Neuronal hyperactivity due to loss of inhibitory tone in APOE4 mice lacking Alzheimer's disease-like pathology

Tal Nuriel, Sergio L Angulo, Usman Khan, Archana Ashok, Qiuying Chen, Helen Y Figueroa, Sheina Emrani, Li Liu, Mathieu Herman, Geoffrey Barrett, Valerie Savage, Luna Buitrago, Efrain Cepeda-Prado, Christine Fung, Eliana Goldberg, Steven S Gross, S Abid Hussaini, Herman Moreno, Scott A Small, Karen E Duff, Tal Nuriel, Sergio L Angulo, Usman Khan, Archana Ashok, Qiuying Chen, Helen Y Figueroa, Sheina Emrani, Li Liu, Mathieu Herman, Geoffrey Barrett, Valerie Savage, Luna Buitrago, Efrain Cepeda-Prado, Christine Fung, Eliana Goldberg, Steven S Gross, S Abid Hussaini, Herman Moreno, Scott A Small, Karen E Duff

Abstract

The ε4 allele of apolipoprotein E (APOE) is the dominant genetic risk factor for late-onset Alzheimer's disease (AD). However, the reason APOE4 is associated with increased AD risk remains a source of debate. Neuronal hyperactivity is an early phenotype in both AD mouse models and in human AD, which may play a direct role in the pathogenesis of the disease. Here, we have identified an APOE4-associated hyperactivity phenotype in the brains of aged APOE mice using four complimentary techniques-fMRI, in vitro electrophysiology, in vivo electrophysiology, and metabolomics-with the most prominent hyperactivity occurring in the entorhinal cortex. Further analysis revealed that this neuronal hyperactivity is driven by decreased background inhibition caused by reduced responsiveness of excitatory neurons to GABAergic inhibitory inputs. Given the observations of neuronal hyperactivity in prodromal AD, we propose that this APOE4-driven hyperactivity may be a causative factor driving increased risk of AD among APOE4 carriers.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1
APOE4 is associated with entorhinal cortex hypermetabolism in aged mice. fMRI analysis was performed on aged APOE mice (mean age = 20 months; 7 APOE3 and 8 APOE4 males), and a voxel-based analysis was performed to generate CBV maps of the whole brain of each mouse, followed by co-registration of each image, ROI selection of the hippocampal formation, and statistical comparison between genotypes by a Student’s t test, followed by multiple test correction. a Voxel-based analyses revealed that aged APOE4 mice had increased CBV in the hippocampal formation compared with APOE3 mice, primarily centered around the EC. b 3D superior view of CBV increase detected in aged APOE4 vs. APOE3 mice, as indicated using a heat scale by p value. c Right sagittal view of the same analysis. d Left sagittal view of the same analysis. For each panel, the colored regions depict the hippocampus and subiculum in green, the lateral entorhinal cortex (LEC) in blue, and the medial entorhinal cortex (MEC) in gray
Fig. 2
Fig. 2
APOE4 is associated with increased neuronal activity in awake freely moving mice. In vivo electrophysiology was performed on aged APOE mice (mean age = 18 months; 5 APOE3 and 4 APOE4 males) using a 16-channel microdrive placed directly into the LEC. a Representative local field potentials (LFPs) in awake, freely moving (5–20 cm/s) APOE3 and APOE4 mice, recorded at a depth of 3.3 mm from the brain surface. b A Nissl-stained brain section from a mouse used in this experiment, showing a track mark made by the implanted tetrode, which was descended to a depth of 3.3 mm from the brain surface when the mouse was killed. c Power spectra of LFPs recorded at a depth of 3.1–3.3 mm from the brain surface in freely moving (5–20 cm/s) APOE3 and APOE4 mice. (Frequencies