A Lipid Rafts Theory of Alzheimer's Disease (2310.20232v2)
Abstract: I present a theory of Alzheimer's Disease (AD) that explains its symptoms, pathology, and risk factors. To do this, I introduce a new theory of brain plasticity that elucidates the physiological roles of AD-related agents. New events generate synaptic and branching candidates competing for long-term enhancement. Competition resolution crucially depends on the formation of membrane lipid rafts, which requires astrocyte-produced cholesterol. Sporadic AD is caused by impaired formation of plasma membrane lipid rafts, which prevents the conversion of short- to long-term memory, and yields excessive tau phosphorylation, intracellular cholesterol accumulation, synaptic dysfunction, and neurodegeneration. Amyloid beta (Abeta) production is promoted by cholesterol during the switch to competition resolution, and cholesterol accumulation stimulates chronic Abeta production, secretion, and aggregation. The theory addresses all of the major established facts known about the disease, and is supported by strong evidence.
- Alzheimer’s Association. 2023 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia. 2023;19(4):1598–1695.
- History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal transduction and targeted therapy. 2019;4(1):29.
- Questions concerning the role of amyloid-β𝛽\betaitalic_β in the definition, aetiology and diagnosis of Alzheimer’s disease. Acta Neuropathologica. 2018;136(5):663–689.
- Murphy C. Olfactory and other sensory impairments in Alzheimer disease. Nature Reviews Neurology. 2019;15(1):11–24.
- Tau seeding activity begins in the transentorhinal/entorhinal regions and anticipates phospho-tau pathology in Alzheimer’s disease and PART. Acta neuropathologica. 2018;136(1):57–67.
- Area-Gomez E, Schon EA. Alzheimer disease. Organelle Contact Sites: From Molecular Mechanism to Disease. 2017;p. 149–156.
- Crews L, Masliah E. Molecular mechanisms of neurodegeneration in Alzheimer’s disease. Human molecular genetics. 2010;19(R1):R12–R20.
- Is plasticity of synapses the mechanism of long-term memory storage? NPJ science of learning. 2019;4(1):1–10.
- Stein IS, Zito K. Dendritic spine elimination: molecular mechanisms and implications. The Neuroscientist. 2019;25(1):27–47.
- Dopamine receptors–IUPHAR review 13. British J Pharmacol. 2015;172(1):1–23.
- Barnes NM, Sharp T. A review of central 5-HT receptors and their function. Neuropharmacol. 1999;38(8):1083–1152.
- Bednarek E, Caroni P. β𝛽\betaitalic_β-Adducin is required for stable assembly of new synapses and improved memory upon environmental enrichment. Neuron. 2011;69(6):1132–1146.
- Transcellular nanoalignment of synaptic function. Neuron. 2017;96(3):680–696.
- Phosphorylation of the AMPA receptor subunit GluR2 differentially regulates its interaction with PDZ domain-containing proteins. Journal of Neuroscience. 2000;20(19):7258–7267.
- The roles of perineuronal nets and the perinodal extracellular matrix in neuronal function. Nature Reviews Neuroscience. 2019;20(8):451–465.
- Role of the endocytosis of caveolae in intracellular signaling and metabolism. Endocytosis and Signaling. 2018;p. 203–234.
- Prominent postsynaptic and dendritic exocytosis of endogenous BDNF vesicles in BDNF-GFP knock-in mice. Molecular neurobiology. 2019;56:6833–6855.
- The story of an exceptional serine protease, tissue-type plasminogen activator (tPA). Revue neurologique. 2016;172(3):186–197.
- Knapska E, Kaczmarek L. Matrix Metalloproteinase 9 (MMP-9) in Learning and Memory. In: Novel Mechanisms of Memory. Springer; 2016. p. 161–181.
- It’s all about tau. Progress in neurobiology. 2019;175:54–76.
- Sustained MAPK/ERK activation in adult Schwann cells impairs nerve repair. Journal of Neuroscience. 2018;38(3):679–690.
- Atorvastatin protects from Aβ𝛽\betaitalic_β 1–40-induced cell damage and depressive-like behavior via ProBDNF cleavage. Molecular neurobiology. 2017;54:6163–6173.
- Cholesterol-dependent modulation of tau phosphorylation in cultured neurons. J neurochem. 2001;76(2):391–400.
- NGF-induced axon growth is mediated by localized inactivation of GSK-3β𝛽\betaitalic_β and functions of the microtubule plus end binding protein APC. Neuron. 2004;42(6):897–912.
