Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2016

Open Access 01-12-2016 | Research article

Alzheimer’s disease-like APP processing in wild-type mice identifies synaptic defects as initial steps of disease progression

Authors: Mickael Audrain, Romain Fol, Patrick Dutar, Brigitte Potier, Jean-Marie Billard, Julien Flament, Sandro Alves, Marie-Anne Burlot, Gaelle Dufayet-Chaffaud, Alexis-Pierre Bemelmans, Julien Valette, Philippe Hantraye, Nicole Déglon, Nathalie Cartier, Jérome Braudeau

Published in: Molecular Neurodegeneration | Issue 1/2016

Login to get access

Abstract

Background

Alzheimer’s disease (AD) is the most frequent form of dementia in the elderly and no effective treatment is currently available. The mechanisms triggering AD onset and progression are still imperfectly dissected. We aimed at deciphering the modifications occurring in vivo during the very early stages of AD, before the development of amyloid deposits, neurofibrillary tangles, neuronal death and inflammation. Most current AD models based on Amyloid Precursor Protein (APP) overproduction beginning from in utero, to rapidly reproduce the histological and behavioral features of the disease within a few months, are not appropriate to study the early steps of AD development. As a means to mimic in vivo amyloid APP processing closer to the human situation in AD, we used an adeno-associated virus (AAV)-based transfer of human mutant APP and Presenilin 1 (PS1) genes to the hippocampi of two-month-old C57Bl/6 J mice to express human APP, without significant overexpression and to specifically induce its amyloid processing.

Results

The human APP, βCTF and Aβ42/40 ratio were similar to those in hippocampal tissues from AD patients. Three months after injection the murine Tau protein was hyperphosphorylated and rapid synaptic failure occurred characterized by decreased levels of both PSD-95 and metabolites related to neuromodulation, on proton magnetic resonance spectroscopy (1H-MRS). Astrocytic GLT-1 transporter levels were lower and the tonic glutamatergic current was stronger on electrophysiological recordings of CA1 hippocampal region, revealing the overstimulation of extrasynaptic N-methyl D-aspartate receptor (NMDAR) which precedes the loss of long-term potentiation (LTP). These modifications were associated with early behavioral impairments in the Open-field, Y-maze and Morris Mater Maze tasks.

