Preclinical and clinical biomarker studies of CT1812: A novel approach to Alzheimer's disease modification

Nicholas J Izzo, Carla M Yuede, Kelsie M LaBarbera, Colleen S Limegrover, Courtney Rehak, Raymond Yurko, Lora Waybright, Gary Look, Gilbert Rishton, Hank Safferstein, Mary E Hamby, Claire Williams, Kelsey Sadlek, Hannah M Edwards, Charles S Davis, Michael Grundman, Lon S Schneider, Steven T DeKosky, Daniel Chelsky, Ian Pike, Christopher Henstridge, Kaj Blennow, Henrik Zetterberg, Harry LeVine 3rd, Tara L Spires-Jones, John R Cirrito, Susan M Catalano, Nicholas J Izzo, Carla M Yuede, Kelsie M LaBarbera, Colleen S Limegrover, Courtney Rehak, Raymond Yurko, Lora Waybright, Gary Look, Gilbert Rishton, Hank Safferstein, Mary E Hamby, Claire Williams, Kelsey Sadlek, Hannah M Edwards, Charles S Davis, Michael Grundman, Lon S Schneider, Steven T DeKosky, Daniel Chelsky, Ian Pike, Christopher Henstridge, Kaj Blennow, Henrik Zetterberg, Harry LeVine 3rd, Tara L Spires-Jones, John R Cirrito, Susan M Catalano

Abstract

Introduction: Amyloid beta (Aβ) oligomers are one of the most toxic structural forms of the Aβ protein and are hypothesized to cause synaptotoxicity and memory failure as they build up in Alzheimer's disease (AD) patients' brain tissue. We previously demonstrated that antagonists of the sigma-2 receptor complex effectively block Aβ oligomer toxicity. CT1812 is an orally bioavailable, brain penetrant small molecule antagonist of the sigma-2 receptor complex that appears safe and well tolerated in healthy elderly volunteers. We tested CT1812's effect on Aβ oligomer pathobiology in preclinical AD models and evaluated CT1812's impact on cerebrospinal fluid (CSF) protein biomarkers in mild to moderate AD patients in a clinical trial (ClinicalTrials.gov NCT02907567).

Methods: Experiments were performed to measure the impact of CT1812 versus vehicle on Aβ oligomer binding to synapses in vitro, to human AD patient post mortem brain tissue ex vivo, and in living APPSwe /PS1dE9 transgenic mice in vivo. Additional experiments were performed to measure the impact of CT1812 versus vehicle on Aβ oligomer-induced deficits in membrane trafficking rate, synapse number, and protein expression in mature hippocampal/cortical neurons in vitro. The impact of CT1812 on cognitive function was measured in transgenic Thy1 huAPPSwe/Lnd+ and wild-type littermates. A multicenter, double-blind, placebo-controlled parallel group trial was performed to evaluate the safety, tolerability, and impact on protein biomarker expression of CT1812 or placebo given once daily for 28 days to AD patients (Mini-Mental State Examination 18-26). CSF protein expression was measured by liquid chromatography with tandem mass spectrometry or enzyme-linked immunosorbent assay in samples drawn prior to dosing (Day 0) and at end of dosing (Day 28) and compared within each patient and between pooled treated versus placebo-treated dosing groups.

Results: CT1812 significantly and dose-dependently displaced Aβ oligomers bound to synaptic receptors in three independent preclinical models of AD, facilitated oligomer clearance into the CSF, increased synaptic number and protein expression in neurons, and improved cognitive performance in transgenic mice. CT1812 significantly increased CSF concentrations of Aβ oligomers in AD patient CSF, reduced concentrations of synaptic proteins and phosphorylated tau fragments, and reversed expression of many AD-related proteins dysregulated in CSF.

Discussion: These preclinical studies demonstrate the novel disease-modifying mechanism of action of CT1812 against AD and Aβ oligomers. The clinical results are consistent with preclinical data and provide evidence of target engagement and impact on fundamental disease-related signaling pathways in AD patients, supporting further development of CT1812.

Keywords: Abeta oligomers; Alzheimer's disease; clinical trial; sigma-2 receptor; synapse.

Conflict of interest statement

N.I., K.M.L., C.L., C.R., R.Y., L.W., G.L., G.R., H.S., M.H., C.W., K.S., and S.C. are employees of Cognition Therapeutics, Inc. C.Y., J.C., H.L., M.S., M.G., and T.S.‐J. are paid consultants of Cognition Therapeutics, Inc. L.S. reports grants and personal fees from Eli Lilly; personal fees from Avraham, Ltd; personal fees from Boehringer Ingelheim; grants and personal fees from Merck; personal fees from Neurim, Ltd; personal fees from Neuronix, Ltd; personal fees from Cognition Therapeutics, Inc.; personal fees from Eisai; personal fees from Takeda; personal fees from vTv; grants and personal fees from Roche/Genentech; grants from Biogen; grants from Novartis; personal fees from Abbott; grants from Biohaven, outside the submitted work. S.D. is a member of the Neuroscience Advisory Board for Amgen; Chair of the Medical Scientific Advisory Board for Acumen; Chair of the Drug Monitoring Committee for Biogen; Chair of the Medical Advisory Board for Cognition Therapeutics, Inc.; and Editor for Dementia for UpToDate. H.Z. has served at scientific advisory boards for Cognition Therapeutics, Roche Diagnostics, Wave, and Samumed; has given lectures in symposia sponsored by Biogen and Alzecure; and is a co‐founder of Brain Biomarker Solutions in Gothenburg AB, a GU Ventures‐based platform company at the University of Gothenburg. T.S. is a scientific advisory board member of Cognition Therapeutics, Inc. and receives collaborative grant funding from two pharmaceutical companies. K.B. has served as a consultant or at advisory boards for Alector, Biogen, Cognition Therapeutics, Lilly, MagQu, Novartis, and Roche Diagnostics, and is a co‐founder of Brain Biomarker Solutions in Gothenburg AB, a GU Venture‐based platform company at the University of Gothenburg. International Patent WO 15/116923 pertains to the results presented in this paper.