- Brain-derived neurotrophic factor-dependent unmasking of “silent” synapses in the developing mouse barrel cortex. Proceedings of the National Academy of Sciences. 2003;100(22):13069–13074.
- Park H, Poo Mm. Neurotrophin regulation of neural circuit development and function. Nat Rev Neurosci. 2013;14(1):7–23.
- Nicoll RA, Schulman H. Synaptic memory and CaMKII: a review. Physiological Reviews;103(4):2877–2925.
- Armijo-Weingart L, Gallo G. It takes a village to raise a branch: cellular mechanisms of the initiation of axon collateral branches. Molecular and Cellular Neuroscience. 2017;84:36–47.
- Henley JM, Wilkinson KA. AMPA receptor trafficking and the mechanisms underlying synaptic plasticity and cognitive aging. Dialogues in clinical neuroscience. 2013;15(1):11–27.
- Ras and Rap control AMPA receptor trafficking during synaptic plasticity. Cell. 2002;110(4):443–455.
- AMPA receptors and their minions: auxiliary proteins in AMPA receptor trafficking. Cellular and Molecular Life Sciences. 2019;76:2133–2169.
- Synaptic incorporation of AMPA receptors during LTP is controlled by a PKC phosphorylation site on GluR1. Neuron. 2006;51(2):213–225.
- Pilpel Y, Segal M. Activation of PKC induces rapid morphological plasticity in dendrites of hippocampal neurons via Rac and Rho-dependent mechanisms. European J Neurosci. 2004;19(12):3151–3164.
- Long-range inhibitory intersection of a retrosplenial thalamocortical circuit by apical tuft-targeting CA1 neurons. Nat Neurosci. 2019;22(4):618–626.
- ApoER2 processing by presenilin-1 modulates reelin expression. FASEB J. 2014;28(4):1543–1554.
- Reelin signaling in development, maintenance, and plasticity of neural networks. Ageing Res Rev. 2013;12(3):815–822.
- Van Der Kant R, Goldstein LS. Cellular functions of the amyloid precursor protein from development to dementia. Developmental cell. 2015;32(4):502–515.
- Ge YW, Lahiri D. Regulation of promoter activity of the APP gene by cytokines and growth factors: implications in Alzheimer’s disease. Annals of the New York Academy of Sciences. 2002;973(1):463–467.
- ApoE2, ApoE3, and ApoE4 differentially stimulate APP transcription and Aβ𝛽\betaitalic_β secretion. Cell. 2017;168(3):427–441.
- Exercise and BDNF reduce Aβ𝛽\betaitalic_β production by enhancing α𝛼\alphaitalic_α-secretase processing of APP. J Neurochem. 2017;142(2):286–296.
- Enhancement of the nonamyloidogenic pathway by exogenous NGF in an Alzheimer transgenic mouse model. Neuropeptides. 2014;48(4):233–238.
- Brain plasmin enhances APP α𝛼\alphaitalic_α-cleavage and Aβ𝛽\betaitalic_β degradation and is reduced in Alzheimer’s disease brains. EMBO reports. 2000;1(6):530–535.
- Matrix metalloproteinase-9 participates in NGF-induced α𝛼\alphaitalic_α-secretase cleavage of amyloid-β𝛽\betaitalic_β protein precursor in PC12 cells. J Alzheimer’s Disease. 2011;24(4):705–719.
- Processing of Alzheimer beta/A4 amyloid precursor protein: modulation by agents that regulate protein phosphorylation. Proc Natl Acad Sci U S A. 1990;87(15):6003–6006.
- Low cholesterol stimulates the nonamyloidogenic pathway by its effect on the α𝛼\alphaitalic_α-secretase ADAM 10. Proc Natl Acad Sci U S A. 2001;98(10):5815–5820.
- Amyloidogenic processing of the Alzheimer β𝛽\betaitalic_β-amyloid precursor protein depends on lipid rafts. J Cell Biol. 2003;160(1):113–123.
- Amyloid precursor protein α𝛼\alphaitalic_α-and β𝛽\betaitalic_β-cleaved ectodomains exert opposing control of cholesterol homeostasis via SREBP2. FASEB J. 2014;28(2):849–860.