Conclusions

Altogether, this demonstrates that an AD-like APP processing, yielding to levels of APP, βCTF and Aβ42/Aβ40 ratio similar to those observed in AD patients, are sufficient to rapidly trigger early steps of the amyloidogenic and Tau pathways in vivo. With this strategy, we identified a sequence of early events likely to account for disease onset and described a model that may facilitate efforts to decipher the factors triggering AD and to evaluate early neuroprotective strategies.
Appendix
Available only for authorised users
Literature
1.
go back to reference Selkoe DJ. Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev. 2001;81(2):741–66.PubMed Selkoe DJ. Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev. 2001;81(2):741–66.PubMed
2.
go back to reference Hardy J, Allsop D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol Sci. 1991;12(10):383–8.PubMedCrossRef Hardy J, Allsop D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol Sci. 1991;12(10):383–8.PubMedCrossRef
5.
go back to reference Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, Bjornsson S, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;488(7409):96–9. doi:10.1038/nature11283.PubMedCrossRef Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, Bjornsson S, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;488(7409):96–9. doi:10.​1038/​nature11283.PubMedCrossRef
6.
7.
go back to reference Doyle E, Bruce MT, Breen KC, Smith DC, Anderton B, Regan CM. Intraventricular infusions of antibodies to amyloid-beta-protein precursor impair the acquisition of a passive avoidance response in the rat. Neurosci Lett. 1990;115(1):97–102.PubMedCrossRef Doyle E, Bruce MT, Breen KC, Smith DC, Anderton B, Regan CM. Intraventricular infusions of antibodies to amyloid-beta-protein precursor impair the acquisition of a passive avoidance response in the rat. Neurosci Lett. 1990;115(1):97–102.PubMedCrossRef
8.
go back to reference Huber G, Martin JR, Loffler J, Moreau JL. Involvement of amyloid precursor protein in memory formation in the rat: an indirect antibody approach. Brain Res. 1993;603(2):348–52.PubMedCrossRef Huber G, Martin JR, Loffler J, Moreau JL. Involvement of amyloid precursor protein in memory formation in the rat: an indirect antibody approach. Brain Res. 1993;603(2):348–52.PubMedCrossRef
9.
go back to reference Weyer SW, Zagrebelsky M, Herrmann U, Hick M, Ganss L, Gobbert J, et al. Comparative analysis of single and combined APP/APLP knockouts reveals reduced spine density in APP-KO mice that is prevented by APPsalpha expression. Acta Neuropathol Commun. 2014;2:36. doi:10.1186/2051-5960-2-36.PubMedPubMedCentralCrossRef Weyer SW, Zagrebelsky M, Herrmann U, Hick M, Ganss L, Gobbert J, et al. Comparative analysis of single and combined APP/APLP knockouts reveals reduced spine density in APP-KO mice that is prevented by APPsalpha expression. Acta Neuropathol Commun. 2014;2:36. doi:10.​1186/​2051-5960-2-36.PubMedPubMedCentralCrossRef
10.
go back to reference Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, et al. Correlative memory deficits, abeta elevation, and amyloid plaques in transgenic mice. Science. 1996;274(5284):99–102.PubMedCrossRef Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, et al. Correlative memory deficits, abeta elevation, and amyloid plaques in transgenic mice. Science. 1996;274(5284):99–102.PubMedCrossRef
11.
go back to reference Moran PM, Higgins LS, Cordell B, Moser PC. Age-related learning deficits in transgenic mice expressing the 751-amino acid isoform of human beta-amyloid precursor protein. Proc Natl Acad Sci U S A. 1995;92(12):5341–5.PubMedPubMedCentralCrossRef Moran PM, Higgins LS, Cordell B, Moser PC. Age-related learning deficits in transgenic mice expressing the 751-amino acid isoform of human beta-amyloid precursor protein. Proc Natl Acad Sci U S A. 1995;92(12):5341–5.PubMedPubMedCentralCrossRef
12.
13.
