Preclinical and randomized clinical evaluation of the p38α kinase inhibitor neflamapimod for basal forebrain cholinergic degeneration

Ying Jiang, John J Alam, Stephen N Gomperts, Paul Maruff, Afina W Lemstra, Ursula A Germann, Philip H Stavrides, Sandipkumar Darji, Sandeep Malampati, James Peddy, Cynthia Bleiwas, Monika Pawlik, Anna Pensalfini, Dun-Sheng Yang, Shivakumar Subbanna, Balapal S Basavarajappa, John F Smiley, Amanda Gardner, Kelly Blackburn, Hui-May Chu, Niels D Prins, Charlotte E Teunissen, John E Harrison, Philip Scheltens, Ralph A Nixon, Ying Jiang, John J Alam, Stephen N Gomperts, Paul Maruff, Afina W Lemstra, Ursula A Germann, Philip H Stavrides, Sandipkumar Darji, Sandeep Malampati, James Peddy, Cynthia Bleiwas, Monika Pawlik, Anna Pensalfini, Dun-Sheng Yang, Shivakumar Subbanna, Balapal S Basavarajappa, John F Smiley, Amanda Gardner, Kelly Blackburn, Hui-May Chu, Niels D Prins, Charlotte E Teunissen, John E Harrison, Philip Scheltens, Ralph A Nixon

Abstract

The endosome-associated GTPase Rab5 is a central player in the molecular mechanisms leading to degeneration of basal forebrain cholinergic neurons (BFCN), a long-standing target for drug development. As p38α is a Rab5 activator, we hypothesized that inhibition of this kinase holds potential as an approach to treat diseases associated with BFCN loss. Herein, we report that neflamapimod (oral small molecule p38α inhibitor) reduces Rab5 activity, reverses endosomal pathology, and restores the numbers and morphology of BFCNs in a mouse model that develops BFCN degeneration. We also report on the results of an exploratory (hypothesis-generating) phase 2a randomized double-blind 16-week placebo-controlled clinical trial (Clinical trial registration: NCT04001517/EudraCT #2019-001566-15) of neflamapimod in mild-to-moderate dementia with Lewy bodies (DLB), a disease in which BFCN degeneration is an important driver of disease expression. A total of 91 participants, all receiving background cholinesterase inhibitor therapy, were randomized 1:1 between neflamapimod 40 mg or matching placebo capsules (taken orally twice-daily if weight <80 kg or thrice-daily if weight >80 kg). Neflamapimod does not show an effect in the clinical study on the primary endpoint, a cognitive-test battery. On two secondary endpoints, a measure of functional mobility and a dementia rating-scale, improvements were seen that are consistent with an effect on BFCN function. Neflamapimod treatment is well-tolerated with no study drug associated treatment discontinuations. The combined preclinical and clinical observations inform on the validity of the Rab5-based pathogenic model of cholinergic degeneration and provide a foundation for confirmatory (hypothesis-testing) clinical evaluation of neflamapimod in DLB.

Conflict of interest statement

J.J.A, A.G., and K.B. are employees of EIP Pharma, the sponsor of the clinical study. J.J.A. is also founder of and has stock ownership in EIP Pharma. U.A.G. receives compensation as a scientific consultant to EIP Pharma. S.N.G. has served on Advisory Boards of Jannsen, Acadia, and Sanofi, has received consulting fees from EIP Pharma, and has received funding from the NIH, the DOD CDMRP, the Michael J. Fox Foundation, the FFFPRI, and the Lewy Body Dementia Association. N.D.P. is CEO and co-owner of Brain Research Center. P.M. is a full-time employee at Cogstate Ltd. J.E.H. reports receipt of personal fees in the past 2 years from Actinogen, AlzeCure, Aptinyx, Astra Zeneca, Athira Therapeutics, Axon Neuroscience, Axovant, Bial Biotech, Biogen Idec, BlackThornRx, Boehringer Ingelheim, Brands2life, Cerecin, Cognito, Cognition Therapeutics, Compass Pathways, Corlieve, Curasen, EIP Pharma, Eisai, G4X Discovery, GfHEU, Heptares, Ki Elements, Lundbeck, Lysosome