Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2018

Open Access 01-12-2018 | Research article

Modifying Rap1-signalling by targeting Pde6δ is neuroprotective in models of Alzheimer’s disease

Authors: Michael Dumbacher, Tom Van Dooren, Katrien Princen, Koen De Witte, Mélissa Farinelli, Sam Lievens, Jan Tavernier, Wim Dehaen, Stefaan Wera, Joris Winderickx, Sara Allasia, Amuri Kilonda, Stéphane Spieser, Arnaud Marchand, Patrick Chaltin, Casper C. Hoogenraad, Gerard Griffioen

Published in: Molecular Neurodegeneration | Issue 1/2018

Login to get access

Abstract

Background

Neuronal Ca2+ dyshomeostasis and hyperactivity play a central role in Alzheimer’s disease pathology and progression. Amyloid-beta together with non-genetic risk-factors of Alzheimer’s disease contributes to increased Ca2+ influx and aberrant neuronal activity, which accelerates neurodegeneration in a feed-forward fashion. As such, identifying new targets and drugs to modulate excessive Ca2+ signalling and neuronal hyperactivity, without overly suppressing them, has promising therapeutic potential.

Methods

Here we show, using biochemical, electrophysiological, imaging, and behavioural tools, that pharmacological modulation of Rap1 signalling by inhibiting its interaction with Pde6δ normalises disease associated Ca2+ aberrations and neuronal activity, conferring neuroprotection in models of Alzheimer’s disease.

Results

The newly identified inhibitors of the Rap1-Pde6δ interaction counteract AD phenotypes, by reconfiguring Rap1 signalling underlying synaptic efficacy, Ca2+ influx, and neuronal repolarisation, without adverse effects in-cellulo or in-vivo. Thus, modulation of Rap1 by Pde6δ accommodates key mechanisms underlying neuronal activity, and therefore represents a promising new drug target for early or late intervention in neurodegenerative disorders.