go back to reference DaRocha-Souto B, Scotton TC, Coma M, Serrano-Pozo A, Hashimoto T, Sereno L, et al. Brain oligomeric beta-amyloid but not total amyloid plaque burden correlates with neuronal loss and astrocyte inflammatory response in amyloid precursor protein/tau transgenic mice. J Neuropathol Exp Neurol. 2011;70(5):360–76. doi:10.1097/NEN.0b013e318217a118.PubMedPubMedCentralCrossRef DaRocha-Souto B, Scotton TC, Coma M, Serrano-Pozo A, Hashimoto T, Sereno L, et al. Brain oligomeric beta-amyloid but not total amyloid plaque burden correlates with neuronal loss and astrocyte inflammatory response in amyloid precursor protein/tau transgenic mice. J Neuropathol Exp Neurol. 2011;70(5):360–76. doi:10.​1097/​NEN.​0b013e318217a118​.PubMedPubMedCentralCrossRef
14.
go back to reference Cheng IH, Scearce-Levie K, Legleiter J, Palop JJ, Gerstein H, Bien-Ly N, et al. Accelerating amyloid-beta fibrillization reduces oligomer levels and functional deficits in Alzheimer disease mouse models. J Biol Chem. 2007;282(33):23818–28. doi:10.1074/jbc.M701078200.PubMedCrossRef Cheng IH, Scearce-Levie K, Legleiter J, Palop JJ, Gerstein H, Bien-Ly N, et al. Accelerating amyloid-beta fibrillization reduces oligomer levels and functional deficits in Alzheimer disease mouse models. J Biol Chem. 2007;282(33):23818–28. doi:10.​1074/​jbc.​M701078200.PubMedCrossRef
15.
go back to reference Tomiyama T, Nagata T, Shimada H, Teraoka R, Fukushima A, Kanemitsu H, et al. A new amyloid beta variant favoring oligomerization in Alzheimer’s-type dementia. Ann Neurol. 2008;63(3):377–87. doi:10.1002/ana.21321.PubMedCrossRef Tomiyama T, Nagata T, Shimada H, Teraoka R, Fukushima A, Kanemitsu H, et al. A new amyloid beta variant favoring oligomerization in Alzheimer’s-type dementia. Ann Neurol. 2008;63(3):377–87. doi:10.​1002/​ana.​21321.PubMedCrossRef
16.
18.
go back to reference Gibson PH. Form and distribution of senile plaques seen in silver impregnated sections in the brains of intellectually normal elderly people and people with Alzheimer-type dementia. Neuropathol Appl Neurobiol. 1983;9(5):379–89.PubMedCrossRef Gibson PH. Form and distribution of senile plaques seen in silver impregnated sections in the brains of intellectually normal elderly people and people with Alzheimer-type dementia. Neuropathol Appl Neurobiol. 1983;9(5):379–89.PubMedCrossRef
19.
go back to reference Mackenzie IR, McLachlan RS, Kubu CS, Miller LA. Prospective neuropsychological assessment of nondemented patients with biopsy proven senile plaques. Neurology. 1996;46(2):425–9.PubMedCrossRef Mackenzie IR, McLachlan RS, Kubu CS, Miller LA. Prospective neuropsychological assessment of nondemented patients with biopsy proven senile plaques. Neurology. 1996;46(2):425–9.PubMedCrossRef
21.
go back to reference Klein WL, Krafft GA, Finch CE. Targeting small abeta oligomers: the solution to an Alzheimer’s disease conundrum? Trends Neurosci. 2001;24(4):219–24.PubMedCrossRef Klein WL, Krafft GA, Finch CE. Targeting small abeta oligomers: the solution to an Alzheimer’s disease conundrum? Trends Neurosci. 2001;24(4):219–24.PubMedCrossRef
24.
go back to reference During MJ, Young D, Baer K, Lawlor P, Klugmann M. Development and optimization of adeno-associated virus vector transfer into the central nervous system. Methods Mol Med. 2003;76:221–36.PubMed During MJ, Young D, Baer K, Lawlor P, Klugmann M. Development and optimization of adeno-associated virus vector transfer into the central nervous system. Methods Mol Med. 2003;76:221–36.PubMed
25.
go back to reference Tenenbaum L, Chtarto A, Lehtonen E, Velu T, Brotchi J, Levivier M. Recombinant AAV-mediated gene delivery to the central nervous system. J Gene Med. 2004;6 Suppl 1:S212–22. doi:10.1002/jgm.506.PubMedCrossRef Tenenbaum L, Chtarto A, Lehtonen E, Velu T, Brotchi J, Levivier M. Recombinant AAV-mediated gene delivery to the central nervous system. J Gene Med. 2004;6 Suppl 1:S212–22. doi:10.​1002/​jgm.​506.PubMedCrossRef
26.