between 55–65 Hz were notch-filtered to remove electrical noise and are extrapolated in the graph using straight lines.) d Percent power distributions comparing LFPs recorded at a depth of 3.1–3.3 mm from the brain surface in freely moving (5–20 cm/s) APOE3 and APOE4 mice (mean values and SEM). A Student’s t test with Welch’s correction was used for statistical comparison between genotypes (delta, p < 0.0001; theta, p = 0.0028; beta, p = 0.0008; low gamma, p < 0.0001; high gamma, p = 0.0015). e Single unit recordings detected at a depth of 3.1–3.3 mm from the brain surface in APOE3 and APOE4 mice, separated into putative excitatory neurons and interneurons based on peak size (black lines represent mean values and SEM). An increased firing rate of excitatory neurons (p < 0.0001) was observed using a Student’s t test with Welch’s correction. (**p < 0.01, ***p < 0.001, ****p < 0.0001)
Fig. 3
Fig. 3
APOE4 is associated with increased expression of small molecules related to energy metabolism. Targeted metabolite profiling was performed on small-molecule metabolites extracted from the EC and PVC of aged APOE mice (14–15 months old; 8 APOE3/3, 9 APOE3/4, and 7 APOE4/4 males). a Scatter dot plots depicting metabolite abundance in the EC for five energy-related metabolites found to be upregulated (Mann–Whitney; malate, p = 0.037; citrate/isocitrate, p = 0.028; ATP, p = 0.049; fructose-6-phosphate, p = 0.011; carnitine, p = 0.021) in APOE4 EC (black lines represent mean values and SEM). b Schematic of the energy metabolism-related metabolites found to be dysregulation in APOE4 EC. (*p ≤ 0.05)
Fig. 4
Fig. 4
APOE4 is associated with abnormal excitability and synaptic plasticity in the hippocampal formation. In vitro electrophysiology was performed on horizontal hippocampal brain slices from aged APOE mice (mean age = 20 months; 5 APOE3 and 4 APOE4 males). a Representative traces of spontaneous extracellular field potentials (sEFPs) recorded in the subiculum (Sub), the dentate gyrus (DG), superficial layers (II/III) of the EC (sEC), and deep layers (V/VI) of the EC (dEC) in APOE3 and APOE4 mice. b Event durations and frequencies comparing APOE3 and APOE4 mice in the different regions recorded (mean values and SEM). Event durations in the Sub, DG, and sEC were found to be increased in aged APOE4 mice (Mann–Whitney; p < 0.001). c Representative sEFPs in APOE4 sEC before and after bath application (25 min) of AP-V (30 μM) and NBQX (10 μM). d A representative horizontal hippocampal brain slice used in this study, with asterisks denoting the location where each electrode was placed and the insert depicting the location of the horizontal slice in the mouse brain. e Cortico-cortical LTP in superficial EC induced by high-frequency stimulation (HFS, 3 trains of 100 pulses at 100 Hz, 10 s interval). Stimulation electrode was placed in lateral EC layer II, and the recording electrode was placed in lateral EC layers III/II. Stimulus intensity was selected to be 50–60% of the maximum amplitude of the fEPSP observed in the input–output curve, and a baseline for 15 mins was obtained before HFS. The slope of the initial component of the fEPSP (10–90%) was normalized to baseline and compared between APOE3 and APOE4 mice (repeated measures ANOVA; F(1,6) = 15.001, p = 0.008). f Mean value of potentiation between 40 and 60 min after LTP induction, comparing APOE3 and APOE4 (t(6) = 3.878, p = 0.008). (**p < 0.01)
Fig. 5
Fig. 5