Conclusion

Targeting the Pde6δ-Rap1 interaction has promising therapeutic potential for disorders characterised by neuronal hyperactivity, such as Alzheimer’s disease.
Appendix
Available only for authorised users
Literature
1.
go back to reference LaFerla FM. Calcium dyshomeostasis and intracellular signalling in Alzheimer’s disease. Nat Rev Neurosci. 2002;3:862–72.CrossRef LaFerla FM. Calcium dyshomeostasis and intracellular signalling in Alzheimer’s disease. Nat Rev Neurosci. 2002;3:862–72.CrossRef
2.
go back to reference Bookheimer SY, et al. Patterns of brain activation in people at risk for Alzheimer’s disease. N Engl J Med. 2000;343:450–6.CrossRef Bookheimer SY, et al. Patterns of brain activation in people at risk for Alzheimer’s disease. N Engl J Med. 2000;343:450–6.CrossRef
3.
go back to reference Elman J a, et al. Neural compensation in older people with brain amyloid-β deposition. Nat Neurosci. 2014;17:1316–8.CrossRef Elman J a, et al. Neural compensation in older people with brain amyloid-β deposition. Nat Neurosci. 2014;17:1316–8.CrossRef
4.
go back to reference Sperling RA, et al. Amyloid deposition is associated with impaired default network function in older persons without dementia. Neuron. 2009;63:178–88.CrossRef Sperling RA, et al. Amyloid deposition is associated with impaired default network function in older persons without dementia. Neuron. 2009;63:178–88.CrossRef
5.
go back to reference Busche MA, et al. Clusters of hyperactive neurons near amyloid plaques in a mouse model of Alzheimer’s disease. Science. 2008;321:1686–9.CrossRef Busche MA, et al. Clusters of hyperactive neurons near amyloid plaques in a mouse model of Alzheimer’s disease. Science. 2008;321:1686–9.CrossRef
6.
go back to reference Bezprozvanny I, Mattson MP. Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease. Trends Neurosci. 2008;31:454–63.CrossRef Bezprozvanny I, Mattson MP. Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease. Trends Neurosci. 2008;31:454–63.CrossRef
7.
go back to reference Morris M, Maeda S, Vossel K, Mucke L. The many faces of tau. Neuron. 2011;70:410–26.CrossRef Morris M, Maeda S, Vossel K, Mucke L. The many faces of tau. Neuron. 2011;70:410–26.CrossRef
8.
go back to reference Holth JK, et al. Tau loss attenuates neuronal network Hyperexcitability in mouse and Drosophila genetic models of epilepsy. J Neurosci. 2013;33:1651–9.CrossRef Holth JK, et al. Tau loss attenuates neuronal network Hyperexcitability in mouse and Drosophila genetic models of epilepsy. J Neurosci. 2013;33:1651–9.CrossRef
9.
go back to reference Ittner LM, Götz J. Amyloid-β and tau--a toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci. 2011;12:65–72.CrossRef Ittner LM, Götz J. Amyloid-β and tau--a toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci. 2011;12:65–72.CrossRef
10.
go back to reference Hall AM, et al. Tau-dependent Kv4.2 depletion and dendritic hyperexcitability in a mouse model of Alzheimer’s disease. J Neurosci. 2015;35:6221–30.CrossRef Hall AM, et al. Tau-dependent Kv4.2 depletion and dendritic hyperexcitability in a mouse model of Alzheimer’s disease. J Neurosci. 2015;35:6221–30.CrossRef
11.
go back to reference Roberson ED, et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science. 2007;316:750–4.CrossRef Roberson ED, et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science. 2007;316:750–4.CrossRef
12.
go back to reference Zempel H, Thies E, Mandelkow E, Mandelkow E-M. Abeta oligomers cause localized ca(2+) elevation, missorting of endogenous tau into dendrites, tau phosphorylation, and destruction of microtubules and spines. J Neurosci. 2010;30:11938–50.CrossRef Zempel H, Thies E, Mandelkow E, Mandelkow E-M. Abeta oligomers cause localized ca(2+) elevation, missorting of endogenous tau into dendrites, tau phosphorylation, and destruction of microtubules and spines. J Neurosci. 2010;30:11938–50.CrossRef
