dis Res. 2016;5:145–157. doi: 10.5582/irdr.2016.01048.
    1. Laggerbauer B, Ostareck D, Keidel EM, Ostareck-Lederer A, Fischer U. Evidence that fragile X mental retardation protein is a negative regulator of translation. Hum Mol Genet. 2001;10:329–338. doi: 10.1093/hmg/10.4.329.
    1. Darnell JC, et al. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell. 2011;146:247–261. doi: 10.1016/j.cell.2011.06.013.
    1. Fmr1 knockout mice: a model to study fragile X mental retardation. The Dutch-Belgian Fragile X Consortium. Cell. 1994;78:23–33.
    1. Morales J, et al. Drosophila fragile X protein, DFXR, regulates neuronal morphology and function in the brain. Neuron. 2002;34:961–972. doi: 10.1016/S0896-6273(02)00731-6.
    1. Choi CH, et al. PDE-4 inhibition rescues aberrant synaptic plasticity in Drosophila and mouse models of fragile X syndrome. J Neurosci. 2015;35:396–408. doi: 10.1523/JNEUROSCI.1356-12.2015.
    1. Kanellopoulos AK, Semelidou O, Kotini AG, Anezaki M, Skoulakis EM. Learning and memory deficits consequent to reduction of the fragile X mental retardation protein result from metabotropic glutamate receptor-mediated inhibition of cAMP signaling in Drosophila. J Neurosci. 2012;32:13111–13124. doi: 10.1523/JNEUROSCI.1347-12.2012.
    1. Berry-Kravis E, Sklena P. Demonstration of abnormal cyclic AMP production in platelets from patients with fragile X syndrome. American Journal of Medical Genetics. 1993;45:81–87. doi: 10.1002/ajmg.1320450120.
    1. Berry-Kravis E, Hicar M, Ciurlionis R. Reduced cyclic AMP production in fragile X syndrome: cytogenetic and molecular correlations. Pediatric Research. 1995;38:638–643. doi: 10.1203/00006450-199511000-00002.
    1. Zhang YQ, et al. Drosophila fragile X-related gene regulates the MAP1B homolog Futsch to control synaptic structure and function. Cell. 2001;107:591–603. doi: 10.1016/S0092-8674(01)00589-X.
    1. Dockendorff TC, et al. Drosophila lacking dfmr1 activity show defects in circadian output and fail to maintain courtship interest. Neuron. 2002;34:973–984. doi: 10.1016/S0896-6273(02)00724-9.
    1. McBride SM, et al. Pharmacological rescue of synaptic plasticity, courtship behavior, and mushroom body defects in a Drosophila model of fragile X syndrome. Neuron. 2005;45:753–764. doi: 10.1016/j.neuron.2005.01.038.
    1. Bolduc FV, Bell K, Cox H, Broadie KS, Tully T. Excess protein synthesis in Drosophila fragile X mutants impairs long-term memory. Nature Neuroscience. 2008;11:1143–1145. doi: 10.1038/nn.2175.
    1. Houslay MD, Schafer P, Zhang KY. Keynote review: phosphodiesterase-4 as a therapeutic target. Drug Discov Today. 2005;10:1503–1519. doi: 10.1016/S1359-6446(05)03622-6.
    1. Gurney, M. E. et al. Heteroaryl Inhibitors of PDE4. United States Patent US 9,221,843 B2 (2015).
    1. Hoffmann R, Wilkinson IR, McCallum JF, Engels P, Houslay MD. cAMP-specific phosphodiesterase HSPDE4D3 mutants which mimic activation and changes in rolipram inhibition triggered by protein kinase A phosphorylation of Ser-54: generation of a molecular model. Biochem J. 1998;333(Pt 1):139–149. doi: 10.1042/bj3330139.
    1. Sette C, Conti M. Phosphorylation and activation of a cAMP-specific phosphodiesterase by the cAMP-dependent protein kinase. Involvement of serine 54 in the enzyme activation. J Biol Chem. 1996;271:16526–16534. doi: 10.1074/jbc.271.28.16526.
    1. Linglart A, et al. PRKAR1A and PDE4D mutations cause acrodysostosis but two distinct syndromes with or without GPCR-signaling hormone resistance. The Journal of Clinical Endocrinology and Metabolism. 2012;97:E2328–2338. doi: 10.1210/jc.2012-2326.
    1. Butler MG, Rames LJ, Wadlington WB. Acrodysostosis: report of a 13-year-old boy with review of literature and metacarpophalangeal pattern profile analysis. American Journal of Medical Genetics. 1988;30:971–980. doi: 10.1002/ajmg.1320300416.