© 2021 The Authors. Alzheimer's & Dementia published by Wiley Periodicals LLC on behalf of Alzheimer's Association.

Figures

FIGURE 1
FIGURE 1
CT1812 is a selective sigma‐2 receptor ligand that prevents and treats amyloid beta (Aβ) oligomer‐induced membrane trafficking deficits. A, CT1812 binds sigma‐2 receptors with a 8.5 nM Kd and sigma‐1 receptors with a 63 nM Kd as measured in radioligand displacement studies (n = 2; means ± standard deviation [SD]). B, Effects of synthetic Aβ oligomers on vesicle trafficking in primary cultures of neurons and glia. Quantification is shown of the amount of formazan within intracellular vesicles in Aβ oligomer‐treated (red dot) or vehicle‐treated (blue square) conditions, and the dose‐dependent restoration of trafficking rate to normal vehicle‐treated values after CT1812 addition 1 hour before addition of Aβ oligomers (gray squares, EC50 = 6.7 μM, n = 11 experiments) or 1 hour after addition of Aβ oligomers (black squares, EC50 = 0.36 μM, n = 12 experiments). Vesicle trafficking was assayed 24 hours after addition of Aβ oligomers (1.75 μM, total peptide; means ± standard error of the mean). N represents the number of replicate experiments from separate cell culture preparations. C, Human Alzheimer's disease brain derived Aβ oligomers (1‐hour treatment) caused a 27 ± 11% (SD; P < 0.001, n = 3) decrease in trafficking rate in cultured neurons and glia. Pretreatment of cultures for 1 hour with 15 μM CT1812 before addition of human derived Aβ oligomers blocked the trafficking deficits (P < 0.001 for Aβ oligomers vs. Aβ oligomers + CT1812, analysis of variance with Tukey's post hoc test, n = 3 experiments). Each point represents results from quadruple replicates from Aβ oligomer preparations isolated from three separate donor brains with the mean of all the points represented by the horizontal lines
FIGURE 2
FIGURE 2
CT1812 prevents amyloid beta (Aβ) oligomer binding to and displaces bound oligomers from neuronal receptors, leading to restoration of synaptic density and protein expression in cultured mouse neurons. A, Aβ oligomers (750 nM, > 80% of Kd) bind specifically and saturably to a single receptor site on some, but not all, neurons in mature (> 21 DIV) primary hippocampal and cortical neuronal cultures. B, CT1812 [10 uM] prevents Aβ binding by 59 ± 8% (standard error of the mean [SEM]) when added 1 hour prior to addition of Aβ oligomers. C, CT1812 [10 uM] displaces Aβ oligomer binding by 42 ± 9% (SEM) when added 1 hour after addition of Aβ oligomers. Scale bar = 20 microns. D, CT1812 prevents binding and more than doubles the Kd of oligomer binding, from 430 nM (at 0 μM CT1812) to 1120 nM (at 10 μM CT1812; n = 6; means ± SEM). E, CT1812 displaces binding and nearly doubles the Kd of oligomer binding, from 610 nM (at 0 μM CT1812) to 1160 nM (for 10 μM CT1812; n = 6; means ± SEM). F, top, Aβ oligomers bind to receptors at neuronal synapses (red, 6E10 immunodetection), resulting in a significant loss of synapses (green, drebrin immunodetection, 12.8 ± 2.6% [SEM] decrease in synapse density, P < 0.01 vs. vehicle‐treated, Student t test [G]; the blue square represents synapse density in vehicle‐treated neurons). F, bottom,CT1812 displaces bound Aβ oligomers and restores synaptic numbers to normal in a dose‐dependent manner. Scale bar = 5 microns. G, CT1812 restores synapse number to normal whether added before (prevention, EC50 = 68 nM) or after (treatment, EC50 = 127 nM) Aβ oligomers (*P < 0.5 vs. Aβ oligomers alone for both conditions, Student's t test, means ± SEM). Treatment with CT1812 restores synaptic protein expression in cultured rat neurons. H, Neurogranin is expressed at high levels in postsynaptic dendrites and synapses in a subset of neurons in primary hippocampal/cortical cultures. I, Addition of Aβ oligomers (0.75 μM, 24 hours) causes a 28% loss of neurons expressing high levels of neurogranin (P = 0.014). J, Treatment with 4.8 μM CT1812 1 hour after addition of Aβ oligomers restores the expression of neurogranin to normal levels (Aβ oligomers vs. Aβ oligomers + CT1812 P = 0.031, vehicle vs. Aβ oligomers + CT1812, n.s., analysis of variance [ANOVA] with Tukey's post hoc test, n = 20). K, Synaptotagmin‐1 is expressed in presynaptic terminals. L, Addition of Aβ oligomers (3.5 μM, 48 hours) cause a 37% loss of synaptotagmin‐1 presynaptic terminals in primary hippocampal/cortical cultures (P = 0.0068, N = 24). M, Treatment with 4.8 μM CT1812 1 hour after addition of Aβ oligomers blocks these losses and restores expression of synaptotagmin‐1 levels (vehicle vs. Aβ oligomers + CT1812, n.s). H–M, ANOVA with Tukey's post hoc test, n = 20. For all cell culture experiments, n represents the number of replicate experiments from separate cell culture preparations, and data points represent means ± SD or SEM as noted