13.
go back to reference Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat. Rev. Mol. Cell Biol. 2007;8:101–12.CrossRef Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat. Rev. Mol. Cell Biol. 2007;8:101–12.CrossRef
14.
go back to reference Spires-Jones TL, Hyman BT. The intersection of amyloid beta and tau at synapses in Alzheimer’s disease. Neuron. 2014;82:756–71.CrossRef Spires-Jones TL, Hyman BT. The intersection of amyloid beta and tau at synapses in Alzheimer’s disease. Neuron. 2014;82:756–71.CrossRef
15.
go back to reference Van Dooren, T., Princen, K., De Witte, K. & Griffioen, G. Derailed intraneuronal signalling drives pathogenesis in sporadic and familial Alzheimer’s disease. Biomed Res Int (2014) 2014, 167024. Van Dooren, T., Princen, K., De Witte, K. & Griffioen, G. Derailed intraneuronal signalling drives pathogenesis in sporadic and familial Alzheimer’s disease. Biomed Res Int (2014) 2014, 167024.
16.
go back to reference Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256:184–5.CrossRef Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256:184–5.CrossRef
17.
go back to reference Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science. 2002;297:353–6.CrossRef Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science. 2002;297:353–6.CrossRef
18.
go back to reference Vossel KA, et al. Seizures and epileptiform activity in the early stages of Alzheimer disease. JAMA Neurol. 2013;70:1158–66.CrossRef Vossel KA, et al. Seizures and epileptiform activity in the early stages of Alzheimer disease. JAMA Neurol. 2013;70:1158–66.CrossRef
19.
go back to reference Sanchez PE, et al. Levetiracetam suppresses neuronal network dysfunction and reverses synaptic and cognitive deficits in an Alzheimer’s disease model. Proc Natl Acad Sci. 2012;109:E2895–903.CrossRef Sanchez PE, et al. Levetiracetam suppresses neuronal network dysfunction and reverses synaptic and cognitive deficits in an Alzheimer’s disease model. Proc Natl Acad Sci. 2012;109:E2895–903.CrossRef
20.
go back to reference Vossel KA, Tartaglia MC, Nygaard HB, Zeman AZ, Miller BL. Epileptic activity in Alzheimer’s disease: causes and clinical relevance. Lancet Neurol. 2017;16:311–22.CrossRef Vossel KA, Tartaglia MC, Nygaard HB, Zeman AZ, Miller BL. Epileptic activity in Alzheimer’s disease: causes and clinical relevance. Lancet Neurol. 2017;16:311–22.CrossRef
21.
go back to reference Moechars D, et al. Early phenotypic changes in transgenic mice that overexpress different mutants of amyloid precursor protein in brain. J Biol Chem. 1999;274:6483–92.CrossRef Moechars D, et al. Early phenotypic changes in transgenic mice that overexpress different mutants of amyloid precursor protein in brain. J Biol Chem. 1999;274:6483–92.CrossRef
22.
go back to reference Dewachter I, et al. Aging increased amyloid peptide and caused amyloid plaques in brain of old APP/V717I transgenic mice by a different mechanism than mutant presenilin1. J Neurosci. 2000;20:6452–8.CrossRef Dewachter I, et al. Aging increased amyloid peptide and caused amyloid plaques in brain of old APP/V717I transgenic mice by a different mechanism than mutant presenilin1. J Neurosci. 2000;20:6452–8.CrossRef
23.
go back to reference Everett KL, Cooper DMF. An improved targeted cAMP sensor to study the regulation of adenylyl cyclase 8 by Ca2+ entry through voltage-gated channels. PLoS One. 2013;8:1–11. Everett KL, Cooper DMF. An improved targeted cAMP sensor to study the regulation of adenylyl cyclase 8 by Ca2+ entry through voltage-gated channels. PLoS One. 2013;8:1–11.
24.
go back to reference Schrödinger LLC. In: LLC S, editor. Small-molecule drug discovery suite 2016–2. New York; 2016. Schrödinger LLC. In: LLC S, editor. Small-molecule drug discovery suite 2016–2. New York; 2016.
25.