Therapeutics, MyCognition, Neurocentria, Neurocog, Neurodyn Inc, Neurotrack, the NHS, Novartis, Novo Nordisk, Nutricia, Probiodrug, Prothena, Recognify, Regeneron, reMYND, Rodin Therapeutics, Samumed, Sanofi, Signant, Syndesi Therapeutics, Takeda, Vivoryon Therapeutics and Winterlight Labs. In addition, he holds stock options in Neurotrack Inc. and is a joint holder of patents with My Cognition Ltd. C.E.T. has a collaboration contracts with ADx Neurosciences, Quanterix and Eli Lilly; performed contract research or received grants from AC-Immune, Axon Neurosciences, Bioconnect, Bioorchestra, Brainstorm Therapeutics, Celgene, EIP Pharma, Eisai, Grifols, Novo Nordisk, PeopleBio, Roche, Toyama, and Vivoryon; and has had speaker contracts for Roche, Grifols, and Novo Nordisk. P.S. has received consultancy fees (paid to the institution) from AC Immune, Brainstorm Cell, EIP Pharma, ImmunoBrain Checkpoint, Genentech, Novartis, Novo Nordisk. P.S. is also principal investigator of studies with AC Immune, FUJI-film/Toyama, UCB, and Vivoryon; and is an employee of EQT Life Sciences (formerly LSP). The remaining authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Normalized Rab5+ endosome number/size and…
Fig. 1. Normalized Rab5+ endosome number/size and restored cholinergic neuronal numbers in Ts2 mice treated with neflamapimod (NFMD).
a Representative images of medial septal nucleus (MSN) regions from vehicle-treated wildtype (2 N, n = 4), vehicle-treated Ts2 (n = 4) and NFMD-treated Ts2 (Ts2-NFMD) mice, labeled with Rab5-GTP (red) and Rab5a (green) antibodies; arrows point to the Rab5-positive neurons, which are shown enlarged in the lower panel (scale bar, 15 μm). b The numbers, sizes and areas of Rab5-GTP-positive endosomes determined by Image J analysis [n = 4 mice per group; for number: F(3,12) = 7.789, R square = 0.661; for 2 N vs Ts2 p = 0.0202, 95% CI = −1.880, −0.151; for Ts2 vs Ts2-NFMD p = 0.0097, 95% CI = 0.273, 2.002; for size: F(3,12) = 6.939, R square = 0.634, for 2 N vs Ts2 p = 0.0036, 95% CI = −1.329, −0.269; for area: F(3,12) = 13.19, R square = 0.767; for 2 N vs Ts2 p = 0.0009; 95% CI = −3.766, −1.079; for Ts2 vs Ts2-NFMD p = 0.0030, 95% CI = 0.7325, 3.419]. c The ratio of Rab5-GTP to total Rab5 intensities determined with Image J [n = 4 mice per group; F(3,12) = 13.23, R square = 0.768; for 2 N vs Ts2 p = 0.001, 95% CI = −0.537, −0.073; for Ts2 vs Ts2-NFMD p = 0.0002, 95% CI = 0.254, 0.719]. d The number of stereologically counted ChAT+ neurons in the MSN region of 2 N (n = 27) and Ts2 (n = 28) mice was graphed versus age of mice in months (Linear regression F = 10.16, R square = 0.281, p = 0.0037, 95% CI = −0.259, −0.056 for Ts2; F = 0.548, R square = 0.0215, p = 0.466, 95% CI = -infinity, −3.98) for 2N. e Representative images of diaminobenzidine (DAB)-stained ChAT+ neurons in the MSN region of 2 N and Ts2 mice treated with either vehicle or NFMD (scale bar, 100 μm). f Quantification of DAB-stained ChAT+ neurons in the MSN region; n = 10 (2 N), n = 11 (2N-NFMD), n = 14 (Ts2) and n = 11 (Ts2-NFMD) mice (F(3, 43) = 11.10, R square = 0.436, for 2 N vs Ts2, p < 0.0001, 95% CI = 0.588, 1.712; for Ts2 vs Ts2-NFMD p = 0.0037, 95% CI = −1.331, −207). g, h Abnormal morphology of ChAT+ neurons was consistently and reproducibly seen in the MSN of Ts2 mice, with swelling (yellow arrows), perikaryal atrophy (red arrows) and below normal ChAT immunoreactivity intensity (green arrows) in the representative images (scale bar,10 μm). Data are presented as mean values ± SEM. Graph made and analyzed with GraphPad Prism8.0.1 with Ordinary One-Way ANOVA/Tukey correction. Statistical significance is represented by asterisks *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. Source data are provided as a Source Data File.