go back to reference Sauvee M, DidierLaurent G, Latarche C, Escanye MC, Olivier JL, Malaplate-Armand C. Additional use of abeta(4)(2)/abeta(4)(0) ratio with cerebrospinal fluid biomarkers P-tau and abeta(4)(2) increases the level of evidence of Alzheimer’s disease pathophysiological process in routine practice. JAD. 2014;41(2):377–86. doi:10.3233/JAD-131838.PubMed Sauvee M, DidierLaurent G, Latarche C, Escanye MC, Olivier JL, Malaplate-Armand C. Additional use of abeta(4)(2)/abeta(4)(0) ratio with cerebrospinal fluid biomarkers P-tau and abeta(4)(2) increases the level of evidence of Alzheimer’s disease pathophysiological process in routine practice. JAD. 2014;41(2):377–86. doi:10.​3233/​JAD-131838.PubMed
27.
go back to reference Bushman DM, Kaeser GE, Siddoway B, Westra JW, Rivera RR, Rehen SK et al. Genomic mosaicism with increased amyloid precursor protein (APP) gene copy number in single neurons from sporadic Alzheimer’s disease brains. eLife. 2015;4. doi:10.7554/eLife.05116 Bushman DM, Kaeser GE, Siddoway B, Westra JW, Rivera RR, Rehen SK et al. Genomic mosaicism with increased amyloid precursor protein (APP) gene copy number in single neurons from sporadic Alzheimer’s disease brains. eLife. 2015;4. doi:10.​7554/​eLife.​05116
28.
go back to reference Zerah M, Piguet F, Colle MA, Raoul S, Deschamps JY, Deniaud J et al. Intracerebral gene therapy using AAVrh.10-hARSA recombinant vector to treat patients with early-onset forms of metachromatic leukodystrophy: preclinical feasibility and safety assessments in non-human primates. Hum Gene Ther Clin Dev. 2015. doi:10.1089/humc.2014.139 Zerah M, Piguet F, Colle MA, Raoul S, Deschamps JY, Deniaud J et al. Intracerebral gene therapy using AAVrh.10-hARSA recombinant vector to treat patients with early-onset forms of metachromatic leukodystrophy: preclinical feasibility and safety assessments in non-human primates. Hum Gene Ther Clin Dev. 2015. doi:10.​1089/​humc.​2014.​139
29.
go back to reference Wanngren J, Franberg J, Svensson AI, Laudon H, Olsson F, Winblad B, et al. The large hydrophilic loop of presenilin 1 is important for regulating gamma-secretase complex assembly and dictating the amyloid beta peptide (Abeta) Profile without affecting Notch processing. J Biol Chem. 2010;285(12):8527–36. doi:10.1074/jbc.M109.055590.PubMedPubMedCentralCrossRef Wanngren J, Franberg J, Svensson AI, Laudon H, Olsson F, Winblad B, et al. The large hydrophilic loop of presenilin 1 is important for regulating gamma-secretase complex assembly and dictating the amyloid beta peptide (Abeta) Profile without affecting Notch processing. J Biol Chem. 2010;285(12):8527–36. doi:10.​1074/​jbc.​M109.​055590.PubMedPubMedCentralCrossRef
30.
go back to reference Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184–5.PubMedCrossRef Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184–5.PubMedCrossRef
31.
go back to reference Jankowsky JL, Fadale DJ, Anderson J, Xu GM, Gonzales V, Jenkins NA, et al. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum Mol Genet. 2004;13(2):159–70. doi:10.1093/hmg/ddh019.PubMedCrossRef Jankowsky JL, Fadale DJ, Anderson J, Xu GM, Gonzales V, Jenkins NA, et al. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum Mol Genet. 2004;13(2):159–70. doi:10.​1093/​hmg/​ddh019.PubMedCrossRef
36.
go back to reference Proctor DT, Coulson EJ, Dodd PR. Reduction in post-synaptic scaffolding PSD-95 and SAP-102 protein levels in the Alzheimer inferior temporal cortex is correlated with disease pathology. JAD. 2010;21(3):795–811. doi:10.3233/JAD-2010-100090.PubMed Proctor DT, Coulson EJ, Dodd PR. Reduction in post-synaptic scaffolding PSD-95 and SAP-102 protein levels in the Alzheimer inferior temporal cortex is correlated with disease pathology. JAD. 2010;21(3):795–811. doi:10.​3233/​JAD-2010-100090.PubMed
37.