go back to reference Chandra A, et al. The GDI-like solubilizing factor PDEδ sustains the spatial organization and signalling of Ras family proteins. Nat Cell Biol. 2012;14:148–58.CrossRef Chandra A, et al. The GDI-like solubilizing factor PDEδ sustains the spatial organization and signalling of Ras family proteins. Nat Cell Biol. 2012;14:148–58.CrossRef
26.
go back to reference Risseeuw MDP, et al. A “clickable” MTX reagent as a practical tool for profiling small-molecule-intracellular target interactions via MASPIT. ChemMedChem. 2013;8:521–6.CrossRef Risseeuw MDP, et al. A “clickable” MTX reagent as a practical tool for profiling small-molecule-intracellular target interactions via MASPIT. ChemMedChem. 2013;8:521–6.CrossRef
27.
go back to reference Rezwan M, Auerbach D. Yeast “N”-hybrid systems for protein–protein and drug–protein interaction discovery. Methods. 2012;57:423–9.CrossRef Rezwan M, Auerbach D. Yeast “N”-hybrid systems for protein–protein and drug–protein interaction discovery. Methods. 2012;57:423–9.CrossRef
28.
go back to reference Jämsä A, Hasslund K, Cowburn RF, Bäckström A, Vasänge M. The retinoic acid and brain-derived neurotrophic factor differentiated SH-SY5Y cell line as a model for Alzheimer’s disease-like tau phosphorylation. Biochem Biophys Res Commun. 2004;319:993–1000.CrossRef Jämsä A, Hasslund K, Cowburn RF, Bäckström A, Vasänge M. The retinoic acid and brain-derived neurotrophic factor differentiated SH-SY5Y cell line as a model for Alzheimer’s disease-like tau phosphorylation. Biochem Biophys Res Commun. 2004;319:993–1000.CrossRef
29.
go back to reference Anekonda TS, et al. L-type voltage-gated calcium channel blockade with isradipine as a therapeutic strategy for Alzheimer’s disease. Neurobiol Dis. 2011;41:62–70.CrossRef Anekonda TS, et al. L-type voltage-gated calcium channel blockade with isradipine as a therapeutic strategy for Alzheimer’s disease. Neurobiol Dis. 2011;41:62–70.CrossRef
30.
go back to reference Grewal SS, et al. Neuronal calcium activates a Rap1 and B-Raf signaling pathway via the cyclic adenosine monophosphate-dependent protein kinase. J Biol Chem. 2000;275:3722–8.CrossRef Grewal SS, et al. Neuronal calcium activates a Rap1 and B-Raf signaling pathway via the cyclic adenosine monophosphate-dependent protein kinase. J Biol Chem. 2000;275:3722–8.CrossRef
31.
go back to reference Pei J-J, et al. Up-regulation of mitogen-activated protein kinases ERK1/2 and MEK1/2 is associated with the progression of neurofibrillary degeneration in Alzheimer’s disease. Mol Brain Res. 2002;109:45–55.CrossRef Pei J-J, et al. Up-regulation of mitogen-activated protein kinases ERK1/2 and MEK1/2 is associated with the progression of neurofibrillary degeneration in Alzheimer’s disease. Mol Brain Res. 2002;109:45–55.CrossRef
32.
go back to reference Marambaud P, Dreses-Werringloer U, Vingtdeux V. Calcium signaling in neurodegeneration. Mol Neurodegener. 2009;4:20.CrossRef Marambaud P, Dreses-Werringloer U, Vingtdeux V. Calcium signaling in neurodegeneration. Mol Neurodegener. 2009;4:20.CrossRef
33.
go back to reference Hanzal-Bayer M, Renault L, Roversi P, Wittinghofer A, Hillig RC. The complex of Arl2-GTP and PDE delta: from structure to function. EMBO J. 2002;21:2095–106.CrossRef Hanzal-Bayer M, Renault L, Roversi P, Wittinghofer A, Hillig RC. The complex of Arl2-GTP and PDE delta: from structure to function. EMBO J. 2002;21:2095–106.CrossRef
34.
go back to reference Hancock JF. Ras proteins: different signals from different locations. Nat Rev Mol Cell Biol. 2003;4:373–85.CrossRef Hancock JF. Ras proteins: different signals from different locations. Nat Rev Mol Cell Biol. 2003;4:373–85.CrossRef
35.
go back to reference Nancy V, et al. The delta subunit of retinal rod cGMP phosphodiesterase regulates the membrane association of Ras and rap GTPases. J Biol Chem. 2002;277:15076–84.CrossRef Nancy V, et al. The delta subunit of retinal rod cGMP phosphodiesterase regulates the membrane association of Ras and rap GTPases. J Biol Chem. 2002;277:15076–84.CrossRef