FIGURE 3
FIGURE 3
CT1812 treatment displaces amyloid beta (Aβ) oligomers from mouse hippocampal receptors and post mortem Alzheimer's disease human patient tissue and facilitates their clearance into cerebrospinal fluid (CSF). A, B, Microimmunoelectrodes (MIE) coated with oligomer‐specific antibody A11 (A) or mAβ40 antibody HJ2 (B) in 12‐month‐old transgenic hAPP/PS1 mice detect soluble Aβ in the hippocampal interstitial fluid (ISF). A, After intravenous injection of 0.3 or 3 mg/kg CT1812 (vertical dashed line), soluble Aβ oligomers are significantly elevated in hippocampal ISF in CT1812‐treated mice (n = 7) compared to vehicle‐treated mice (n = 4, means ± standard error of the mean [SEM]; *** P < 0.001, **** P < 0.0001, two‐way analysis of variance [ANOVA] with Sidak's post hoc test for effect of drug vs. vehicle). B, CT1812 treatment does not cause a change in total Aβ levels in the hippocampus (n = 5) compared to vehicle treatment (n = 4, means ± SEM). C–E, Aβ oligomers displaced from post mortem brain tissue sections from 8 AD patients were quantified by enzyme‐linked immunosorbent assay (ELISA; D) and native western blots (C [representative individual patient], E), and show the expected dose‐dependent increase in concentration in CT1812‐treated compared to the vehicle‐treated condition. For western blots, the sum of the intensity of discrete Aβ protein bands at 22, 33, 43, 48, and 85 Kd was quantified for each donor tissue section (C). In (D) and (E), values for Aβ measured by ELISA for each brain tissue section and for intensity of western blot bands for each brain tissue section normalized to vehicle‐treated controls (dashed line) from the same donor are shown, with the mean of all the points represented by the horizontal lines (*P < 0.05, ANOVA with Tukey's post hoc test for each treatment vs. vehicle treated controls). F (representative individual patient), G, H, Aβ oligomers (red) located in a 2 μM halo surrounding compact thioflavin‐S positive plaques (green) are displaced from frozen post mortem human AD brain tissue sections (N = 5 patients) by CT1812 in a dose‐dependent manner normalized to vehicle treated brain sections (dashed line) from the same individual with the mean of all the points represented by the horizontal lines (*P < 0.05, ANOVA with Tukey's post hoc test for each CT1812‐treated vs. vehicle‐treated controls). H, Aβ intensity within plaques does not change. Scale bar = 20 μM. I, J, Microimmunoelectrodes coated with oligomer‐specific antibody A11 (I) or pan‐Aβ antibody HJ2 (J) in 12‐month‐old transgenic hAPP/PS1 mice detect soluble Aβ in the lateral ventricle CSF. I, After intravenous injection of 0.3 (n = 7) or 3 (n = 5) mg/kg CT1812 (vertical dashed line), soluble Aβ oligomers are significantly elevated in lateral ventricle CSF compared to vehicle‐treated mice (n = 5, means ± SEM). J, CT1812 treatment (n = 5) did not cause a change in Aβ monomer levels in the CSF compared to vehicle treatment (n = 4 means ± SEM). *** P < 0.001, **** P < 0.0001, two‐way ANOVA Sidak's post hoc test for effect of drug vs. vehicle
FIGURE 4
FIGURE 4
Treatment with CT1812 improves learning and memory deficits in transgenic Alzheimer's mice. A, Transgenic Thy‐1 huAPPSwe/Ldn+ male mice treated with CT1812 (Tg + CT1812) learn the Morris water maze task significantly better than do transgenic vehicle‐treated mice (Tg + vehicle; P = 0.016, two‐way repeated measures analysis of variance, Bonferroni post hoc *P < 0.5; mean ± standard error of the mean). CT1812 treatment does not affect non‐transgenic animal performance (nTg + CT1812). B, Transgenic mice treated with CT1812 remember previous arms entered in the Y maze task significantly better (P = 0.013, Student's t test) than chance (dashed line), but transgenic vehicle‐treated animals do not (nTg + vehicle, 62.7 ± standard deviation [SD] 12.2%; Tg + vehicle, 56.1 ± SD 9.2%; Tg + CT1812, 58.5 ± SD 9.4%; nTg + CT1812, 65.3 ± SD 6.0%). C, Transgenic mice show deficits in the Contextual Fear Conditioning test (P = 0.037, Student's t test), while transgenic and nontransgenic mice treated with CT1812 do not (nTg + vehicle, 52.5 ± SD 5.4%; Tg + vehicle, 37.9 ± SD 6.4%; Tg + CT1812, 44.6 ± SD 6.5%; nTg + CT1812, 50.9 ± SD 5.1%). Each data point in (B) and (C) represents an individual mouse with the mean of all the points represented by the horizontal lines. D, Plasma and brain concentration of CT1812 after a single p.o. dose of 10 mg/kg (n = 3 mice per time point, mean ± SD)