go back to reference Schneider I, Reverse D, Dewachter I, Ris L, Caluwaerts N, Kuiperi C, et al. Mutant presenilins disturb neuronal calcium homeostasis in the brain of transgenic mice, decreasing the threshold for excitotoxicity and facilitating long-term potentiation. J Biol Chem. 2001;276(15):11539–44. doi:10.1074/jbc.M010977200.PubMedCrossRef Schneider I, Reverse D, Dewachter I, Ris L, Caluwaerts N, Kuiperi C, et al. Mutant presenilins disturb neuronal calcium homeostasis in the brain of transgenic mice, decreasing the threshold for excitotoxicity and facilitating long-term potentiation. J Biol Chem. 2001;276(15):11539–44. doi:10.​1074/​jbc.​M010977200.PubMedCrossRef
38.
go back to reference Duff K, Eckman C, Zehr C, Yu X, Prada CM, Perez-tur J, et al. Increased amyloid-beta42(43) in brains of mice expressing mutant presenilin 1. Nature. 1996;383(6602):710–3. doi:10.1038/383710a0.PubMedCrossRef Duff K, Eckman C, Zehr C, Yu X, Prada CM, Perez-tur J, et al. Increased amyloid-beta42(43) in brains of mice expressing mutant presenilin 1. Nature. 1996;383(6602):710–3. doi:10.​1038/​383710a0.PubMedCrossRef
39.
go back to reference Walker JM, Fowler SW, Miller DK, Sun AY, Weisman GA, Wood WG, et al. Spatial learning and memory impairment and increased locomotion in a transgenic amyloid precursor protein mouse model of Alzheimer’s disease. Behav Brain Res. 2011;222(1):169–75. doi:10.1016/j.bbr.2011.03.049.PubMedCrossRef Walker JM, Fowler SW, Miller DK, Sun AY, Weisman GA, Wood WG, et al. Spatial learning and memory impairment and increased locomotion in a transgenic amyloid precursor protein mouse model of Alzheimer’s disease. Behav Brain Res. 2011;222(1):169–75. doi:10.​1016/​j.​bbr.​2011.​03.​049.PubMedCrossRef
40.
go back to reference Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov. 2011;10(9):698–712. doi:10.1038/nrd3505.PubMedCrossRef Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov. 2011;10(9):698–712. doi:10.​1038/​nrd3505.PubMedCrossRef
41.
go back to reference Quon D, Wang Y, Catalano R, Scardina JM, Murakami K, Cordell B. Formation of beta-amyloid protein deposits in brains of transgenic mice. Nature. 1991;352(6332):239–41. doi:10.1038/352239a0.PubMedCrossRef Quon D, Wang Y, Catalano R, Scardina JM, Murakami K, Cordell B. Formation of beta-amyloid protein deposits in brains of transgenic mice. Nature. 1991;352(6332):239–41. doi:10.​1038/​352239a0.PubMedCrossRef
42.
go back to reference Higgins LS, Catalano R, Quon D, Cordell B. Transgenic mice expressing human beta-APP751, but not mice expressing beta-APP695, display early Alzheimer’s disease-like histopathology. Ann N Y Acad Sci. 1993;695:224–7.PubMedCrossRef Higgins LS, Catalano R, Quon D, Cordell B. Transgenic mice expressing human beta-APP751, but not mice expressing beta-APP695, display early Alzheimer’s disease-like histopathology. Ann N Y Acad Sci. 1993;695:224–7.PubMedCrossRef
44.
go back to reference Sudoh S, Kawamura Y, Sato S, Wang R, Saido TC, Oyama F, et al. Presenilin 1 mutations linked to familial Alzheimer’s disease increase the intracellular levels of amyloid beta-protein 1-42 and its N-terminally truncated variant(s) which are generated at distinct sites. J Neurochem. 1998;71(4):1535–43.PubMedCrossRef Sudoh S, Kawamura Y, Sato S, Wang R, Saido TC, Oyama F, et al. Presenilin 1 mutations linked to familial Alzheimer’s disease increase the intracellular levels of amyloid beta-protein 1-42 and its N-terminally truncated variant(s) which are generated at distinct sites. J Neurochem. 1998;71(4):1535–43.PubMedCrossRef
45.
go back to reference Jabaudon D, Shimamoto K, Yasuda-Kamatani Y, Scanziani M, Gahwiler BH, Gerber U. Inhibition of uptake unmasks rapid extracellular turnover of glutamate of nonvesicular origin. Proc Natl Acad Sci U S A. 1999;96(15):8733–8.PubMedPubMedCentralCrossRef Jabaudon D, Shimamoto K, Yasuda-Kamatani Y, Scanziani M, Gahwiler BH, Gerber U. Inhibition of uptake unmasks rapid extracellular turnover of glutamate of nonvesicular origin. Proc Natl Acad Sci U S A. 1999;96(15):8733–8.PubMedPubMedCentralCrossRef