36.
go back to reference Glatter T, Wepf A, Aebersold R, Gstaiger M. An integrated workflow for charting the human interaction proteome: insights into the PP2A system. Mol Syst Biol. 2009;5(237). Glatter T, Wepf A, Aebersold R, Gstaiger M. An integrated workflow for charting the human interaction proteome: insights into the PP2A system. Mol Syst Biol. 2009;5(237).
37.
go back to reference Ismail S a, et al. Arl2-GTP and Arl3-GTP regulate a GDI-like transport system for farnesylated cargo. Nat Chem Biol. 2011;7:942–9.CrossRef Ismail S a, et al. Arl2-GTP and Arl3-GTP regulate a GDI-like transport system for farnesylated cargo. Nat Chem Biol. 2011;7:942–9.CrossRef
38.
go back to reference Wätzlich D, et al. The interplay between RPGR, PDEδ and Arl2/3 regulate the ciliary targeting of farnesylated cargo. EMBO Rep. 2013;14:465–72.CrossRef Wätzlich D, et al. The interplay between RPGR, PDEδ and Arl2/3 regulate the ciliary targeting of farnesylated cargo. EMBO Rep. 2013;14:465–72.CrossRef
39.
go back to reference Baehr W. Membrane protein transport in photoreceptors: the function of PDEδ: the Proctor lecture. Invest Ophthalmol Vis Sci. 2014;55:8653–66.CrossRef Baehr W. Membrane protein transport in photoreceptors: the function of PDEδ: the Proctor lecture. Invest Ophthalmol Vis Sci. 2014;55:8653–66.CrossRef
40.
go back to reference Zimmermann G, et al. Small molecule inhibition of the KRAS–PDEδ interaction impairs oncogenic KRAS signalling. Nature. 2013;497:638–42.CrossRef Zimmermann G, et al. Small molecule inhibition of the KRAS–PDEδ interaction impairs oncogenic KRAS signalling. Nature. 2013;497:638–42.CrossRef
41.
go back to reference Schmick M, et al. KRas localizes to the plasma membrane by spatial cycles of Solubilization, trapping and vesicular transport. Cell. 2014;157:459–71.CrossRef Schmick M, et al. KRas localizes to the plasma membrane by spatial cycles of Solubilization, trapping and vesicular transport. Cell. 2014;157:459–71.CrossRef
42.
go back to reference York RD, et al. Rap1 mediates sustained MAP kinase activation induced by nerve growth factor. Nature. 1998;392:622–6.CrossRef York RD, et al. Rap1 mediates sustained MAP kinase activation induced by nerve growth factor. Nature. 1998;392:622–6.CrossRef
43.
go back to reference Mochizuki N, et al. Spatio-temporal images of growth-factor-induced activation of Ras and Rap1. Nature. 2001;411:1065–8.CrossRef Mochizuki N, et al. Spatio-temporal images of growth-factor-induced activation of Ras and Rap1. Nature. 2001;411:1065–8.CrossRef
44.
go back to reference Wu GY, Deisseroth K, Tsien RW. Spaced stimuli stabilize MAPK pathway activation and its effects on dendritic morphology. Nat Neurosci. 2001;4:151–8.CrossRef Wu GY, Deisseroth K, Tsien RW. Spaced stimuli stabilize MAPK pathway activation and its effects on dendritic morphology. Nat Neurosci. 2001;4:151–8.CrossRef
45.
go back to reference Ohba Y. Mechanism of the spatio-temporal regulation of Ras and Rap1. EMBO J. 2003;22:859–69.CrossRef Ohba Y. Mechanism of the spatio-temporal regulation of Ras and Rap1. EMBO J. 2003;22:859–69.CrossRef
46.
go back to reference Wang Z, et al. Rap1-Mediated Activation of Extracellular Signal-Regulated Kinases by Cyclic AMP Is Dependent on the Mode of Rap1 Activation Rap1-Mediated Activation of Extracellular Signal-Regulated Kinases by Cyclic AMP Is Dependent on the Mode of Rap1 Activation. Mol Cell Biol. 2006;26:2130–45.CrossRef Wang Z, et al. Rap1-Mediated Activation of Extracellular Signal-Regulated Kinases by Cyclic AMP Is Dependent on the Mode of Rap1 Activation Rap1-Mediated Activation of Extracellular Signal-Regulated Kinases by Cyclic AMP Is Dependent on the Mode of Rap1 Activation. Mol Cell Biol. 2006;26:2130–45.CrossRef