- Age-dependent accumulation of soluble amyloid β𝛽\betaitalic_β (Aβ𝛽\betaitalic_β) oligomers reverses the neuroprotective effect of soluble amyloid precursor protein-α𝛼\alphaitalic_α (sAPPα𝛼\alphaitalic_α) by modulating phosphatidylinositol 3-kinase (PI3K)/Akt-GSK-3β𝛽\betaitalic_β pathway in Alzheimer mouse model. J Biological Chem. 2011;286(21):18414–18425.
- Insulin inhibits Abeta production through modulation of APP processing in a cellular model of Alzheimer’s disease. Neuroendocrinol Lett. 2014;35:224–229.
- Secreted amyloid precursor protein-alpha enhances LTP through the synthesis and trafficking of Ca2+-permeable AMPA receptors. Frontiers in Molecular Neuroscience. 2021;14:660208.
- Acute function of secreted amyloid precursor protein fragment APPsα𝛼\alphaitalic_α in synaptic plasticity. Acta neuropathologica. 2015;129(1):21–37.
- The physiological role of the amyloid precursor protein as an adhesion molecule in the developing nervous system. J Neurochem. 2017;143(1):11–29.
- Soluble amyloid precursor protein-α𝛼\alphaitalic_α modulates β𝛽\betaitalic_β-secretase activity and amyloid-β𝛽\betaitalic_β generation. Nat Comm. 2012;3(1):1–9.
- Brain-derived neurotrophic factor reduces amyloidogenic processing through control of SORLA gene expression. J Neurosci. 2009;29(49):15472–15478.
- Gugustea R, Jia Z. Genetic manipulations of AMPA glutamate receptors in hippocampal synaptic plasticity. Neuropharmacology. 2021;194:108630.
- Brain cholesterol in normal and pathological aging. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 2010;1801(8):934–944.
- Brain-derived neurotrophic factor modulates cholesterol homeostasis and Apolipoprotein E synthesis in human cell models of astrocytes and neurons. J Cellular Physiol. 2018;233(9):6925–6943.
- Map kinase and PKC signaling pathways modulate NGF-mediated apoE transcription. Neurosci Letters. 2015;595:54–59.
- Activation of estrogen receptor α𝛼\alphaitalic_α increases and estrogen receptor β𝛽\betaitalic_β decreases apolipoprotein E expression in hippocampus in vitro and in vivo. Proc Natl Acad Sci U S A. 2006;103(45):16983–16988.
- Mahley RW, Huang Y. Apolipoprotein E sets the stage: response to injury triggers neuropathology. Neuron. 2012;76(5):871–885.
- Shimano H, Sato R. SREBP-regulated lipid metabolism: convergent physiology–divergent pathophysiology. Nat Rev Endocrinol. 2017;13(12):710.
- Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. 2019;15(9):501–518.
- Better memory and neural efficiency in young apolipoprotein E ε𝜀\varepsilonitalic_ε4 carriers. Cerebral cortex. 2007;17(8):1934–1947.
- The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nature reviews Molecular cell biology. 2017;18(6):361–374.
- Interaction of membrane/lipid rafts with the cytoskeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal arrangement and cell signaling. Biochimica et Biophysica Acta (BBA)-Biomembranes. 2014;1838(2):532–545.
- Membrane raft disruption results in neuritic retraction prior to neuronal death in cortical neurons. Bioscience trends. 2012;6(4):183–191.
- Palmitoylated Proteins in Dendritic Spine Remodeling. Front Synaptic Neurosci. 2020;12.
- Pike LJ. Growth factor receptors, lipid rafts and caveolae: an evolving story. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2005;1746(3):260–273.
- The role of cholesterol in the activation of nicotinic acetylcholine receptors. In: Current topics in membranes. vol. 80. Elsevier; 2017. p. 95–137.
- CNS synaptogenesis promoted by glia-derived cholesterol. Science. 2001;294(5545):1354–1357.
- Lipid rafts in the maintenance of synapses, dendritic spines, and surface AMPA receptor stability. J Neurosci. 2003;23(8):3262–3271.
- Cholesterol depletion inhibits synaptic transmission and synaptic plasticity in rat hippocampus. Exp Neurol. 2008;212(2):407–414.
- Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. The EMBO journal. 2012;31(21):4106–4123.
- Waugh MG. Raft-like membranes from the trans-Golgi network and endosomal compartments. Nature Protocols. 2013;8(12):2429–2439.
- Chowdhury D, Hell JW. Ca2+/calmodulin binding to PSD-95 downregulates its palmitoylation and ampars in long-term depression. Frontiers in Synaptic Neuroscience. 2019;11:6.