APOE4 is associated with decreased background synaptic inhibition in layer II entorhinal cortex pyramidal neurons. Miniature inhibitory post-synaptic currents (mIPSCs) were recorded using the patch-clamp whole-cell modality on pyramidal cells (n = 7 cells from APOE3 and n = 8 cells from APOE4) in the layer II EC of aged APOE mice (mean age = 20 months). a Representative traces of mIPSCs of layer II EC pyramidal neurons of APOE3 and APOE4 mice from recordings in voltage-clamp modality (holding at −65 mV) in the presence of APV (10 µM), NBQX (16 µM), and TTX (1 µM). b Representative average mIPSCs from a 3 min recording of an APOE3 and APOE4 mouse. c Plotted are mean + SEM values of peak amplitude of mIPSCs in the different genotypes as indicated (APOE3 26.09 ± 0.73 pA; APOE4 21.6 ± 0.48 pA; p < 0.001). d Cumulative probability distribution of peak amplitude in APOE3 and APOE4 mice, showing lower values for APOE4 in the entire distribution. e Shown are mean + SEM values of instantaneous frequencies in the different genotypes (APOE3 4.92 ± 0.3 Hz; APOE4 5.36 ± 0.31 Hz; t(748) = −1.035, p = 0.301). f Cumulative probability distribution of instantaneous frequencies in APOE3 and APOE4 mice, showing no significant differences. g Plotted are mean + SEM values of decay time constant in both groups (APOE3 9.56 ± 1.61 ms; APOE4 9.19 ± 0.53; p = 0.819). (***p < 0.001)

References

    1. Farrer LA, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997;278:1349–1356. doi: 10.1001/jama.1997.03550160069041.
    1. Mahley RW, Rall SC., Jr. Apolipoprotein E: far more than a lipid transport protein. Annu. Rev. Genomics Hum. Genet. 2000;1:507–537. doi: 10.1146/annurev.genom.1.1.507.
    1. Han X. The role of apolipoprotein E in lipid metabolism in the central nervous system. Cell Mol. Life Sci. 2004;61:1896–1906. doi: 10.1007/s00018-004-4009-z.
    1. Holtzman DM, Herz J, Bu G. Apolipoprotein E and apolipoprotein E receptors: normal biology and roles in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2012;2:a006312.
    1. Bales KR, et al. Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition. Nat. Genet. 1997;17:263–264. doi: 10.1038/ng1197-263.
    1. Castano EM, et al. Fibrillogenesis in Alzheimer’s disease of amyloid beta peptides and apolipoprotein E. Biochem. J. 1995;306(Pt 2):599–604. doi: 10.1042/bj3060599.
    1. Rebeck GW, Reiter JS, Strickland DK, Hyman BT. Apolipoprotein E in sporadic Alzheimer’s disease: allelic variation and receptor interactions. Neuron. 1993;11:575–580. doi: 10.1016/0896-6273(93)90070-8.
    1. Schmechel DE, et al. Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc. Natl Acad. Sci. USA. 1993;90:9649–9653. doi: 10.1073/pnas.90.20.9649.
    1. Ma J, et al. Amyloid-associated proteins alpha 1-antichymotrypsin and apolipoprotein E promote assembly of Alzheimer beta-protein into filaments. Nature. 1994;372:92–94. doi: 10.1038/372092a0.
    1. Castellano JM, et al. Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci. Transl. Med. 2011;3:89ra57. doi: 10.1126/scitranslmed.3002156.
    1. Holtzman DM, et al. Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer’s disease. Proc. Natl Acad. Sci. USA. 2000;97:2892–2897. doi: 10.1073/pnas.050004797.
    1. Huang Y. Abeta-independent roles of apolipoprotein E4 in the pathogenesis of Alzheimer’s disease. Trends Mol. Med. 2010;16:287–294. doi: 10.1016/j.molmed.2010.04.004.
    1. Wolf AB, et al. Apolipoprotein E as a beta-amyloid-independent factor in alzheimer’s disease. Alzheimers Res. Ther. 2013;5:38. doi: 10.1186/alzrt204.
    1. Busche MA, et al. Decreased amyloid-beta and increased neuronal hyperactivity by immunotherapy in Alzheimer’s models. Nat. Neurosci. 2015;18:1725–7. doi: 10.1038/nn.4163.