Fig. 2. Slower decline of LTP and…
Fig. 2. Slower decline of LTP and normalized scores of behavioral tests in Ts2 mice treated with NFMD.
a Input/output relationship plots of hippocampal slices from 2 N and Ts2 mice treated with either vehicle or NFMD (n = 4 mice for Ts2 group and n = 5 mice for the other treatment groups), and graphed using GraphPad Prism8.0.1 [no significant difference between the slopes, Linear regression F (3, 40) = 0.107, p = 0.956]. b Plots of LTP in the Schaffer collateral synapses (CA3-CA1) induced by theta-burst stimulation (TBS) of mice from four treatment groups [Linear regression for, significant difference between the slopes of 2 N vs Ts2, F(1, 236) = 6.925, p = 0.0091; no significant difference between the slopes of Ts2 vs Ts2-NFMD, F(1, 236) = 2.103, p = 0.148]. c Averages of fEPSP slopes at 1, 50, and 110 min following tetanic stimulation; NFMD treatment increased the fEPSP slope of Ts2 mice by 18% at the 110 min time point [Ordinary One-Way ANOVA with Tukey correction, at 1 min time point, F(3, 34) = 20.75, R square = 0.647, for 2 N vs Ts2, p < 0.0001, 95% CI = 54.62, 139.5; at 50 min time point, F(3, 34) = 16.44, R square = 0.592, for 2 N vs Ts2, p < 0.0001, 95% CI = 49.54, 144.5; at 110 min time point, F(3, 34) = 19.00, R square = 0.626, for 2 N vs Ts2, p < 0.0001, 95% CI = 72.09, 139.6, for Ts2 vs Ts2-NFMD, p = 0.0334, 95% CI = −67.71, −2.938]. d Novel Object Recognition (NOR) test at 24 h after familiarization session, represented by recognition index in 2 N (n = 8) and Ts2 (n = 8) mice before (Pre) and after (Post) 4 weeks of NFMD treatments [Ordinary One-Way ANOVA with Tukey correction, F(3, 24) = 1.967, R square = 0.197, for Ts2-pre vs Ts2-post, p = 0.0262, 95% CI = 2.406, 34.97]. e Open field test results including speed, distance and percentage of time spent in thigmotaxis for 2 N (n = 8) and Ts2 mice (n = 8) Pre and Post 4 weeks of NFMD treatment [Ordinary One-Way ANOVA with Tukey correction, For Speed F(3, 22) = 5.936, R square = 0.447, for 2N-NFMD-pre vs Ts2-NFMD-pre, p = 0.0148, 95% CI = −36.97, −4.47; for Ts2-NFMD-pre vs Ts2-NFMD-post p = 0.0148, 95% CI = 3.572, 34.89; For Distance F(3, 22) = 9.784, R square = 0.572, for 2N-NFMD-pre vs Ts2-NFMD-pre, p = 0.0434, 95% CI = −27.75, −0.295; for Ts2-NFMD-pre vs Ts2-NFMD-post p = 0.0003, 95% CI = 9.205, 32.77). Data are presented as mean values ± SEM. Statistical significance represented by asterisks *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. Source data are provided as a Source Data File.
Fig. 3. The positive effects of neflamapimod…
Fig. 3. The positive effects of neflamapimod on pathology and function in Ts2 mice appear to be mediated by inhibition of the p38α kinase pathway and related to BACE1 and βCTF reductions.