47.
go back to reference de Rooij J, et al. Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature. 1998;396:474–7.CrossRef de Rooij J, et al. Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature. 1998;396:474–7.CrossRef
48.
go back to reference Kim J, Wei D, Hoffman DA. Kv4 potassium channel subunits control action potential repolarization and frequency-dependent broadening in rat hippocampal CA1 pyramidal neurones. J Physiol. 2005;569:41–57.CrossRef Kim J, Wei D, Hoffman DA. Kv4 potassium channel subunits control action potential repolarization and frequency-dependent broadening in rat hippocampal CA1 pyramidal neurones. J Physiol. 2005;569:41–57.CrossRef
49.
go back to reference Morozov A, et al. Rap1 couples cAMP signaling to a distinct pool of p42/44MAPK regulating excitability, synaptic plasticity, learning, and memory. Neuron. 2003;39:309–25.CrossRef Morozov A, et al. Rap1 couples cAMP signaling to a distinct pool of p42/44MAPK regulating excitability, synaptic plasticity, learning, and memory. Neuron. 2003;39:309–25.CrossRef
50.
go back to reference Purgert C a, et al. Intracellular mGluR5 can mediate synaptic plasticity in the Hippocampus. J Neurosci. 2014;34:4589–98.CrossRef Purgert C a, et al. Intracellular mGluR5 can mediate synaptic plasticity in the Hippocampus. J Neurosci. 2014;34:4589–98.CrossRef
51.
go back to reference Gladding CM, Fitzjohn SM, Molnár E, Molnar E. Metabotropic glutamate receptor-mediated long-term depression: molecular mechanisms. Pharmacol Rev. 2009;61:395–412.CrossRef Gladding CM, Fitzjohn SM, Molnár E, Molnar E. Metabotropic glutamate receptor-mediated long-term depression: molecular mechanisms. Pharmacol Rev. 2009;61:395–412.CrossRef
52.
go back to reference Gallagher SM. Extracellular signal-regulated protein kinase activation is required for metabotropic glutamate receptor-dependent long-term depression in hippocampal area CA1. J Neurosci. 2004;24:4859–64.CrossRef Gallagher SM. Extracellular signal-regulated protein kinase activation is required for metabotropic glutamate receptor-dependent long-term depression in hippocampal area CA1. J Neurosci. 2004;24:4859–64.CrossRef
53.
go back to reference Jong Y-JI, Sergin I, Purgert C, O’Malley KL. Location-dependent signaling of the group 1 metabotropic glutamate receptor mGlu5. Mol Pharmacol. 2014;86:774–85.CrossRef Jong Y-JI, Sergin I, Purgert C, O’Malley KL. Location-dependent signaling of the group 1 metabotropic glutamate receptor mGlu5. Mol Pharmacol. 2014;86:774–85.CrossRef
54.
go back to reference Yuan L, Adams JP, Swank M, Sweatt JD, Johnston D. Protein kinase modulation of dendritic K+ channels in hippocampus involves a mitogen-activated protein kinase pathway. J Neurosci. 2002;22:4860–8.CrossRef Yuan L, Adams JP, Swank M, Sweatt JD, Johnston D. Protein kinase modulation of dendritic K+ channels in hippocampus involves a mitogen-activated protein kinase pathway. J Neurosci. 2002;22:4860–8.CrossRef
55.
go back to reference Scala F, et al. Intraneuronal Aβ accumulation induces hippocampal neuron hyperexcitability through A-type K+ current inhibition mediated by activation of caspases and GSK-3. Neurobiol Aging. 2015;36:886–900.CrossRef Scala F, et al. Intraneuronal Aβ accumulation induces hippocampal neuron hyperexcitability through A-type K+ current inhibition mediated by activation of caspases and GSK-3. Neurobiol Aging. 2015;36:886–900.CrossRef
56.
go back to reference Ping Y, et al. Linking Abeta42-induced Hyperexcitability to neurodegeneration, learning and motor deficits, and a shorter lifespan in an Alzheimer’s model. PLoS Genet. 2015;11:1–25.CrossRef Ping Y, et al. Linking Abeta42-induced Hyperexcitability to neurodegeneration, learning and motor deficits, and a shorter lifespan in an Alzheimer’s model. PLoS Genet. 2015;11:1–25.CrossRef