    1. Busche MA, et al. Clusters of hyperactive neurons near amyloid plaques in a mouse model of Alzheimer’s disease. Science. 2008;321:1686–1689. doi: 10.1126/science.1162844.
    1. Davis KE, Fox S, Gigg J. Increased hippocampal excitability in the 3xTgAD mouse model for Alzheimer’s disease in vivo. PLoS ONE. 2014;9:e91203. doi: 10.1371/journal.pone.0091203.
    1. 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. doi: 10.1016/j.neuron.2007.07.025.
    1. Hamalainen A, et al. Increased fMRI responses during encoding in mild cognitive impairment. Neurobiol. Aging. 2007;28:1889–1903. doi: 10.1016/j.neurobiolaging.2006.08.008.
    1. Kircher TT, et al. Hippocampal activation in patients with mild cognitive impairment is necessary for successful memory encoding. J. Neurol. Neurosurg. Psychiatry. 2007;78:812–818. doi: 10.1136/jnnp.2006.104877.
    1. Bakker A, et al. Reduction of hippocampal hyperactivity improves cognition in amnestic mild cognitive impairment. Neuron. 2012;74:467–474. doi: 10.1016/j.neuron.2012.03.023.
    1. Dickerson BC, et al. Increased hippocampal activation in mild cognitive impairment compared to normal aging and AD. Neurology. 2005;65:404–411. doi: 10.1212/01.wnl.0000171450.97464.49.
    1. Miller SL, et al. Hippocampal activation in adults with mild cognitive impairment predicts subsequent cognitive decline. J. Neurol. Neurosurg. Psychiatry. 2008;79:630–635. doi: 10.1136/jnnp.2007.124149.
    1. Quiroz YT, et al. Hippocampal hyperactivation in presymptomatic familial Alzheimer’s disease. Ann. Neurol. 2010;68:865–875. doi: 10.1002/ana.22105.
    1. Sepulveda-Falla D, Glatzel M, Lopera F. Phenotypic profile of early-onset familial Alzheimer’s disease caused by presenilin-1 E280A mutation. J. Alzheimers Dis. 2012;32:1–12.
    1. Das U, et al. Activity-induced convergence of APP and BACE-1 in acidic microdomains via an endocytosis-dependent pathway. Neuron. 2013;79:447–460. doi: 10.1016/j.neuron.2013.05.035.
    1. Cirrito JR, et al. Synaptic activity regulates interstitial fluid amyloid-beta levels in vivo. Neuron. 2005;48:913–922. doi: 10.1016/j.neuron.2005.10.028.
    1. Yamamoto K, et al. Chronic optogenetic activation augments abeta pathology in a mouse model of Alzheimer disease. Cell Rep. 2015;11:859–865. doi: 10.1016/j.celrep.2015.04.017.
    1. Pooler AM, Phillips EC, Lau DH, Noble W, Hanger DP. Physiological release of endogenous tau is stimulated by neuronal activity. EMBO Rep. 2013;14:389–394. doi: 10.1038/embor.2013.15.
    1. Yamada K, et al. Neuronal activity regulates extracellular tau in vivo. J. Exp. Med. 2014;211:387–393. doi: 10.1084/jem.20131685.
    1. Wu JW, et al. Neuronal activity enhances tau propagation and tau pathology in vivo. Nat. Neurosci. 2016;19:1085–92. doi: 10.1038/nn.4328.
    1. Filippini N, et al. Distinct patterns of brain activity in young carriers of the APOE-epsilon4 allele. Proc. Natl Acad. Sci. USA. 2009;106:7209–7214. doi: 10.1073/pnas.0811879106.
    1. Bookheimer SY, et al. Patterns of brain activation in people at risk for Alzheimer’s disease. N. Engl. J. Med. 2000;343:450–456. doi: 10.1056/NEJM200008173430701.