    1. Lee H, et al. Exome sequencing identifies PDE4D mutations in acrodysostosis. American Journal of Human Genetics. 2012;90:746–751. doi: 10.1016/j.ajhg.2012.03.004.
    1. Michot C, et al. Exome sequencing identifies PDE4D mutations as another cause of acrodysostosis. American Journal of Human Genetics. 2012;90:740–745. doi: 10.1016/j.ajhg.2012.03.003.
    1. Lynch DC, et al. Identification of novel mutations confirms Pde4d as a major gene causing acrodysostosis. Human Mutation. 2013;34:97–102. doi: 10.1002/humu.22222.
    1. Lindstrand A, et al. Different mutations in PDE4D associated with developmental disorders with mirror phenotypes. Journal of Medical Genetics. 2014;51:45–54. doi: 10.1136/jmedgenet-2013-101937.
    1. Kaname T, et al. Heterozygous mutations in cyclic AMP phosphodiesterase-4D (PDE4D) and protein kinase A (PKA) provide new insights into the molecular pathology of acrodysostosis. Cellular Signalling. 2014;26:2446–2459. doi: 10.1016/j.cellsig.2014.07.025.
    1. Gurney ME, D’Amato EC, Burgin AB. Phosphodiesterase-4 (PDE4) Molecular Pharmacology and Alzheimer’s Disease. Neurotherapeutics: The Journal of the American Society for Experimental NeuroTherapeutics. 2015;12:49–56. doi: 10.1007/s13311-014-0309-7.
    1. Gervasi N, Tchenio P, Preat T. PKA dynamics in a Drosophila learning center: coincidence detection by rutabaga adenylyl cyclase and spatial regulation by dunce phosphodiesterase. Neuron. 2010;65:516–529. doi: 10.1016/j.neuron.2010.01.014.
    1. Burgin AB, et al. Design of Phosphodiesterase Type 4D (PDE4D) Allosteric Modulators for Cognition with Improved Safety. Nature Biotechnology. 2010;28:63–70. doi: 10.1038/nbt.1598.
    1. Comery TA, et al. Abnormal dendritic spines in fragile X knockout mice: maturation and pruning deficits. Proc Natl Acad Sci USA. 1997;94:5401–5404. doi: 10.1073/pnas.94.10.5401.
    1. Cruz-Martin A, Crespo M, Portera-Cailliau C. Delayed stabilization of dendritic spines in fragile X mice. J Neurosci. 2010;30:7793–7803. doi: 10.1523/JNEUROSCI.0577-10.2010.
    1. Thomas AM, et al. Genetic reduction of group 1 metabotropic glutamate receptors alters select behaviors in a mouse model for fragile X syndrome. Behav Brain Res. 2011;223:310–321. doi: 10.1016/j.bbr.2011.04.049.
    1. Deacon RM, Croucher A, Rawlins JN. Hippocampal cytotoxic lesion effects on species-typical behaviours in mice. Behav Brain Res. 2002;132:203–213. doi: 10.1016/S0166-4328(01)00401-6.
    1. He CX, Portera-Cailliau C. The trouble with spines in fragile X syndrome: density, maturity and plasticity. Neuroscience. 2013;251:120–128. doi: 10.1016/j.neuroscience.2012.03.049.
    1. Irwin SA, Galvez R, Greenough WT. Dendritic spine structural anomalies in fragile-X mental retardation syndrome. Cereb Cortex. 2000;10:1038–1044. doi: 10.1093/cercor/10.10.1038.
    1. Cherry JA, Davis RL. Cyclic AMP phosphodiesterases are localized in regions of the mouse brain associated with reinforcement, movement, and affect. The Journal of Comparative Neurology. 1999;407:287–301. doi: 10.1002/(SICI)1096-9861(19990503)407:2<287::AID-CNE9>;2-R.
    1. Bobon D, et al. Is phosphodiesterase inhibition a new mechanism of antidepressant action? A double blind double-dummy study between rolipram and desipramine in hospitalized major and/or endogenous depressives. Eur Arch Psychiatry Neurol Sci. 1988;238:2–6. doi: 10.1007/BF00381071.
    1. Bertolino A, et al. Rolipram versus imipramine in inpatients with major, “minor” or atypical depressive disorder: a double-blind double-dummy study aimed at testing a novel therapeutic approach. Int Clin Psychopharmacol. 1988;3:245–253. doi: 10.1097/00004850-198807000-00006.