57.
go back to reference Snyder EM, et al. Internalization of ionotropic glutamate receptors in response to mGluR activation. Nat Neurosci. 2001;4:1079–85.CrossRef Snyder EM, et al. Internalization of ionotropic glutamate receptors in response to mGluR activation. Nat Neurosci. 2001;4:1079–85.CrossRef
58.
go back to reference Subramanian J, Dye L, Morozov A. Rap1 signaling prevents L-type calcium channel-dependent neurotransmitter release. J Neurosci. 2013;33:7245–52.CrossRef Subramanian J, Dye L, Morozov A. Rap1 signaling prevents L-type calcium channel-dependent neurotransmitter release. J Neurosci. 2013;33:7245–52.CrossRef
59.
go back to reference Catterall WA. Voltage-Gated Calcium Channels. Cold Spring Harb. Perspect. Biol. 3; 2011. p. a003947. Catterall WA. Voltage-Gated Calcium Channels. Cold Spring Harb. Perspect. Biol. 3; 2011. p. a003947.
60.
go back to reference Sawada M, Sato M. The effect of dimethyl sulfoxide on the neuronal excitability and cholinergic transmission in Aplysia ganglion cells. Ann N Y Acad Sci. 1975;243:337–57.CrossRef Sawada M, Sato M. The effect of dimethyl sulfoxide on the neuronal excitability and cholinergic transmission in Aplysia ganglion cells. Ann N Y Acad Sci. 1975;243:337–57.CrossRef
61.
go back to reference Morley P, Whitfield JF. The differentiation inducer, dimethyl sulfoxide, transiently increases the intracellular calcium ion concentration in various cell types. J Cell Physiol. 1993;156:219–25.CrossRef Morley P, Whitfield JF. The differentiation inducer, dimethyl sulfoxide, transiently increases the intracellular calcium ion concentration in various cell types. J Cell Physiol. 1993;156:219–25.CrossRef
62.
go back to reference Terwel D, et al. Changed conformation of mutant tau-P301L underlies the moribund Tauopathy, absent in progressive, nonlethal Axonopathy of tau-4R/2N transgenic mice. J Biol Chem. 2005;280:3963–73.CrossRef Terwel D, et al. Changed conformation of mutant tau-P301L underlies the moribund Tauopathy, absent in progressive, nonlethal Axonopathy of tau-4R/2N transgenic mice. J Biol Chem. 2005;280:3963–73.CrossRef
63.
go back to reference Zhang B, et al. Retarded axonal transport of R406W mutant tau in transgenic mice with a neurodegenerative tauopathy. J Neurosci. 2004;24:4657–67.CrossRef Zhang B, et al. Retarded axonal transport of R406W mutant tau in transgenic mice with a neurodegenerative tauopathy. J Neurosci. 2004;24:4657–67.CrossRef
64.
go back to reference Zhang H, Constantine R, Frederick JM, Baehr W. The prenyl-binding protein PrBP/δ: a chaperone participating in intracellular trafficking. Vis Res. 2012;75:19–25.CrossRef Zhang H, Constantine R, Frederick JM, Baehr W. The prenyl-binding protein PrBP/δ: a chaperone participating in intracellular trafficking. Vis Res. 2012;75:19–25.CrossRef
65.
go back to reference Bero AW, et al. Neuronal activity regulates the regional vulnerability to amyloid-β deposition. Nat Neurosci. 2011;14:750–6.CrossRef Bero AW, et al. Neuronal activity regulates the regional vulnerability to amyloid-β deposition. Nat Neurosci. 2011;14:750–6.CrossRef
66.
go back to reference Palop JJ, et al. Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer’s disease. Neuron. 2007;55:697–711.CrossRef Palop JJ, et al. Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer’s disease. Neuron. 2007;55:697–711.CrossRef
67.
go back to reference Meldrum BS, Rogawski MA. Molecular targets for antiepileptic drug development. Neurotherapeutics. 2007;4:18–61.CrossRef Meldrum BS, Rogawski MA. Molecular targets for antiepileptic drug development. Neurotherapeutics. 2007;4:18–61.CrossRef
68.
go back to reference Bernard C, et al. Acquired dendritic channelopathy in temporal lobe epilepsy. Science. 2004;305:532–5.CrossRef Bernard C, et al. Acquired dendritic channelopathy in temporal lobe epilepsy. Science. 2004;305:532–5.CrossRef