- Diversity in NMDA receptor composition: many regulators, many consequences. The Neuroscientist. 2013;19(1):62–75.
- Transient incorporation of native GluR2-lacking AMPA receptors during hippocampal long-term potentiation. Nature neuroscience. 2006;9(5):602–604.
- Tau phosphorylation and tau mislocalization mediate soluble Aβ𝛽\betaitalic_β oligomer-induced AMPA glutamate receptor signaling deficits. European Journal of Neuroscience. 2014;39(7):1214–1224.
- Regulation of synaptic nanodomain by liquid–liquid phase separation: A novel mechanism of synaptic plasticity. Current Opinion in Neurobiology. 2021;69:84–92.
- Spike Timing Dependent Plasticity: A Consequence of More Fundamental Learning Rules. Frontiers in Computational Neuroscience. 2010;4:19.
- Reciprocal activation within a kinase-effector complex underlying persistence of structural LTP. Neuron. 2019;102(6):1199–1210.
- Sacktor TC, Fenton AA. What does LTP tell us about the roles of CaMKII and PKMζ𝜁\zetaitalic_ζ in memory? Molecular brain. 2018;11:1–9.
- Membrane lipid rafts and neurobiology: age-related changes in membrane lipids and loss of neuronal function. J Physiol. 2016;594(16):4565–4579.
- Bakota L, Brandt R. Why kiss-and-hop explains that tau does not stabilize microtubules and does not interfere with axonal transport (at physiological conditions). Cytoskeleton;.
- Multivalent cross-linking of actin filaments and microtubules through the microtubule-associated protein Tau. Nature communications. 2017;8(1):1981.
- Gong CX, Iqbal K. Hyperphosphorylation of microtubule-associated protein tau: a promising therapeutic target for Alzheimer disease. Current medicinal chemistry. 2008;15(23):2321–2328.
- Leucine carboxyl methyltransferase 1 (LCMT1)-dependent methylation regulates the association of protein phosphatase 2A and Tau protein with plasma membrane microdomains in neuroblastoma cells. J Biological Chem. 2013;288(38):27396–27405.
- Neurotrophin receptor p75 mediates amyloid β𝛽\betaitalic_β-induced tau pathology. Neurobiol of Disease. 2019;132:104567.
- DHCR24 knock-down induced tau hyperphosphorylation at Thr181, Ser199, Thr231, Ser262, Ser396 epitopes and inhibition of autophagy by overactivation of GSK3β𝛽\betaitalic_β/mTOR signaling. Frontiers in Aging Neuroscience. 2021;13:513605.
- The cell biology of tau secretion. Frontiers in Molecular Neuroscience. 2020;13:569818.
- Cholesterol depletion inhibits the generation of β𝛽\betaitalic_β-amyloid in hippocampal neurons. Proc Natl Acad Sci U S A. 1998;95(11):6460–6464.
- Independent inhibition of Alzheimer disease β𝛽\betaitalic_β-and γ𝛾\gammaitalic_γ-secretase cleavage by lowered cholesterol levels. Journal of Biological Chemistry. 2008;283(17):11302–11311.
- Regulation of cholesterol and sphingomyelin metabolism by amyloid-β𝛽\betaitalic_β and presenilin. Nat cell biol. 2005;7(11):1118–1123.
- Preferred endocytosis of amyloid precursor protein from cholesterol-enriched lipid raft microdomains. Molecules. 2020;25(23):5490.
- A TrkA-to-p75NTR molecular switch activates amyloid β𝛽\betaitalic_β-peptide generation during aging. Biochemical Journal. 2005;391(1):59–67.
- Amyloid-β𝛽\betaitalic_β 42 activates the expression of BACE1 through the JNK pathway. Journal of Alzheimer’s Disease. 2011;27(4):871–883.
- AMPAR removal underlies Aβ𝛽\betaitalic_β-induced synaptic depression and dendritic spine loss. Neuron. 2006;52(5):831–843.
- Amyloid β𝛽\betaitalic_β-induced nerve growth factor dysmetabolism in Alzheimer disease. J Neuropathol Exp Neurol. 2009;68(8):857–869.
- Knafo S, Esteban JA. PTEN: local and global modulation of neuronal function in health and disease. Trends Neurosci. 2017;40(2):83–91.