FIGURE 5
FIGURE 5
CT1812 treatment significantly impacts cerebrospinal fluid (CSF) biomarkers of target engagement and disease‐related biology. Protein concentrations in CSF samples from Alzheimer's disease (AD) patients in a 28‐day phase 1b/2a clinical trial of CT1812 vs. placebo were measured via enzyme‐linked immunosorbent assay (ELISA), liquid chromatography tandem mass spectrometry (LC‐MS/MS), or gel electrophoresis. A, After 28 days the concentration of amyloid beta (Aβ) oligomers measured via western blot in CT1812‐treated AD patient CSF increases compared to the patient's own baseline and vs. placebo (P = 0.014, Student's t test, n = 3 placebo, 10 CT1812‐treated), providing supporting evidence of clinical target engagement. At day 28 of the study, the concentration of synaptic damage proteins neurogranin (B, measured by ELISA) and synaptotagmin‐1 (C, measured by LC‐MS/MS) decreases compared to the patient's own baseline and vs. placebo (P = 0.050 analysis of covariance, n = 5 placebo, 11 CT1812‐treated; and F1,12 = 8.8, P = 0.011, n = 4 placebo, 9 CT1812‐treated, respectively) providing evidence of a positive effect on synapses in patients with AD. A, B, and C, Data from individual patients are displayed with the treatment group mean represented by the horizontal lines. These changes are corroborated (D) by significant expression changes (P < 0.05) in multiple synaptic proteome proteins in the CSF measured by unbiased LC‐MS/MS in AD patients treated for 28 days with CT1812 vs. placebo. E, CSF proteomics identifies a subset of proteins significantly altered with CT1812 that have been reported to be significantly regulated in AD. Data are expressed as mean % change in protein concentration in CSF from CT1812 treated (n = 11) vs. placebo (n = 4; blue) and AD vs. control to illustrate how these proteins are altered in AD relative to cognitively normal age‐matched controls. All analytes shown are significantly regulated (P < 0.05) in AD patients vs. control and in the present study. F, After 28 days of treatment, the abundance of six tau phosphorylation peptides decreased by 30% (the threshold for noise distribution), or more after treatment with CT1812 compared to placebo (T52, T205, S262, S263, S285, S305) while one site increased more than 30% (S191), but the concentration of unphosphorylated tau did not change, providing evidence of CT1812 impact on pathological disease signaling. Colored bars represent amino acid sites within tau that are phosphorylated by GSK3β with priming (blue bars) or without priming (green bars),
FIGURE 6
FIGURE 6
Hypothesized mechanism of action of CT1812: synaptoprotective displacement of toxic amyloid beta (Aβ) oligomers from neurons. A, In the absence of Aβ oligomers, pre‐ and post‐synaptic proteins such as synaptotagmin‐1 and neurogranin are expressed at normal levels. B, The sigma‐2 receptor complex tightly regulates the oligomer receptor complex by stabilizing plasma membrane expression of oligomer receptor component proteins. C, Oligomer binding results in changing expression/localization of synapse associated proteins, spine loss, and memory failure, as well as upregulation of the sigma‐2 receptor complex. D, Fragmented pre‐ and post‐synaptic proteins such as synaptotagmin‐1 and neurogranin move into interstitial fluid and then cerebrospinal fluid. E, CT1812 binds to the sigma‐2 receptor ligand binding site within the sigma‐2 receptor complex, acting as a negative allosteric modulator of Aβ oligomer binding. This destabilizes the oligomer binding site and displaces oligomers from synapses without affecting normal synapse protein function. F, Synapse number and memory are restored to normal levels