a Western blot analysis of p38α, phosphorylated-p38 (p-p38), and downstream substrates MK2 and MNK1 in tissue homogenates of the brain cortex after 2 weeks of either vehicle or NFMD treatment (n = 5 for Ts2-NFMD, and n = 6 for other treatment groups). b Quantification of the western blot images shown with Image J and graphed with GraphPad Prism 8.0.1 [Ordinary One-Way ANOVA, For p-p38 F(3, 19) = 6.658, R square = 0.540, for 2 N vs Ts2, p = 0.0044, 95% CI = −0.760, −0.131; for 2 N vs Ts2-NFMD p = 0.0495, 95% CI = −0.656 −0.000194; For MK2 F(3, 19) = 10.33, R square = 0.610, for 2 N vs Ts2-NFMD, p = 0.0005, 95% CI = 0.182, 0.541; for Ts2 vs Ts2-NFMD p = 0.0166, 95% CI = 0.0457, 0.404; For pMK2 F(3, 19) = 2.850, R square = 0.310, for 2 N vs Ts2, p = 0.0178, 95% CI = −1.095, −0.117; for 2 N vs 2N-NFMD p = 0.0469, 95% CI = −0.985, −0.0076; for Ts2 vs Ts2-NFMD p = 0.0166, 95% CI = 0.0457, 0.404; For MNK1 F(3, 19) = 15.11, R square = 0.705, for 2 N vs Ts2, p = 0.0003, 95% CI = 0.235, 0.668; for 2 N vs 2N-NFMD p = 0.023, 95% CI = 0.0393, 0.472, for Ts2 vs Ts2-NFMD, p = 0.0347, 95% CI = 0.0197, 0.473; For pMNK1 F(3, 19) = 6.285, R square = 0.498, for 2 N vs Ts2-k NFMD, p = 0.0063, 95% CI = 0.156, 0.824; for TS2 vs Ts2-NFMD p = 0.0428, 95% CI = 0.125, 0.681]. c, d Subsequent western blot analysis for full length APP (APPfl), APP-βCTF and BACE1 and western blot image quantified with Image J and graphed with GraphPad Prism 8.0.1 (Ordinary One-Way ANOVA, For APPfl F(3, 19) = 23.33, R square = 0.786, for 2 N vs Ts2, p < 0.0001, 95% CI = −684, −0.376; for 2 N vs Ts2-NFMD p = 0.0001, 95% CI = −0.532, −0.209; For APP-βCTF F(3, 19) = 8.167, R square = 0.563, for 2 N vs Ts2, p = 0.0003, 95% CI = −1.027, −0.367; for Ts2 vs Ts2-NFMD p = 0.0007, 95% CI = 0.3187, 1.010; For BACE1 F(3, 19) = 12.86, R square = 0.670, for 2 N vs Ts2-NFMD, p = 0.0001, 95% CI = 0.158, 0.491; for Ts2 vs TS2-NFMD p = 0.0469, 95% CI = −0.00185, 0.335). Data are presented as mean values ± SEM. Statistical significance is represented by asterisks *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. Source data are provided as a Source Data File.
Fig. 4. CONSORT flow diagram for the…
Fig. 4. CONSORT flow diagram for the AscenD-LB Phase 2a clinical trial.
CONSORT 2010 Statement flow diagram showing participant flow through each stage of the AscenD-LB phase 2a randomized controlled trial from randomization through to follow-up and data analysis.

References

    1. Grothe MJ, et al. Atrophy of the cholinergic basal forebrain in dementia with Lewy bodies and Alzheimer’s disease dementia. J. Neurol. 2014;261:1939–1948. doi: 10.1007/s00415-014-7439-z.
    1. Duda JE. Pathology and neurotransmitter abnormalities of dementia with Lewy bodies. Dement Geriatr. Cogn. Disord. 2004;17:3–14. doi: 10.1159/000074677.
    1. Schumacher, J. et al. Cholinergic white matter pathways in dementia with Lewy bodies and Alzheimer’s disease. Brain (2021).
    1. Schumacher J, et al. In vivo nucleus basalis of Meynert degeneration in mild cognitive impairment with Lewy bodies. Neuroimage Clin. 2021;30:102604. doi: 10.1016/j.nicl.2021.102604.
    1. Schmitz TW, Spreng RN, The-Alzheimer’s-Disease-Neuroimaging-Initiative. Basal forebrain degeneration precedes and predicts the cortical spread of Alzheimer’s pathology. Nat. Commun. 2016;7:13249. doi: 10.1038/ncomms13249.
    1. Hampel H, et al. Revisiting the cholinergic hypothesis in alzheimer’s disease: emerging evidence from translational and clinical research. J. Prev. Alzheimers Dis. 2019;6:2–15.
    1. Fernandez-Cabello S, et al. Basal forebrain volume reliably predicts the cortical spread of Alzheimer’s degeneration. Brain. 2020;143:993–1009. doi: 10.1093/brain/awaa012.
    1. Wilson J, et al. Cholinergic basal forebrain volumes predict gait decline in Parkinson’s Disease. Mov. Disord. 2021;36:611–621. doi: 10.1002/mds.28453.