69.
go back to reference Lugo JN, et al. Altered phosphorylation and localization of the A-type channel, Kv4.2 in status epilepticus. J Neurochem. 2008;106:1929–40.CrossRef Lugo JN, et al. Altered phosphorylation and localization of the A-type channel, Kv4.2 in status epilepticus. J Neurochem. 2008;106:1929–40.CrossRef
70.
go back to reference Zhang Z, et al. GABAB receptor promotes its own surface expression by recruiting a Rap1-dependent signaling cascade. J Cell Sci. 2015;128:2302–13.CrossRef Zhang Z, et al. GABAB receptor promotes its own surface expression by recruiting a Rap1-dependent signaling cascade. J Cell Sci. 2015;128:2302–13.CrossRef
71.
go back to reference Cagnol S, Chambard J-C. ERK and cell death: mechanisms of ERK-induced cell death--apoptosis, autophagy and senescence. FEBS J. 2010;277:2–21.CrossRef Cagnol S, Chambard J-C. ERK and cell death: mechanisms of ERK-induced cell death--apoptosis, autophagy and senescence. FEBS J. 2010;277:2–21.CrossRef
72.
go back to reference Chong YH, et al. ERK1/2 activation mediates Abeta oligomer-induced neurotoxicity via caspase-3 activation and tau cleavage in rat organotypic hippocampal slice cultures. J Biol Chem. 2006;281:20315–25.CrossRef Chong YH, et al. ERK1/2 activation mediates Abeta oligomer-induced neurotoxicity via caspase-3 activation and tau cleavage in rat organotypic hippocampal slice cultures. J Biol Chem. 2006;281:20315–25.CrossRef
73.
go back to reference Satoh T, et al. Neuroprotection by MAPK / ERK kinase inhibition with U0126 against oxidative stress in a mouse neuronal cell line and rat primary cultured cortical neurons. Neurosci Lett. 2000;288:163–6.CrossRef Satoh T, et al. Neuroprotection by MAPK / ERK kinase inhibition with U0126 against oxidative stress in a mouse neuronal cell line and rat primary cultured cortical neurons. Neurosci Lett. 2000;288:163–6.CrossRef
74.
go back to reference Amadoro G, et al. NMDA receptor mediates tau-induced neurotoxicity by calpain and ERK/MAPK activation. Proc Natl Acad Sci U S A. 2006;103:2892–7.CrossRef Amadoro G, et al. NMDA receptor mediates tau-induced neurotoxicity by calpain and ERK/MAPK activation. Proc Natl Acad Sci U S A. 2006;103:2892–7.CrossRef
75.
go back to reference Feld M, et al. Decrease of ERK/MAPK overactivation in prefrontal cortex reverses early memory deficit in a mouse model of Alzheimer’s disease. J Alzheimers Dis. 2014;40:69–82.CrossRef Feld M, et al. Decrease of ERK/MAPK overactivation in prefrontal cortex reverses early memory deficit in a mouse model of Alzheimer’s disease. J Alzheimers Dis. 2014;40:69–82.CrossRef
76.
go back to reference Yau KW, et al. Microtubule minus-end binding protein CAMSAP2 controls axon specification and dendrite development. Neuron. 2014;82:1058–73.CrossRef Yau KW, et al. Microtubule minus-end binding protein CAMSAP2 controls axon specification and dendrite development. Neuron. 2014;82:1058–73.CrossRef
77.
go back to reference Klein WL. Abeta toxicity in Alzheimer’s disease: globular oligomers (ADDLs) as new vaccine and drug targets. Neurochem Int. 2002;41:345–52.CrossRef Klein WL. Abeta toxicity in Alzheimer’s disease: globular oligomers (ADDLs) as new vaccine and drug targets. Neurochem Int. 2002;41:345–52.CrossRef
Metadata
Title
Modifying Rap1-signalling by targeting Pde6δ is neuroprotective in models of Alzheimer’s disease
Authors
Michael Dumbacher
Tom Van Dooren
Katrien Princen
Koen De Witte
Mélissa Farinelli
Sam Lievens
Jan Tavernier
Wim Dehaen
Stefaan Wera
Joris Winderickx
Sara Allasia
Amuri Kilonda
Stéphane Spieser
Arnaud Marchand
Patrick Chaltin
Casper C. Hoogenraad
Gerard Griffioen
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2018
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-018-0283-3

Other articles of this Issue 1/2018

Molecular Neurodegeneration 1/2018 Go to the issue