References

    1. Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer's disease drug development pipeline: 2019. Alzheimer's Dement Transl Res Clin Interv. 2019;5:272‐293. 10.1016/j.trci.2019.05.008.
    1. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol Med. 2016;8:595‐608. 10.15252/emmm.201606210.
    1. Hardy J. The Discovery of Alzheimer causing Mutations in the APP Gene and the Formulation of the “Amyloid Cascade Hypothesis. FEBS J. 2017;38:42‐49. 10.1111/febs.14004.
    1. Walsh DM, Selkoe DJ. Amyloid β‐protein and beyond: the path forward in Alzheimer's disease. Curr Opin Neurobiol. 2020;61:116‐124. 10.1016/j.conb.2020.02.003.
    1. Jonsson T, Atwal JK, Steinberg S, et al. A mutation in APP protects against Alzheimer's disease and age‐related cognitive decline. Nature. 2012;488:96‐99. 10.1038/nature11283.
    1. Limegrover CS, LeVine H, Izzo NJ, et al. Alzheimer's protection effect of A673T mutation may be driven by lower Aβ oligomer binding affinity. J Neurochem. 2020. 10.1111/jnc.15212. jnc.15212.
    1. van Dyck CH. Anti‐Amyloid‐β Monoclonal Antibodies for Alzheimer's Disease: pitfalls and Promise. Biol Psychiatry. 2018;83:311‐319. 10.1016/j.biopsych.2017.08.010.
    1. Viola KL, Klein WL. Amyloid beta oligomers in Alzheimer's disease pathogenesis, treatment, and diagnosis. Acta Neuropathol. 2015;129:183‐206. 10.1007/s00401-015-1386-3.
    1. Park J, Strittmatter S. Nogo receptor interacts with brain APP and Aβ to reduce pathologic changes in Alzheimers transgenic mice. Curr Alzheimer Res. 2007;4:568‐570. 10.2174/156720507783018235.
    1. Kim T, Vidal GS, Djurisic M, et al. Human LilrB2 is a β‐amyloid receptor and its murine homolog PirB regulates synaptic plasticity in an Alzheimer's model. Science. 2013;341:1399‐1404. 10.1126/science.1242077.
    1. Smith LM, Kostylev MA, Lee S, Strittmatter SM. Systematic and standardized comparison of reported amyloid‐β receptors for sufficiency, affinity, and Alzheimer's disease relevance. J Biol Chem. 2019. 10.1074/jbc.RA118.006252. jbc.RA118.006252.
    1. Shrestha BR, Vitolo OV, Joshi P, Lordkipanidze T, Shelanski M, Dunaevsky A. Amyloid beta peptide adversely affects spine number and motility in hippocampal neurons. Mol Cell Neurosci. 2006;33:274‐282. 10.1016/j.mcn.2006.07.011.
    1. Lacor PN, Buniel MC, Furlow PW, et al. Abeta oligomer‐induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer's disease. J Neurosci. 2007;27:796‐807. 10.1523/JNEUROSCI.3501-06.2007.
    1. Shankar GM, Li S, Mehta TH, et al. Amyloid‐beta protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat Med. 2008;14:837‐842. 10.1038/nm1782.
    1. Calabrese B, Shaked GM, Tabarean IV, Braga J, Koo EH, Halpain S. Rapid, concurrent alterations in pre‐ and postsynaptic structure induced by naturally‐secreted amyloid‐beta protein. Mol Cell Neurosci. 2007;35:183‐193. 10.1016/j.mcn.2007.02.006.
    1. Wang D, Govindaiah G, Liu R, De Arcangelis V, Cox CL, Xiang YK. Binding of amyloid beta peptide to beta2 adrenergic receptor induces PKA‐dependent AMPA receptor hyperactivity. FASEB J. 2010;24:3511‐3521. 10.1096/fj.10-156661.
    1. Lambert MP, Barlow AK, Chromy BA, et al. Diffusible, nonfibrillar ligands derived from Abeta1‐42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A. 1998;95:6448‐6453.
    1. Li S, Hong S, Shepardson NE, Walsh DM, Shankar GM, Selkoe D. Soluble oligomers of amyloid β protein facilitate hippocampal long‐term depression by disrupting neuronal glutamate uptake. Neuron. 2009;62:788‐801. 10.1016/j.neuron.2009.05.012.
    1. Wang Q, Walsh D, Rowan MJ, Selkoe DJ, Anwyl R. Block of long‐term potentiation by naturally secreted and synthetic amyloid‐peptide in hippocampal slices is mediated via activation of the kinases c‐Jun N‐terminal kinase, cyclin‐dependent kinase 5, and p38 mitogen‐activated protein kinase as well as Me. J Neurosci. 2004;24:3370‐3378. 10.1523/JNEUROSCI.1633-03.2004.
    1. Walsh DM, Klyubin I, Fadeeva JV, et al. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long‐term potentiation in vivo. Nature. 2002;416:535‐539. 10.1038/416535a.
    1. Walsh DM, Townsend M, Podlisny MB, et al. Certain inhibitors of synthetic amyloid beta‐peptide (Abeta) fibrillogenesis block oligomerization of natural Abeta and thereby rescue long‐term potentiation. J Neurosci. 2005;25:2455‐2462. 10.1523/JNEUROSCI.4391-04.2005.
    1. Klyubin I, Walsh DM, Lemere CA, et al. Amyloid β protein immunotherapy neutralizes Aβ oligomers that disrupt synaptic plasticity in vivo. Nat Med. 2005;11:556‐561. 10.1038/nm1234.
    1. Klyubin I, Betts V, Welzel AT, et al. Amyloid beta protein dimer‐containing human CSF disrupts synaptic plasticity: prevention by systemic passive immunization. J Neurosci. 2008;28:4231‐4237. 10.1523/JNEUROSCI.5161-07.2008.