    1. Wilkins KB, Parker JE, Bronte-Stewart HM. Gait variability is linked to the atrophy of the Nucleus Basalis of Meynert and is resistant to STN DBS in Parkinson’s disease. Neurobiol. Dis. 2020;146:105134. doi: 10.1016/j.nbd.2020.105134.
    1. Dalrymple WA, et al. Cholinergic nucleus 4 atrophy and gait impairment in Parkinson’s disease. J. Neurol. 2021;268:95–101. doi: 10.1007/s00415-020-10111-2.
    1. Jarzebowski, P., Tang, C.S., Paulsen, O. & Hay, Y.A. Impaired spatial learning and suppression of sharp wave ripples by cholinergic activation at the goal location. Elife10, e65998 (2021).
    1. Nixon RA. Amyloid precursor protein and endosomal-lysosomal dysfunction in Alzheimer’s disease: inseparable partners in a multifactorial disease. FASEB J. 2017;31:2729–2743. doi: 10.1096/fj.201700359.
    1. Holtzman DM, et al. Developmental abnormalities and age-related neurodegeneration in a mouse model of Down syndrome. Proc. Natl Acad. Sci. USA. 1996;93:13333–13338. doi: 10.1073/pnas.93.23.13333.
    1. Kim S, et al. Evidence that the rab5 effector APPL1 mediates APP-betaCTF-induced dysfunction of endosomes in Down syndrome and Alzheimer’s disease. Mol. Psychiatry. 2016;21:707–716. doi: 10.1038/mp.2015.97.
    1. Salehi AH, et al. NRAGE, a novel MAGE protein, interacts with the p75 neurotrophin receptor and facilitates nerve growth factor-dependent apoptosis. Neuron. 2000;27:279–288. doi: 10.1016/S0896-6273(00)00036-2.
    1. Cataldo AM, et al. Endocytic pathway abnormalities precede amyloid beta deposition in sporadic Alzheimer’s disease and Down syndrome: differential effects of APOE genotype and presenilin mutations. Am. J. Pathol. 2000;157:277–286. doi: 10.1016/S0002-9440(10)64538-5.
    1. Cataldo AM, et al. Down syndrome fibroblast model of Alzheimer-related endosome pathology: accelerated endocytosis promotes late endocytic defects. Am. J. Pathol. 2008;173:370–384. doi: 10.2353/ajpath.2008.071053.
    1. Jiang Y, et al. Partial BACE1 reduction in a Down syndrome mouse model blocks Alzheimer-related endosomal anomalies and cholinergic neurodegeneration: role of APP-CTF. Neurobiol. Aging. 2016;39:90–98. doi: 10.1016/j.neurobiolaging.2015.11.013.
    1. Cooper JD, et al. Failed retrograde transport of NGF in a mouse model of Down’s syndrome: reversal of cholinergic neurodegenerative phenotypes following NGF infusion. Proc. Natl Acad. Sci. USA. 2001;98:10439–10444. doi: 10.1073/pnas.181219298.
    1. Chen XQ, Mobley WC. Exploring the pathogenesis of Alzheimer Disease in basal forebrain cholinergic neurons: converging insights from alternative hypotheses. Front Neurosci. 2019;13:446. doi: 10.3389/fnins.2019.00446.
    1. Cavalli V, et al. The stress-induced MAP kinase p38 regulates endocytic trafficking via the GDI:Rab5 complex. Mol. Cell. 2001;7:421–432. doi: 10.1016/S1097-2765(01)00189-7.
    1. Gibbs KL, et al. Inhibiting p38 MAPK alpha rescues axonal retrograde transport defects in a mouse model of ALS. Cell Death Dis. 2018;9:596. doi: 10.1038/s41419-018-0624-8.
    1. Alam JJ. Selective brain-targeted antagonism of p38 MAPKalpha reduces hippocampal IL-1beta levels and improves Morris water maze performance in aged rats. J. Alzheimers Dis. 2015;48:219–227. doi: 10.3233/JAD-150277.
    1. Alam J, Blackburn K, Patrick D. Neflamapimod: Clinical phase 2b-ready oral small molecule inhibitor of p38alpha to reverse synaptic dysfunction in early Alzheimer’s disease. J. Prev. Alzheimer’s Dis. 2017;4:273–278.