    1. Trachtenberg AJ, Filippini N, Mackay CE. The effects of APOE-epsilon4 on the BOLD response. Neurobiol. Aging. 2012;33:323–334. doi: 10.1016/j.neurobiolaging.2010.03.009.
    1. Fleisher AS, et al. Cerebral perfusion and oxygenation differences in Alzheimer’s disease risk. Neurobiol. Aging. 2009;30:1737–1748. doi: 10.1016/j.neurobiolaging.2008.01.012.
    1. Wierenga CE, et al. Effect of mild cognitive impairment and APOE genotype on resting cerebral blood flow and its association with cognition. J. Cereb. Blood Flow Metab. 2012;32:1589–1599. doi: 10.1038/jcbfm.2012.58.
    1. Jack CR, Jr, et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 2013;12:207–216. doi: 10.1016/S1474-4422(12)70291-0.
    1. Sullivan PM, et al. Targeted replacement of the mouse apolipoprotein E gene with the common human APOE3 allele enhances diet-induced hypercholesterolemia and atherosclerosis. J. Biol. Chem. 1997;272:17972–17980. doi: 10.1074/jbc.272.29.17972.
    1. Sullivan PM, Mace BE, Maeda N, Schmechel DE. Marked regional differences of brain human apolipoprotein E expression in targeted replacement mice. Neuroscience. 2004;124:725–733. doi: 10.1016/j.neuroscience.2003.10.011.
    1. Moreno H, Hua F, Brown T, Small SA. Longitudinal mapping of mouse cerebral blood volume with MRI. NMR in Biomedicine. 2006;19:535–543. doi: 10.1002/nbm.1022.
    1. Moreno H, et al. Imaging the abeta-related neurotoxicity of Alzheimer disease. Arch. Neurol. 2007;64:1467–1477. doi: 10.1001/archneur.64.10.1467.
    1. Raichle ME. Positron emission tomography. Annu. Rev. Neurosci. 1983;6:249–267. doi: 10.1146/annurev.ne.06.030183.001341.
    1. Leenders KL, et al. Cerebral blood flow, blood volume and oxygen utilization. Normal values and effect of age. Brain. 1990;113:27–47. doi: 10.1093/brain/113.1.27.
    1. Gonzalez RG, et al. Functional MR in the evaluation of dementia: correlation of abnormal dynamic cerebral blood volume measurements with changes in cerebral metabolism on positron emission tomography with fludeoxyglucose F 18. Am. J. Neuroradiol. 1995;16:1763–1770.
    1. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta. Neuropathol. 1991;82:239–259. doi: 10.1007/BF00308809.
    1. Richards K, et al. Segmentation of the mouse hippocampal formation in magnetic resonance images. Neuroimage. 2011;58:732–740. doi: 10.1016/j.neuroimage.2011.06.025.
    1. Hussaini SA, Kempadoo KA, Thuault SJ, Siegelbaum SA, Kandel ER. Increased size and stability of CA1 and CA3 place fields in HCN1 knockout mice. Neuron. 2011;72:643–653. doi: 10.1016/j.neuron.2011.09.007.
    1. Kohara K, et al. Cell type-specific genetic and optogenetic tools reveal hippocampal CA2 circuits. Nat. Neurosci. 2014;17:269–279. doi: 10.1038/nn.3614.
    1. Quirk MC, Sosulski DL, Feierstein CE, Uchida N, Mainen ZF. A defined network of fast-spiking interneurons in orbitofrontal cortex: responses to behavioral contingencies and ketamine administration. Front. Syst. Neurosci. 2009;3:13. doi: 10.3389/neuro.06.013.2009.
    1. Chen Q, et al. Untargeted plasma metabolite profiling reveals the broad systemic consequences of xanthine oxidoreductase inactivation in mice. PLoS ONE. 2012;7:e37149. doi: 10.1371/journal.pone.0037149.
    1. Wang X, Michaelis ML, Michaelis EK. Functional genomics of brain aging and Alzheimer’s disease: focus on selective neuronal vulnerability. Curr. Genomics. 2010;11:618–633. doi: 10.2174/138920210793360943.