    1. Puzzo D, Vitolo O, Trinchese F, Jacob JP, Palmeri A, Arancio O. Amyloid‐beta peptide inhibits activation of the nitric oxide/cGMP/cAMP‐responsive element‐binding protein pathway during hippocampal synaptic plasticity. J Neurosci. 2005;25:6887‐6897. 10.1523/JNEUROSCI.5291-04.2005.
    1. Puzzo D, Privitera L, Leznik E, et al. Picomolar amyloid‐beta positively modulates synaptic plasticity and memory in hippocampus. J Neurosci. 2008;28:14537‐14545. 10.1523/JNEUROSCI.2692-08.2008.
    1. Izzo NJ, Staniszewski A, To L, et al. Alzheimer's therapeutics targeting amyloid beta 1‐42 oligomers I: abeta 42 oligomer binding to specific neuronal receptors is displaced by drug candidates that improve cognitive deficits. PLoS One. 2014;9:e111898. 10.1371/journal.pone.0111898.
    1. Cleary JP, Walsh DM, Hofmeister JJ, et al. Natural oligomers of the amyloid‐beta protein specifically disrupt cognitive function. Nat Neurosci. 2005;8:79‐84. 10.1038/nn1372.
    1. Izzo NJ, Xu J, Zeng C, et al. Alzheimer's therapeutics targeting amyloid beta 1‐42 oligomers II: sigma‐2/PGRMC1 receptors mediate Abeta 42 oligomer binding and synaptotoxicity. PLoS One. 2014;9:e111899. 10.1371/journal.pone.0111899.
    1. Grundman M, Morgan R, Lickliter JD, et al. A phase 1 clinical trial of the sigma‐2 receptor complex allosteric antagonist CT1812, a novel therapeutic candidate for Alzheimer's disease. Alzheimer's Dement (New York, N Y). 2019;5:20‐26. 10.1016/j.trci.2018.11.001.
    1. Oyer HM, Sanders CM, Kim FJ. Small‐molecule modulators of sigma1 and sigma2/TMEM97 in the context of cancer: foundational concepts and emerging themes. Front Pharmacol. 2019;10:1141. 10.3389/fphar.2019.01141.
    1. Riad A, Zeng C, Weng C‐CC, et al. Sigma‐2 receptor/TMEM97 and PGRMC‐1 increase the rate of internalization of LDL by LDL receptor through the formation of a. Ternary Complex Sci Rep. 2018;8:16845. 10.1038/s41598-018-35430-3.
    1. Ahmed ISA, Chamberlain C, Craven RJ. S2R(Pgrmc1): the cytochrome‐related sigma‐2 receptor that regulates lipid and drug metabolism and hormone signaling. Expert Opin Drug Metab Toxicol. 2012;8:361‐370. 10.1517/17425255.2012.658367.
    1. Cahill MA. Progesterone receptor membrane component 1: an integrative review. J Steroid Biochem Mol Biol. 2007;105:16‐36. 10.1016/j.jsbmb.2007.02.002.
    1. Cahill MA, Jazayeri JA, Catalano SM, Toyokuni S, Kovacevic Z, Richardson DR. The emerging role of progesterone receptor membrane component 1 (PGRMC1) in cancer biology. Biochim Biophys Acta. 2016;1866:339‐349. 10.1016/j.bbcan.2016.07.004.
    1. Nguyen T, Su C, Singh M. Let‐7i inhibition enhances progesterone‐induced functional recovery in a mouse model of ischemia. Proc Natl Acad Sci U S A. 2018;115:E9668‐E9677. 10.1073/pnas.1803384115.
    1. Su C, Cunningham RL, Rybalchenko N, Singh M. Progesterone increases the release of brain‐derived neurotrophic factor from glia via progesterone receptor membrane component 1 (Pgrmc1)‐dependent ERK5 signaling. Endocrinology. 2012;153:4389‐4400. 10.1210/en.2011-2177.
    1. Izzo NJ, Colom‐Cadena M, Riad AA, et al. Proceedings from the Fourth International Symposium on sigma‐2 Receptors: Role in Health and Disease. ENeuro 2020:ENEURO.0317‐20.2020. 10.1523/ENEURO.0317-20.2020.
    1. Shankar GM, Bloodgood BL, Townsend M, Walsh DM, Selkoe DJ, Sabatini BL. Natural oligomers of the Alzheimer amyloid‐beta protein induce reversible synapse loss by modulating an NMDA‐type glutamate receptor‐dependent signaling pathway. J Neurosci. 2007;27:2866‐2875. 10.1523/JNEUROSCI.4970-06.2007.
    1. Scheff SW, Price DA, Schmitt FA, Mufson EJ. Hippocampal synaptic loss in early Alzheimer's disease and mild cognitive impairment. Neurobiol Aging. 2006;27:1372‐1384. 10.1016/j.neurobiolaging.2005.09.012.
    1. Yuede CM, Lee H, Restivo JL, et al. Rapid in vivo measurement of β‐amyloid reveals biphasic clearance kinetics in an Alzheimer's mouse model. J Exp Med. 2016;213:677‐685. 10.1084/jem.20151428.
    1. Kayed R, Head E, Thompson JL, et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003;300:486‐489. 10.1126/science.1079469.
    1. Savonenko A, Xu GM, Melnikova T, et al. Episodic‐like memory deficits in the APPswe/PS1dE9 mouse model of Alzheimer's disease: relationships to beta‐amyloid deposition and neurotransmitter abnormalities. Neurobiol Dis. 2005;18:602‐617. 10.1016/j.nbd.2004.10.022.
    1. Koffie RM, Meyer‐Luehmann M, Hashimoto T, et al. Oligomeric amyloid beta associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc Natl Acad Sci U S A. 2009;106:4012‐4017. 10.1073/pnas.0811698106.
    1. Koffie RM, Hashimoto T, Tai H‐C, et al. Apolipoprotein E4 effects in Alzheimer's disease are mediated by synaptotoxic oligomeric amyloid‐β. Brain. 2012;135:2155‐2168. 10.1093/brain/aws127.
    1. Faizi M, Bader PL, Saw N, et al. Thy1‐hAPP(Lond/Swe+) mouse model of Alzheimer's disease displays broad behavioral deficits in sensorimotor, cognitive and social function. Brain Behav. 2012;2:142‐154. 10.1002/brb3.41.