    1. Prins ND, et al. A phase 2 double-blind placebo-controlled 24-week treatment clinical study of the p38 alpha kinase inhibitor neflamapimod in mild Alzheimer’s disease. Alzheimer’s Res. Ther. 2021;13:106. doi: 10.1186/s13195-021-00843-2.
    1. Trempolec N, Dave-Coll N, Nebreda AR. SnapShot: p38 MAPK substrates. Cell. 2013;152:924–924.e921. doi: 10.1016/j.cell.2013.01.047.
    1. Lawson SK, Dobrikova EY, Shveygert M, Gromeier M. p38alpha mitogen-activated protein kinase depletion and repression of signal transduction to translation machinery by miR-124 and −128 in neurons. Mol. Cell Biol. 2013;33:127–135. doi: 10.1128/MCB.00695-12.
    1. Schnoder L, et al. Deficiency of neuronal p38alpha MAPK attenuates amyloid pathology in Alzheimer disease mouse and cell models through facilitating lysosomal degradation of BACE1. J. Biol. Chem. 2016;291:2067–2079. doi: 10.1074/jbc.M115.695916.
    1. McKeith IG, et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology. 2017;89:88–100. doi: 10.1212/WNL.0000000000004058.
    1. Thomas AJ, et al. Autopsy validation of 123I-FP-CIT dopaminergic neuroimaging for the diagnosis of DLB. Neurology. 2017;88:276–283. doi: 10.1212/WNL.0000000000003512.
    1. Wesnes KA, et al. Effects of rivastigmine on cognitive function in dementia with lewy bodies: a randomised placebo-controlled international study using the cognitive drug research computerised assessment system. Dement Geriatr. Cogn. Disord. 2002;13:183–192. doi: 10.1159/000048651.
    1. Patel, B. et al. Outcome measures for dementia with lewy body clinical trials: a review. Alzheimer Dis. Assoc. Disord.36, 64–72 (2021).
    1. McKeith IG, Wesnes KA, Perry E, Ferrara R. Hallucinations predict attentional improvements with rivastigmine in dementia with lewy bodies. Dement Geriatr. Cogn. Disord. 2004;18:94–100. doi: 10.1159/000077816.
    1. Niewiadomska G, Komorowski S, Baksalerska-Pazera M. Amelioration of cholinergic neurons dysfunction in aged rats depends on the continuous supply of NGF. Neurobiol. Aging. 2002;23:601–613. doi: 10.1016/S0197-4580(01)00345-1.
    1. Loy B, et al. p38alpha and p38beta mitogen-activated protein kinases determine cholinergic transdifferentiation of sympathetic neurons. J. Neurosci. 2011;31:12059–12067. doi: 10.1523/JNEUROSCI.0448-11.2011.
    1. Scali C, et al. The selective cyclooxygenase-2 inhibitor rofecoxib suppresses brain inflammation and protects cholinergic neurons from excitotoxic degeneration in vivo. Neuroscience. 2003;117:909–919. doi: 10.1016/S0306-4522(02)00839-4.
    1. Miyakawa T, Yamada M, Duttaroy A, Wess J. Hyperactivity and intact hippocampus-dependent learning in mice lacking the M1 muscarinic acetylcholine receptor. J. Neurosci. 2001;21:5239–5250. doi: 10.1523/JNEUROSCI.21-14-05239.2001.
    1. Valuskova P, Riljak V, Forczek ST, Farar V, Myslivecek J. Variability in the drug response of M4 muscarinic receptor knockout mice during day and night time. Front Pharm. 2019;10:237. doi: 10.3389/fphar.2019.00237.
    1. Pepeu, G., Scali, C. & Giovannini, M.G. Chapter 25. The role of cholinergic system in novel object recognition. in Handbook of Behavioral Neuroscience (eds. Ennaceur, A. & deSouza Silva, M.) 371–378 (Elsevier, Amsterdam, 2018).
    1. Kaur G, et al. Glutamatergic transmission aberration: a major cause of behavioral deficits in a murine model of Down’s syndrome. J. Neurosci. 2014;34:5099–5106. doi: 10.1523/JNEUROSCI.5338-13.2014.
    1. Kurt MA, Kafa MI, Dierssen M, Davies DC. Deficits of neuronal density in CA1 and synaptic density in the dentate gyrus, CA3 and CA1, in a mouse model of Down syndrome. Brain Res. 2004;1022:101–109. doi: 10.1016/j.brainres.2004.06.075.