- The β𝛽\betaitalic_β-amyloid protein of Alzheimer’s disease increases neuronal CRMP-2 phosphorylation by a Rho-GTP mechanism. Brain. 2008;131(1):90–108.
- β𝛽\betaitalic_β-Amyloid regulation of presynaptic nicotinic receptors in rat hippocampus and neocortex. J Neurosci. 2003;23(17):6740–6747.
- Towards understanding the roles of heparan sulfate proteoglycans in Alzheimer’s disease. BioMed research international;2014.
- Interaction of Alzheimer’s β𝛽\betaitalic_β-amyloid peptides with cholesterol: mechanistic insights into amyloid pore formation. Biochemistry. 2014;53(28):4489–4502.
- Picomolar amyloid-β𝛽\betaitalic_β positively modulates synaptic plasticity and memory in hippocampus. Journal of Neuroscience. 2008;28(53):14537–14545.
- Finnie PS, Nader K. Amyloid beta secreted during consolidation prevents memory malleability. Curr Biol. 2020;30(10):1934–1940.
- Rasch B, Born J. About sleep’s role in memory. Physiol Rev. 2013;93(2):681–766.
- Lipid Dys-Homeostasis Contributes to APOE4-Associated AD Pathology. Cells. 2022;11(22):3616.
- Loss of astrocyte cholesterol synthesis disrupts neuronal function and alters whole-body metabolism. Proc Natl Acad Sci U S A. 2017;114(5):1189–1194.
- Stewart KL, Radford SE. Amyloid plaques beyond Aβ𝛽\betaitalic_β: a survey of the diverse modulators of amyloid aggregation. Biophysical Rev. 2017;9(4):405–419.
- Tau binds to lipid membrane surfaces via short amphipathic helices located in its microtubule-binding repeats. Biophysical J. 2014;107(6):1441–1452.
- Longitudinal tau PET in ageing and Alzheimer’s disease. Brain. 2018;141(5):1517–1528.
- Sex differences in Alzheimer disease – the gateway to precision medicine. Nat Rev Neurol. 2018;14(8):457–469.
- Autosomal-dominant Alzheimer’s disease: a review and proposal for the prevention of Alzheimer’s disease. Alzheimer’s research & therapy. 2011;3(1):1–13.
- Raft disorganization leads to reduced plasmin activity in Alzheimer’s disease brains. EMBO reports. 2003;4(12):1190–1196.
- Altered lipid composition in cortical lipid rafts occurs at early stages of sporadic Alzheimer’s disease and facilitates APP/BACE1 interactions. Neurobiology of aging. 2014;35(8):1801–1812.
- Structural membrane alterations in Alzheimer brains found to be associated with regional disease development; increased density of gangliosides GM1 and GM2 and loss of cholesterol in detergent-resistant membrane domains. Journal of neurochemistry. 2005;92(1):171–182.
- Flotillin is a novel diagnostic blood marker of Alzheimer’s disease. Journal of Alzheimer’s Disease. 2019;72(4):1165–1176.
- Neuronal ER-Signalosome Proteins as Early Biomarkers in Prodromal Alzheimer’s Disease Independent of Amyloid-β𝛽\betaitalic_β Production and Tau Phosphorylation. Frontiers in Molecular Neuroscience. 2022;15:879146.
- Robust quantitation of gangliosides and sulfatides in human brain using UHPLC-MRM-MS: Method development and application in Alzheimer’s disease. Talanta. 2023;256:124264.
- Accumulation of flotillin-1 in tangle-bearing neurones of Alzheimer’s disease. Neuropathology and applied neurobiology. 2003;29(5):451–461.
- Foley P. Lipids in Alzheimer’s disease: A century-old story. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 2010;1801(8):750–753.
- Evidence for changes in the Alzheimer’s disease brain cortical membrane structure mediated by cholesterol. Neurobiol Aging. 1992;13(3):413–419.
- ABCA1-mediated cholesterol efflux capacity to cerebrospinal fluid is reduced in patients with mild cognitive impairment and Alzheimer’s disease. Journal of the American Heart Association. 2016;5(2):e002886.
- Alterations of cholesterol precursor levels in Alzheimer’s disease. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 2010;1801(8):945–950.
- In Vivo and In Silico Evidence: Hippocampal Cholesterol Metabolism Decreases with Aging and Increases with Alzheimer’s Disease–Modeling Brain Aging and Disease. In: 2011 IEEE 11th International Conference on Data Mining Workshops. IEEE; 2011. p. 1064–1070.