    1. Yi B, Sahn JJ, Ardestani PM, et al. Small molecule modulator of sigma 2 receptor is neuroprotective and reduces cognitive deficits and neuro‐inflammation in experimental models of Alzheimer's disease. J Neurochem. 2017;140:561‐575. 10.1111/jnc.13917.
    1. Zetterberg H, Blennow K. Neurogranin levels in cerebrospinal fluid: a new addition to the Alzheimer disease dagnostic toolbox. JAMA Neurol. 2015;72:1‐3. 10.1001/jamaneurol.2015.2075.
    1. Kvartsberg H, Lashley T, Murray CE, et al. The intact postsynaptic protein neurogranin is reduced in brain tissue from patients with familial and sporadic Alzheimer's disease. Acta Neuropathol. 2018. 10.1007/s00401-018-1910-3.
    1. Öhrfelt A, Brinkmalm A, Dumurgier J, et al. The pre‐synaptic vesicle protein synaptotagmin is a novel biomarker for Alzheimer's disease. Alzheimers Res Ther. 2016;8:41. 10.1186/s13195-016-0208-8.
    1. Willemse EAJ, De Vos A, Herries EM, et al. Neurogranin as cerebrospinal fluid biomarker for Alzheimer sisease: an assay comparison study. Clin Chem. 2018. 10.1373/clinchem.2017.283028. 000:clinchem.2017.283028.
    1. Portelius E, Zetterberg H, Skillbäck T, et al. Cerebrospinal fluid neurogranin: relation to cognition and neurodegeneration in Alzheimer's disease. Brain. 2015;138:3373‐3385. 10.1093/brain/awv267.
    1. Lleó A, Núñez‐Llaves R, Alcolea D, et al. Changes in synaptic proteins precede neurodegeneration markers in preclinical Alzheimer's disease cerebrospinal fluid. Mol Cell Proteomics. 2019;18:546‐560. 10.1074/mcp.RA118.001290.
    1. Higginbotham L, Ping L, Dammer E, et al. Integrated proteomics reveals brain‐based cerebrospinal fluid biomarkers in asymptomatic and symptomatic Alzheimer's disease (4738). Neurology. 2020;94:4738. 10.1101/806752.
    1. Noble W, Hanger DP, Miller CCJ, Lovestone S. The importance of tau phosphorylation for neurodegenerative diseases. Front Neurol. 2013;4:1‐11. 10.3389/fneur.2013.00083. JUL.
    1. Scheltens P, Blennow K, Breteler MMB, et al. Alzheimer's disease. Lancet (London, England). 2016;388:505‐517. 10.1016/S0140-6736(15)01124-1.
    1. Gomez‐Isla T. Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer's disease. Ann Neurol. 1997;41:17‐24.
    1. Serrano‐Pozo A, Frosch MP, Masliah E, Hyman BT. Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med. 2011;1:a006189. 10.1101/cshperspect.a006189.
    1. Liu C, Song X, Nisbet R, Götz J. Co‐immunoprecipitation with Tau isoform‐specific antibodies reveals distinct protein interactions and highlights a putative role for 2N Tau in disease. J Biol Chem. 2016;291:8173‐8188. 10.1074/jbc.M115.641902.
    1. Filtz TM, Vogel WK, Leid M. Regulation of transcription factor activity by interconnected, post‐translational modifications. Trends Pharmacol Sci. 2014;35:76‐85.
    1. Saito S, Yamaguchi H, Higashimoto Y, et al. Phosphorylation site interdependence of human p53 post‐translational modifications in response to stress. J Biol Chem. 2003;278:37536‐37544. 10.1074/jbc.M305135200.
    1. Russell CL, Mitra V, Hansson K, et al. Comprehensive quantitative profiling of tau and phosphorylated tau peptides in cerebrospinal fluid by mass spectrometry provides new biomarker candidates. J Alzheimer's Dis. 2016;55:303‐313. 10.3233/JAD-160633.
    1. Dashinimaev EB, Artyuhov AS, Bolshakov AP, Vorotelyak EA, Vasiliev AV. Neurons derived from induced pluripotent stem cells of patients with Down syndrome reproduce early stages of Alzheimer's disease type pathology in vitro. J Alzheimer's Dis. 2017;56:1‐13. 10.3233/JAD-160945.
    1. Blennow K, Zetterberg H, Rinne JO, et al. Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Arch Neurol. 2012;69:1002‐1010. 10.1001/archneurol.2012.90.
    1. Mormino EC, Papp KV. Amyloid accumulation and cognitive decline in clinically normal older individuals: implications for aging and early Alzheimer's disease. J Alzheimers Dis. 2018;64:S633‐S646.
    1. Kumar D, Ganeshpurkar A, Kumar D, Modi G, Gupta SK, Singh SK. Secretase inhibitors for the treatment of Alzheimer's disease: long road ahead. Eur J Med Chem. 2018;148:436‐452. 10.1016/j.ejmech.2018.02.035.
    1. Harper AR, Nayee S, Topol EJ. Protective alleles and modifier variants in human health and disease. Nat Rev Genet. 2015;16:689‐701. 10.1038/nrg4017.
    1. Yang T, Li S, Xu H, Walsh DM, Selkoe DJ. Large soluble oligomers of amyloid β‐Protein from Alzheimer brain are far less neuroactive than the smaller oligomers to which they dissociate. J Neurosci. 2017;37:152‐163. 10.1523/JNEUROSCI.1698-16.2017.
    1. Riad A, Lengyel‐Zhand Z, Zeng C, et al. The sigma‐2 receptor/TMEM97, PGRMC1, and LDL receptor complex are responsible for the cellular uptake of Aβ42 and its protein aggregates. Mol Neurobiol. 2020;57:3803‐3813. 10.1007/s12035-020-01988-1.