    1. Dickinson-Anson H, et al. Acetylcholine-secreting cells improve age-induced memory deficits. Mol. Ther. 2003;8:51–61. doi: 10.1016/S1525-0016(03)00145-X.
    1. Alam JJ, Krakovsky M, Germann U, Levy A. Continuous administration of a p38alpha inhibitor during the subacute phase after transient ischemia-induced stroke in the rat promotes dose-dependent functional recovery accompanied by increase in brain BDNF protein level. PLoS ONE. 2020;15:e0233073. doi: 10.1371/journal.pone.0233073.
    1. Conner JM, Chiba AA, Tuszynski MH. The basal forebrain cholinergic system is essential for cortical plasticity and functional recovery following brain injury. Neuron. 2005;46:173–179. doi: 10.1016/j.neuron.2005.03.003.
    1. Pensalfini A, et al. Endosomal dysfunction induced by directly overactivating rab5 recapitulates prodromal and neurodegenerative features of Alzheimer’s Disease. Cell Rep. 2020;33:108420. doi: 10.1016/j.celrep.2020.108420.
    1. Ge B, et al. MAPKK-independent activation of p38alpha mediated by TAB1-dependent autophosphorylation of p38alpha. Science. 2002;295:1291–1294. doi: 10.1126/science.1067289.
    1. Alam J, Scheper W. Targeting neuronal MAPK14/p38alpha activity to modulate autophagy in the Alzheimer disease brain. Autophagy. 2016;12:2516–2520. doi: 10.1080/15548627.2016.1238555.
    1. Colie S, et al. Neuronal p38alpha mediates synaptic and cognitive dysfunction in an Alzheimer’s mouse model by controlling beta-amyloid production. Sci. Rep. 2017;7:45306. doi: 10.1038/srep45306.
    1. Schnoder L, et al. P38alpha-MAPK phosphorylates Snapin and reduces Snapin-mediated BACE1 transportation in APP-transgenic mice. FASEB J. 2021;35:e21691. doi: 10.1096/fj.202100017R.
    1. Kanel P, et al. Topography of cholinergic changes in dementia with lewy bodies and key neural network hubs. J. Neuropsychiatry Clin. Neurosci. 2020;32:370–375. doi: 10.1176/appi.neuropsych.19070165.
    1. Muller P, et al. Structural MRI of the basal forebrain as predictor of cognitive response to galantamine in healthy older adults-A randomized controlled double-blinded crossover study. Alzheimers Dement (N.Y.) 2021;7:e12153.
    1. Avram M, et al. Lower cholinergic basal forebrain volumes link with cognitive difficulties in schizophrenia. Neuropsychopharmacology. 2021;46:2320–2329. doi: 10.1038/s41386-021-01070-x.
    1. Oswal A, et al. Cortical connectivity of the nucleus basalis of Meynert in Parkinson’s disease and Lewy body dementias. Brain. 2021;144:781–788. doi: 10.1093/brain/awaa411.
    1. Matsunaga, S., Kishi, T., Yasue, I. & Iwata, N. Cholinesterase inhibitors for lewy body disorders: a meta-analysis. Int. J. Neuropsychopharmacol19, 10.1093/ijnp/pyv086 (2015).
    1. Yoo, H.S. et al. Association of beta-amyloid and basal forebrain with cortical thickness and cognition in alzheimer and lewy body disease spectra. Neurology, 98 e947–e957 (2021).
    1. Hall S, et al. Plasma phospho-tau identifies Alzheimer’s co-pathology in patients with lewy body disease. Mov. Disord. 2021;36:767–771. doi: 10.1002/mds.28370.
    1. Kantarci K, et al. Longitudinal atrophy in prodromal dementia with Lewy bodies points to cholinergic degeneration. Brain Commun. 2022;4:fcac013. doi: 10.1093/braincomms/fcac013.
    1. Asih PR, et al. Functions of p38 MAP kinases in the Central Nervous System. Front Mol. Neurosci. 2020;13:570586. doi: 10.3389/fnmol.2020.570586.
    1. Bachstetter AD, Van Eldik LJ. The p38 MAP kinase family as regulators of proinflammatory cytokine production in degenerative diseases of the CNS. Aging Dis. 2010;1:199–211.