    1. Pecknold JC, McClure DJ, Appeltauer L, Wrzesinski L, Allan T. Treatment of anxiety using fenobam (a nonbenzodiazepine) in a double-blind standard (diazepam) placebo-controlled study. Journal of Clinical Psychopharmacology. 1982;2:129–133. doi: 10.1097/00004714-198204000-00010.
    1. Berry-Kravis EM, et al. Effects of STX209 (arbaclofen) on neurobehavioral function in children and adults with fragile X syndrome: a randomized, controlled, phase 2 trial. Science Translational Medicine. 2012;4:152ra127. doi: 10.1126/scitranslmed.3004214.
    1. Berry-Kravis E, Huttenlocher PR. Cyclic AMP metabolism in fragile X syndrome. Annals of Neurology. 1992;31:22–26. doi: 10.1002/ana.410310105.
    1. Gresele P, Momi S, Falcinelli E. Anti-platelet therapy: phosphodiesterase inhibitors. Br J Clin Pharmacol. 2011;72:634–646. doi: 10.1111/j.1365-2125.2011.04034.x.
    1. Ito M, et al. Characterization of the isoenzymes of cyclic nucleotide phosphodiesterase in human platelets and the effects of E4021. Cellular Signalling. 1996;8:575–581. doi: 10.1016/S0898-6568(96)00112-X.
    1. Richter W, Jin SL, Conti M. Splice variants of the cyclic nucleotide phosphodiesterase PDE4D are differentially expressed and regulated in rat tissue. Biochem J. 2005;388:803–811. doi: 10.1042/BJ20050030.
    1. Kelly MP, et al. Select 3′,5′-cyclic nucleotide phosphodiesterases exhibit altered expression in the aged rodent brain. Cellular Signalling. 2014;26:383–397. doi: 10.1016/j.cellsig.2013.10.007.
    1. Michalon A, et al. Chronic pharmacological mGlu5 inhibition corrects fragile X in adult mice. Neuron. 2012;74:49–56. doi: 10.1016/j.neuron.2012.03.009.
    1. Yan QJ, Rammal M, Tranfaglia M, Bauchwitz RP. Suppression of two major Fragile X Syndrome mouse model phenotypes by the mGluR5 antagonist MPEP. Neuropharmacology. 2005;49:1053–1066. doi: 10.1016/j.neuropharm.2005.06.004.
    1. Pacey LK, Tharmalingam S, Hampson DR. Subchronic administration and combination metabotropic glutamate and GABAB receptor drug therapy in fragile X syndrome. J Pharmacol Exp Ther. 2011;338:897–905. doi: 10.1124/jpet.111.183327.
    1. Kelley DJ, et al. The cyclic AMP cascade is altered in the fragile X nervous system. PLoS One. 2007;2:e931. doi: 10.1371/journal.pone.0000931.
    1. Dlaboga D, Hajjhussein H, O’Donnell JM. Regulation of phosphodiesterase-4 (PDE4) expression in mouse brain by repeated antidepressant treatment: comparison with rolipram. Brain Res. 2006;1096:104–112. doi: 10.1016/j.brainres.2006.04.032.
    1. Fujita, M. et al. cAMP signaling in brain is decreased in unmedicated depressed patients and increased by treatment with a selective serotonin reuptake inhibitor. Molecular Psychiatry (2016).
    1. Bailey CH, Bartsch D, Kandel ER. Toward a molecular definition of long-term memory storage. Proc Natl Acad Sci USA. 1996;93:13445–13452. doi: 10.1073/pnas.93.24.13445.
    1. Kandel ER. The biology of memory: a forty-year perspective. J Neurosci. 2009;29:12748–12756. doi: 10.1523/JNEUROSCI.3958-09.2009.
    1. Tully T, Bourtchouladze R, Scott R, Tallman J. Targeting the CREB pathway for memory enhancers. Nat Rev Drug Discov. 2003;2:267–277. doi: 10.1038/nrd1061.
    1. Brown JA, Gianino SM, Gutmann DH. Defective cAMP generation underlies the sensitivity of CNS neurons to neurofibromatosis-1 heterozygosity. J Neurosci. 2010;30:5579–5589. doi: 10.1523/JNEUROSCI.3994-09.2010.
    1. Bourtchouladze R, et al. A mouse model of Rubinstein-Taybi syndrome: defective long-term memory is ameliorated by inhibitors of phosphodiesterase 4. Proc Natl Acad Sci USA. 2003;100:10518–10522. doi: 10.1073/pnas.1834280100.
    1. Bu Q, et al. CREB Signaling Is Involved in Rett Syndrome Pathogenesis. J Neurosci. 2017;37:3671–3685. doi: 10.1523/JNEUROSCI.3735-16.2017.

Source: PubMed

3
Abonneren