    1. Minoshima S, et al. Metabolic reduction in the posterior cingulate cortex in very early Alzheimer’s disease. Ann. Neurol. 1997;42:85–94. doi: 10.1002/ana.410420114.
    1. Cepeda-Prado E, et al. R6/2 Huntington’s disease mice develop early and progressive abnormal brain metabolism and seizures. J. Neurosci. 2012;32:6456–6467. doi: 10.1523/JNEUROSCI.0388-12.2012.
    1. Walther H, Lambert JD, Jones RS, Heinemann U, Hamon B. Epileptiform activity in combined slices of the hippocampus, subiculum and entorhinal cortex during perfusion with low magnesium medium. Neurosci. Lett. 1986;69:156–161. doi: 10.1016/0304-3940(86)90595-1.
    1. Kunitake A, Kunitake T, Stewart M. Differential modulation by carbachol of four separate excitatory afferent systems to the rat subiculum in vitro. Hippocampus. 2004;14:986–999. doi: 10.1002/hipo.20016.
    1. Criscuolo C, et al. BDNF prevents amyloid-dependent impairment of LTP in the entorhinal cortex by attenuating p38 MAPK phosphorylation. Neurobiol. Aging. 2015;36:1303–1309. doi: 10.1016/j.neurobiolaging.2014.11.016.
    1. Canto CB, Witter MP. Cellular properties of principal neurons in the rat entorhinal cortex. I. The lateral entorhinal cortex. Hippocampus. 2012;22:1256–1276. doi: 10.1002/hipo.20997.
    1. Alonso A, Klink R. Differential electroresponsiveness of stellate and pyramidal-like cells of medial entorhinal cortex layer II. J. Neurophysiol. 1993;70:128–143.
    1. Prange O, Murphy TH. Correlation of miniature synaptic activity and evoked release probability in cultures of cortical neurons. J. Neurosci. 1999;19:6427–6438.
    1. Liraz O, Boehm-Cagan A, Michaelson DM. ApoE4 induces Abeta42, tau, and neuronal pathology in the hippocampus of young targeted replacement apoE4 mice. Mol. Neurodegener. 2013;8:16. doi: 10.1186/1750-1326-8-16.
    1. Sullivan PM, Mace BE, Estrada JC, Schmechel DE, Alberts MJ. Human apolipoprotein E4 targeted replacement mice show increased prevalence of intracerebral hemorrhage associated with vascular amyloid deposition. J. Stroke Cerebrovasc. Dis. 2008;17:303–311. doi: 10.1016/j.jstrokecerebrovasdis.2008.03.011.
    1. Klein RC, Acheson SK, Mace BE, Sullivan PM, Moore SD. Altered neurotransmission in the lateral amygdala in aged human apoE4 targeted replacement mice. Neurobiol. Aging. 2014;35:2046–2052. doi: 10.1016/j.neurobiolaging.2014.02.019.
    1. Hunter JM, et al. Emergence of a seizure phenotype in aged apolipoprotein epsilon 4 targeted replacement mice. Brain Res. 2012;1467:120–132. doi: 10.1016/j.brainres.2012.05.048.
    1. Gillespie AK, et al. Apolipoprotein E4 causes age-dependent disruption of slow gamma Oscillations during Hippocampal sharp-wave ripples. Neuron. 2016;90:740–751. doi: 10.1016/j.neuron.2016.04.009.
    1. Sullivan D, et al. Relationships between hippocampal sharp waves, ripples, and fast gamma oscillation: influence of dentate and entorhinal cortical activity. J. Neurosci. 2011;31:8605–8616. doi: 10.1523/JNEUROSCI.0294-11.2011.
    1. Haier RJ, et al. Temporal cortex hypermetabolism in down syndrome prior to the onset of dementia. Neurology. 2003;61:1673–1679. doi: 10.1212/01.WNL.0000098935.36984.25.