    1. Bracco L, Bessi V, Padiglioni S, Marini S, Pepeu G. Do cholinesterase inhibitors act primarily on attention deficit? A naturalistic study in Alzheimer’s disease patients. J. Alzheimers Dis. 2014;40:737–742. doi: 10.3233/JAD-131154.
    1. Teipel SJ, et al. Predictors of cognitive decline and treatment response in a clinical trial on suspected prodromal Alzheimer’s disease. Neuropharmacology. 2016;108:128–135. doi: 10.1016/j.neuropharm.2016.02.005.
    1. Edmonds EC, et al. Unmasking the benefits of donepezil via psychometrically precise identification of mild cognitive impairment: a secondary analysis of the ADCS vitamin E and donepezil in MCI study. Alzheimers Dement (N.Y.) 2018;4:11–18. doi: 10.1016/j.trci.2017.11.001.
    1. Mendez MF. Early-onset Alzheimer disease and its variants. Contin. (Minneap. Minn.) 2019;25:34–51.
    1. Al-Shaikh FSH, et al. Selective vulnerability of the nucleus basalis of Meynert among neuropathic subtypes of Alzheimer disease. JAMA Neurol. 2021;77:225–233. doi: 10.1001/jamaneurol.2019.3606.
    1. Martinez JL, Zammit MD, West NR, Christian BT, Bhattacharyya A. Basal forebrain cholinergic neurons: linking down syndrome and Alzheimer’s disease. Front Aging Neurosci. 2021;13:703876. doi: 10.3389/fnagi.2021.703876.
    1. Fang F, et al. Synuclein impairs trafficking and signaling of BDNF in a mouse model of Parkinson’s disease. Sci. Rep. 2017;7:3868. doi: 10.1038/s41598-017-04232-4.
    1. Spencer B, et al. Anti-alpha-synuclein immunotherapy reduces alpha-synuclein propagation in the axon and degeneration in a combined viral vector and transgenic model of synucleinopathy. Acta Neuropathol. Commun. 2017;5:7. doi: 10.1186/s40478-016-0410-8.
    1. Bassil F, et al. Amyloid-beta (abeta) plaques promote seeding and spreading of alpha-synuclein and tau in a mouse model of lewy body disorders with abeta pathology. Neuron. 2020;105:260–275 e266. doi: 10.1016/j.neuron.2019.10.010.
    1. Mandler M, et al. Effects of single and combined immunotherapy approach targeting amyloid beta protein and alpha-synuclein in a dementia with Lewy bodies-like model. Alzheimers Dement. 2019;15:1133–1148. doi: 10.1016/j.jalz.2019.02.002.
    1. Cataldo AM, et al. App gene dosage modulates endosomal abnormalities of Alzheimer’s disease in a segmental trisomy 16 mouse model of down syndrome. J. Neurosci. 2003;23:6788–6792. doi: 10.1523/JNEUROSCI.23-17-06788.2003.
    1. Jiang Y, et al. Alzheimer’s-related endosome dysfunction in Down syndrome is Abeta-independent but requires APP and is reversed by BACE-1 inhibition. Proc. Natl Acad. Sci. USA. 2010;107:1630–1635. doi: 10.1073/pnas.0908953107.
    1. Jiang Y, et al. Lysosomal dysfunction in down syndrome is app-dependent and mediated by APP-betaCTF (C99) J. Neurosci. 2019;39:5255–5268. doi: 10.1523/JNEUROSCI.0578-19.2019.
    1. West MJ, Slomianka L, Gundersen HJ. Unbiased stereological estimation of the total number of neurons in thesubdivisions of the rat hippocampus using the optical fractionator. Anat. Rec. 1991;231:482–497. doi: 10.1002/ar.1092310411.
    1. Seibenhener, M.L. & Wooten, M.C. Use of the Open Field Maze to measure locomotor and anxiety-like behavior in mice. J. Vis. Exp.96, e52434 (2015).
    1. Hanninen L, Pastell M. CowLog: open-source software for coding behaviors from digital video. Behav. Res Methods. 2009;41:472–476. doi: 10.3758/BRM.41.2.472.
    1. Rodriguez A, et al. ToxTrac: a fast and robust software for tracking organisms. Methods Ecol. Evolution. 2018;9:460–464. doi: 10.1111/2041-210X.12874.

Source: PubMed

3
購読する