- The role of DHCR24 in the pathogenesis of AD: re-cognition of the relationship between cholesterol and AD pathogenesis. Acta Neuropathologica Communications. 2022;10(1):35.
- Abnormal brain cholesterol homeostasis in Alzheimer’s disease–a targeted metabolomic and transcriptomic study. npj Aging and Mechanisms of Disease. 2021;7(1):11.
- Tangle-bearing neurons contain more free cholesterol than adjacent tangle-free neurons. Acta neuropathologica. 2001;101:547–554.
- Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer’s disease. Proc Natl Acad Sci U S A. 2004;101(7):2070–2075.
- Comparative lipidomic analysis of mouse and human brain with Alzheimer disease. J Biological Chem. 2012;287(4):2678–2688.
- Cholesterol retention in Alzheimer’s brain is responsible for high β𝛽\betaitalic_β-and γ𝛾\gammaitalic_γ-secretase activities and Aβ𝛽\betaitalic_β production. Neurobiol of disease. 2008;29(3):422–437.
- Increased expression of cholesterol transporter ABCA1 is highly correlated with severity of dementia in AD hippocampus. Brain research. 2010;1318:167–177.
- STARD1 and NPC1 expression as pathological markers associated with astrogliosis in post-mortem brains from patients with Alzheimer’s disease and Down syndrome. Aging (Albany NY). 2020;12(1):571.
- Time-of-flight secondary ion mass spectrometry (TOF-SIMS) imaging reveals cholesterol overload in the cerebral cortex of Alzheimer disease patients. Acta neuropathologica. 2013;125(1):133–144.
- The sterol regulatory element-binding protein 2 is dysregulated by tau alterations in Alzheimer disease. Brain Pathol. 2019;29(4):530–543.
- Elevated cellular cholesterol in Familial Alzheimer’s presenilin 1 mutation is associated with lipid raft localization of β𝛽\betaitalic_β-amyloid precursor protein. PLoS One. 2019;14(1):e0210535.
- Cerebral and extracerebral cholesterol metabolism and CSF markers of Alzheimer’s disease. Biochemical pharmacology. 2013;86(1):37–42.
- Cholesterol metabolism is associated with soluble amyloid precursor protein production in Alzheimer’s disease. Journal of neurochemistry. 2012;123(2):310–316.
- Proprotein convertase subtilisin/kexin type 9, brain cholesterol homeostasis and potential implication for Alzheimer’s disease. Frontiers in aging neuroscience. 2019;11:120.
- The Alzheimer’s disease-associated C99 fragment of APP regulates cellular cholesterol trafficking. The EMBO journal. 2020;39(20):e103791.
- Altered cholesterol ester cycle in skin fibroblasts from patients with Alzheimer’s disease. Journal of Alzheimer’s disease. 2009;18(4):829–841.
- Endo-lysosomal dysregulations and late-onset Alzheimer’s disease: impact of genetic risk factors. Molecular neurodegeneration. 2019;14(1):20.
- APP function and lipids: a bidirectional link. Frontiers in molecular neuroscience. 2017;10:63.
- Chung SH. Aberrant phosphorylation in the pathogenesis of Alzheimer’s disease. BMB reports. 2009;42(8):467–474.
- Florencia Iulita M, Claudio Cuello A. The NGF metabolic pathway in the CNS and its dysregulation in Down syndrome and Alzheimer’s disease. Current Alzheimer Research. 2016;13(1):53–67.
- Differential preservation of AMPA receptor subunits in the hippocampi of Alzheimer’s disease patients according to Braak stage. Experimental neurology. 2004;187(2):299–309.
- A pathophysiological framework of hippocampal dysfunction in ageing and disease. Nature Reviews Neuroscience. 2011;12(10):585–601.
- Braak H, Braak E. Alzheimer’s disease affects limbic nuclei of the thalamus. Acta Neuropathologica. 1991;81(3):261–268.
- Laminar activity in the hippocampus and entorhinal cortex related to novelty and episodic encoding. Nature communications. 2014;5(1):5547.
- Localization and proteomic characterization of cholesterol-rich membrane microdomains in the inner ear. Journal of proteomics. 2014;103:178–193.
- Functional role of lipid raft microdomains in cyclic nucleotide-gated channel activation. Molecular pharmacology. 2004;65(3):503–511.