    1. Tarawneh R, D'Angelo G, Crimmins D, et al. Diagnostic and prognostic utility of the synaptic marker neurogranin in Alzheimer disease. JAMA Neurol. 2016;73:561‐571. 10.1001/jamaneurol.2016.0086.
    1. Blennow K, Zetterberg H. Biomarkers for Alzheimer disease ‐ current status and prospects for the future. J Intern Med. 2018:0‐2. 10.1111/joim.12816.
    1. Wellington H, Paterson RW, Portelius E, et al. Increased CSF neurogranin concentration is specific to Alzheimer disease. Neurology. 2016;86:829‐835. 10.1212/WNL.0000000000002423.
    1. Mairet‐Coello G, Courchet J, Pieraut S, et al. The CAMKK2‐AMPK kinase pathway mediates the synaptotoxic effects of Aβ oligomers through tau phosphorylation. Neuron. 2013;78:94‐108. 10.1016/j.neuron.2013.02.003.
    1. Crespo‐Biel N, Theunis C, Borghgraef P, et al. Phosphorylation of protein Tau by GSK3β prolongs survival of bigenic Tau.P301L × GSK3β mice by delaying brainstem tauopathy. Neurobiol Dis. 2014;67:119‐132.
    1. Llorens‐Martín M, Jurado J, Hernández F, Ávila J. GSK‐3β, a pivotal kinase in Alzheimer disease. Front Mol Neurosci. 2014.
    1. Mendoza J, Sekiya M, Taniguchi T, Iijima KM, Wang R, Ando K. Global analysis of phosphorylation of tau by the checkpoint kinases Chk1 and Chk2 in vitro. J Proteome Res. 2013;12:2654‐2665. 10.1021/pr400008f.
    1. Ittner A, Chua SW, Bertz J, et al. Site‐specific phosphorylation of tau inhibits amyloid‐β toxicity in Alzheimer's mice. Science (80‐). 2016;354:904‐908. 10.1126/science.aah6205.
    1. Barthélemy NR, Mallipeddi N, Moiseyev P, Sato C, Bateman RJ. Tau phosphorylation rates measured by mass spectrometry differ in the intracellular brain vs. extracellular cerebrospinal fluid compartments and are differentially affected by Alzheimer's disease. Front Aging Neurosci. 2019;11:1‐18. 10.3389/fnagi.2019.00121.
    1. Jarosz‐Griffiths HH, Noble E, Rushworth JV, Hooper NM. Amyloid‐β receptors: the good, the bad and the prion protein. J Biol Chem. 2015. 10.1074/jbc.R115.702704. jbc.R115.702704.
    1. Hsieh H, Boehm J, Sato C, et al. AMPAR removal underlies Abeta‐induced synaptic depression and dendritic spine loss. Neuron. 2006;52:831‐843. 10.1016/j.neuron.2006.10.035.
    1. Snyder EM, Nong Y, Almeida CG, et al. Regulation of NMDA receptor trafficking by amyloid‐β. Nat Neurosci. 2005;8:1051‐1058. 10.1038/nn1503.
    1. Ganapathy ME, Prasad PD, Huang W, Seth P, Leibach FH, Ganapathy V. Molecular and ligand‐binding characterization of the sigma‐receptor in the Jurkat human T lymphocyte cell line. J Pharmacol Exp Ther. 1999;289:251‐260.
    1. Klein WL. Abeta toxicity in Alzheimer's disease: globular oligomers (ADDLs) as new vaccine and drug targets. Neurochem Int. 2002;41:345‐352.
    1. Lambert MP, Viola KL, Chromy BA, et al. Vaccination with soluble Abeta oligomers generates toxicity‐neutralizing antibodies. J Neurochem. 2001;79:595‐605.
    1. Liu Y, Peterson DA, Schubert D. Amyloid beta peptide alters intracellular vesicle trafficking and cholesterol homeostasis. Proc Natl Acad Sci U S A. 1998;95:13266‐13271.
    1. McDonald JM, Cairns NJ, Taylor‐Reinwald L, Holtzman D, Walsh DM. The levels of water‐soluble and triton‐soluble Aβ are increased in Alzheimer's disease brain. Brain Res. 2012;1450:138‐147. 10.1016/j.brainres.2012.02.041.
    1. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671‐675.
    1. Armstrong‐James M, Millar J. Carbon fibre microelectrodes. J Neurosci Methods. 1979;1:279‐287.
    1. Portelius E, Olsson B, Höglund K, et al. Cerebrospinal fluid neurogranin concentration in neurodegeneration: relation to clinical phenotypes and neuropathology. Acta Neuropathol. 2018;136:363‐376. 10.1007/s00401-018-1851-x.
    1. Brinkmalm A, Brinkmalm G, Honer WG, et al. SNAP‐25 is a promising novel cerebrospinal fluid biomarker for synapse degeneration in Alzheimer's disease. Mol Neurodegener. 2014;9:53. 10.1186/1750-1326-9-53.
    1. Croisé P, Houy S, Gand M, et al. Cdc42 and Rac1 activity is reduced in human pheochromocytoma and correlates with FARP1 and ARHGEF1 expression. Endocr Relat Cancer. 2016;23:281‐293. 10.1530/ERC-15-0502.
    1. Russell CL, Heslegrave A, Mitra V, et al. Combined tissue and fluid proteomics with tandem mass tags to identify low‐abundance protein biomarkers of disease in peripheral body fluid: an Alzheimer's disease case study. Rapid Commun Mass Spectrom. 2017;31:153‐159. 10.1002/rcm.7777.
    1. Johnson ECB, Dammer EB, Duong DM, et al. Large‐scale proteomic analysis of Alzheimer's disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation. Nat Med. 2020. 10.1038/s41591-020-0815-6.
    1. Rankin CA, Sun Q, Gamblin TC. Tau phosphorylation by GSK‐3ß promotes tangle‐like filament morphology. Mol Neurodegener. 2007;2:1‐14. 10.1186/1750-1326-2-12.
    1. Salcedo‐Tello P, Ortiz‐Matamoros A, Arias C. GSK3 function in the brain during development, neuronal plasticity, and neurodegeneration. Int J Alzheimers Dis. 2011;2011:7‐10. 10.4061/2011/189728.

Source: PubMed

3
订阅