    1. Small SA, Perera GM, DeLaPaz R, Mayeux R, Stern Y. Differential regional dysfunction of the hippocampal formation among elderly with memory decline and Alzheimer’s disease. Ann. Neurol. 1999;45:466–472. doi: 10.1002/1531-8249(199904)45:4<466::AID-ANA8>;2-Q.
    1. Small SA, Tsai WY, DeLaPaz R, Mayeux R, Stern Y. Imaging hippocampal function across the human life span: is memory decline normal or not? Ann. Neurol. 2002;51:290–295. doi: 10.1002/ana.10105.
    1. de Leon MJ, et al. Prediction of cognitive decline in normal elderly subjects with 2-[(18)F]fluoro-2-deoxy-D-glucose/poitron-emission tomography (FDG/PET) Proc. Natl Acad. Sci. USA. 2001;98:10966–10971. doi: 10.1073/pnas.191044198.
    1. Karow DS, et al. Relative capability of MR imaging and FDG PET to depict changes associated with prodromal and early Alzheimer disease. Radiology. 2010;256:932–942. doi: 10.1148/radiol.10091402.
    1. Khan UA, et al. Molecular drivers and cortical spread of lateral entorhinal cortex dysfunction in preclinical Alzheimer’s disease. Nat. Neurosci. 2014;17:304–311. doi: 10.1038/nn.3606.
    1. Walsh DM, et al. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–539. doi: 10.1038/416535a.
    1. Shankar GM, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat. Med. 2008;14:837–842. doi: 10.1038/nm1782.
    1. Kamenetz F, et al. APP processing and synaptic function. Neuron. 2003;37:925–937. doi: 10.1016/S0896-6273(03)00124-7.
    1. Gomez-Isla T, et al. Profound loss of layer II entorhinal cortex neurons occurs in very mild Alzheimer’s disease. J. Neurosci. 1996;16:4491–4500.
    1. Hoover BR, et al. Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron. 2010;68:1067–1081. doi: 10.1016/j.neuron.2010.11.030.
    1. Brickman AM, Small SA, Fleisher A. Pinpointing synaptic loss caused by Alzheimer’s disease with fMRI. Behav. Neurol. 2009;21:93–100. doi: 10.1155/2009/246892.
    1. Sabuncu MR, Yeo BT, Van Leemput K, Vercauteren T, Golland P. Asymmetric image-template registration. Med. Image Comput. Comput. Assist. Interv. 2009;12:565–573.
    1. Matyash V, Liebisch G, Kurzchalia TV, Shevchenko A, Schwudke D. Lipid extraction by methyl-tert-butyl ether for high-throughput lipidomics. J. Lipid Res. 2008;49:1137–1146. doi: 10.1194/jlr.D700041-JLR200.
    1. Giavalisco P, et al. Elemental formula annotation of polar and lipophilic metabolites using (13) C, (15) N and (34) S isotope labelling, in combination with high-resolution mass spectrometry. Plant J. 2011;68:364–376. doi: 10.1111/j.1365-313X.2011.04682.x.
    1. Woodhall GL, Bailey SJ, Thompson SE, Evans DI, Jones RS. Fundamental differences in spontaneous synaptic inhibition between deep and superficial layers of the rat entorhinal cortex. Hippocampus. 2005;15:232–245. doi: 10.1002/hipo.20047.
    1. von Kienlin M, et al. Altered metabolic profile in the frontal cortex of PS2APP transgenic mice, monitored throughout their life span. Neurobiol. Dis. 2005;18:32–39. doi: 10.1016/j.nbd.2004.09.005.
    1. Salek RM, et al. A metabolomic study of the CRND8 transgenic mouse model of Alzheimer’s disease. Neurochem. Int. 2010;56:937–947. doi: 10.1016/j.neuint.2010.04.001.

Source: PubMed

3
Se inscrever