- Reggies/flotillins regulate retinal axon regeneration in the zebrafish optic nerve and differentiation of hippocampal and N2a neurons. Journal of Neuroscience. 2009;29(20):6607–6615.
- A population-based study on the association between statin use and sudden sensorineural hearing loss. Otolaryngology–Head and Neck Surgery. 2015;152(2):319–325.
- Olfactory drug effects approached from human-derived data. Drug discovery today. 2015;20(11):1398–1406.
- Hearing loss and hair cell death in mice given the cholesterol-chelating agent hydroxypropyl-β𝛽\betaitalic_β-cyclodextrin. PloS one. 2012;7(12):e53280.
- Integrated late onset Alzheimer’s disease (LOAD) susceptibility genes: cholesterol metabolism and trafficking perspectives. Gene. 2017;597:10–16.
- Neurofibrillary tangles in Niemann-Pick disease type C. Brain. 1995;118(1):119–129.
- Castello MA, Soriano S. Rational heterodoxy: cholesterol reformation of the amyloid doctrine. Ageing Res Rev. 2013;12(1):282–288.
- Multiscale analysis of independent Alzheimer’s cohorts finds disruption of molecular, genetic, and clinical networks by human herpesvirus. Neuron. 2018;99(1):64–82.
- Anti-herpetic medications and reduced risk of dementia in patients with herpes simplex virus infections–a nationwide, population-based cohort study in Taiwan. Neurotherapeutics. 2018;15(2):417–429.
- Causal evidence that herpes zoster vaccination prevents a proportion of dementia cases. medRxiv. 2023;p. 2023–05.
- Itzhaki RF, Wozniak MA. Herpes simplex virus type 1, apolipoprotein E, and cholesterol: a dangerous liaison in Alzheimer’s disease and other disorders. Prog Lipid Res. 2006;45(1):73–90.
- An update on the association between traumatic brain injury and Alzheimer’s disease: focus on tau pathology and synaptic dysfunction. Neurosci & Biobehavioral Rev. 2020;.
- The 20-year voyage aboard the journal of Alzheimer’s disease: docking at ‘Type 3 Diabetes’, environmental/exposure factors, pathogenic mechanisms, and potential treatments. Journal of Alzheimer’s Disease. 2018;62(3):1381–1390.
- BMI and risk of dementia in two million people over two decades: a retrospective cohort study. The lancet Diabetes & endocrinology. 2015;3(6):431–436.
- Elucidating the role of TREM2 in Alzheimer’s disease. Neuron. 2017;94(2):237–248.
- A preclinical perspective on the enhanced vulnerability to Alzheimer’s disease after early-life stress. Neurobiol Stress. 2018;8:172–185.
- Increased levels of tau protein in SH-SY5Y cells after treatment with cholinesterase inhibitors and nicotinic agonists. Journal of neurochemistry. 2000;74(2):777–784.
- Bilingualism for delaying the onset of Alzheimer’s disease: a systematic review and meta-analysis. European Geriatric Medicine. 2020;p. 1–8.
- Bilingualism and “brain reserve": A matter of age. Neurobiol Aging. 2019;81:157–165.
- The impact of bilingualism on brain reserve and metabolic connectivity in Alzheimer’s dementia. Proc Natl Acad Sci U S A. 2017;114(7):1690–1695.
- Relationship Between Exercise and Alzheimer’s Disease: A Narrative Literature Review. Front Neurosci. 2020;14:131.
- Alzheimer’s disease: the cholesterol connection. Nat Neurosci. 2003;6(4):345–351.
- Mattson MP. Hormesis defined. Ageing research reviews. 2008;7(1):1–7.
- Adult neurogenesis in the hippocampus: from stem cells to behavior. Cell. 2016;167(4):897–914.
- Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat Med. 2019;25(4):554–560.
Paper Prompts
Sign up for free to create and run prompts on this paper using GPT-5.
Top Community Prompts
Explain it Like I'm 14
A simple explanation of “A Lipid Rafts Theory of Alzheimer’s Disease”
What this paper is about
The paper proposes a new, big-picture explanation for Alzheimer’s disease (AD). It says that a tiny part of brain cell membranes, called lipid rafts, doesn’t form properly in many people with AD. Because these rafts need cholesterol to work, the paper ties many AD problems to how brain cells get and use cholesterol. Using this idea, it explains memory loss, brain changes seen under the microscope, and known risk factors like the ApoE4 gene.