48.
go back to reference Schallier A, Smolders I, Van Dam D, Loyens E, De Deyn PP, Michotte A, et al. Region- and age-specific changes in glutamate transport in the AbetaPP23 mouse model for Alzheimer’s disease. JAD. 2011;24(2):287–300. doi:10.3233/JAD-2011-101005.PubMed Schallier A, Smolders I, Van Dam D, Loyens E, De Deyn PP, Michotte A, et al. Region- and age-specific changes in glutamate transport in the AbetaPP23 mouse model for Alzheimer’s disease. JAD. 2011;24(2):287–300. doi:10.​3233/​JAD-2011-101005.PubMed
56.
go back to reference Tokutake T, Kasuga K, Yajima R, Sekine Y, Tezuka T, Nishizawa M, et al. Hyperphosphorylation of Tau induced by naturally secreted amyloid-beta at nanomolar concentrations is modulated by insulin-dependent Akt-GSK3beta signaling pathway. J Biol Chem. 2012;287(42):35222–33. doi:10.1074/jbc.M112.348300.PubMedPubMedCentralCrossRef Tokutake T, Kasuga K, Yajima R, Sekine Y, Tezuka T, Nishizawa M, et al. Hyperphosphorylation of Tau induced by naturally secreted amyloid-beta at nanomolar concentrations is modulated by insulin-dependent Akt-GSK3beta signaling pathway. J Biol Chem. 2012;287(42):35222–33. doi:10.​1074/​jbc.​M112.​348300.PubMedPubMedCentralCrossRef
57.
go back to reference Augustinack JC, Schneider A, Mandelkow E-M, Hyman BT. Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer’s disease. Acta Neuropathol. 2001;103(1):26–35. doi:10.1007/s004010100423.CrossRef Augustinack JC, Schneider A, Mandelkow E-M, Hyman BT. Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer’s disease. Acta Neuropathol. 2001;103(1):26–35. doi:10.​1007/​s004010100423.CrossRef
60.
go back to reference Volianskis A, Kostner R, Molgaard M, Hass S, Jensen MS. Episodic memory deficits are not related to altered glutamatergic synaptic transmission and plasticity in the CA1 hippocampus of the APPswe/PS1deltaE9-deleted transgenic mice model of ss-amyloidosis. Neurobiol Aging. 2010;31(7):1173–87. doi:10.1016/j.neurobiolaging.2008.08.005.PubMedCrossRef Volianskis A, Kostner R, Molgaard M, Hass S, Jensen MS. Episodic memory deficits are not related to altered glutamatergic synaptic transmission and plasticity in the CA1 hippocampus of the APPswe/PS1deltaE9-deleted transgenic mice model of ss-amyloidosis. Neurobiol Aging. 2010;31(7):1173–87. doi:10.​1016/​j.​neurobiolaging.​2008.​08.​005.PubMedCrossRef
61.
go back to reference Berger A, Lorain S, Josephine C, Desrosiers M, Peccate C, Voit T, et al. Repair of Rhodopsin mRNA by spliceosome-mediated RNA trans-splicing: a new approach for autosomal dominant retinitis pigmentosa. Mol Ther. 2015. doi:10.1038/mt.2015.11.PubMedPubMedCentral Berger A, Lorain S, Josephine C, Desrosiers M, Peccate C, Voit T, et al. Repair of Rhodopsin mRNA by spliceosome-mediated RNA trans-splicing: a new approach for autosomal dominant retinitis pigmentosa. Mol Ther. 2015. doi:10.​1038/​mt.​2015.​11.PubMedPubMedCentral
62.
go back to reference Anderson WW, Collingridge GL. The LTP Program: a data acquisition program for on-line analysis of long-term potentiation and other synaptic events. J Neurosci Methods. 2001;108(1):71–83.PubMedCrossRef Anderson WW, Collingridge GL. The LTP Program: a data acquisition program for on-line analysis of long-term potentiation and other synaptic events. J Neurosci Methods. 2001;108(1):71–83.PubMedCrossRef
Metadata
Title
Alzheimer’s disease-like APP processing in wild-type mice identifies synaptic defects as initial steps of disease progression
Authors
Mickael Audrain
Romain Fol
Patrick Dutar
Brigitte Potier
Jean-Marie Billard
Julien Flament
Sandro Alves
Marie-Anne Burlot
Gaelle Dufayet-Chaffaud
Alexis-Pierre Bemelmans
Julien Valette
Philippe Hantraye
Nicole Déglon
Nathalie Cartier
Jérome Braudeau
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2016
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-016-0070-y

Other articles of this Issue 1/2016

Molecular Neurodegeneration 1/2016 Go to the issue