What questions the paper tries to answer
- Why does Alzheimer’s start with trouble making new memories?
- Why do we see two famous brain changes in AD: tau tangles inside cells and amyloid-beta (Aβ) plaques outside cells?
- Why is the ApoE4 version of a cholesterol-carrying gene such a strong risk factor?
- Why do some people have many Aβ plaques but still think and remember normally?
- How do many known risks (aging, infection, insulin resistance, stress, poor sleep) fit together?
How the author approached the problem
Instead of running new lab experiments, the author built a theory by carefully piecing together results from a very large number of existing studies over many years. He first introduces a simple model for how the brain learns and changes (plasticity), and then shows how problems in that process can lead to Alzheimer’s.
To make it easier to understand, think of brain plasticity like upgrading a city’s roads after a traffic jam:
- Step 1: Candidate generation (Cgen). The city quickly tries lots of new side streets and detours. It’s messy, but it helps right away. This is like short-term memory.
- Step 2: Competition resolution (Cres). The city studies which routes worked best, builds them properly, removes bad ones, and sets everything back into a stable state. This is like turning short-term memory into long-term memory.
The key switch from step 1 to step 2 needs lipid rafts—tiny, organized “platforms” in the cell membrane—and these rafts need cholesterol.
What are lipid rafts and why does cholesterol matter?
- Lipid rafts are like small, sturdy docks in the cell’s outer membrane. They help important proteins gather and work together, and they link the outside world to the cell’s inner skeleton.
- Brain neurons get most of their cholesterol from helper cells called astrocytes. Astrocytes send cholesterol using a protein called ApoE (like delivery trucks). Different “truck types” exist: ApoE2, ApoE3, and ApoE4. ApoE4 trucks tend to deliver cholesterol less effectively to the right place, especially as we age.
- Without enough cholesterol in the right spot, rafts don’t form well. Without rafts, the brain can’t complete the switch from short-term to long-term memory.
The new plasticity model in simple terms
- Step 1 (Cgen): The brain briefly loosens its structure, boosts activity, and creates many “candidate” connections (new or strengthened synapses and branches). This feels like short-term memory—quick but fragile. A piece of the APP protein called sAPPα helps this building phase and also boosts cholesterol supply.
- Step 2 (Cres): The brain picks the best candidates and makes them strong and stable, while removing the weaker ones. This saves the memory long-term. Here, cholesterol-rich lipid rafts are essential. Another APP piece, amyloid-beta (Aβ), normally helps with the clean‑up by removing “loser” connections.
- Tau is a protein that acts like scaffolding. When turned “on,” tau helps link the inner skeleton to the membrane and stabilizes “winner” connections. When turned “off,” it lets the cell remove “losers.”
- Calcium acts like a referee: strong, focused bursts help pick winners; lower, lingering amounts help remove losers. The same molecule can help or harm depending on dose—this “double-edged” behavior is a recurring theme.
What goes wrong in Alzheimer’s, according to this theory
- Main idea: Neurons can’t form good lipid rafts because they aren’t getting or using cholesterol properly (often worse with ApoE4 and aging).
- If rafts don’t form, the brain gets stuck mostly in step 1 (Cgen). That means:
- Short-term memories don’t convert into long-term memories (classic early AD symptom).
- Tau can’t be properly turned “on” at the right places, so it stays overly “off” (hyper‑phosphorylated) and forms tangles inside cells.
- Many signals that rely on the membrane’s organized platforms (rafts) are weakened, and cells become fragile over time.
- Neurons try to compensate by making their own cholesterol. This often increases Aβ production. Two things can then happen: 1) Some neurons manage to form rafts and work fairly well, but produce lots of Aβ that can gather into plaques. These plaques can exist even when thinking is still okay—explaining why plaques don’t always match symptoms. 2) Other neurons still can’t form rafts. They show memory failure, tau problems, and ongoing Aβ production that adds stress.
- The “double-edged” calcium signal becomes a slow, chronic clean‑up signal. That means the brain keeps pruning and removing connections, which leads to degeneration.
What evidence the paper points to (in brief)
- In brain areas hit early by AD, membranes and lipid rafts look abnormal, with disturbed cholesterol and related lipids.
- Tau problems correlate well with where and when symptoms appear.
- ApoE4 (poorer cholesterol delivery) is a strong risk factor, just as the raft idea predicts.
- Plaques often contain cholesterol and raft lipids, linking Aβ to cholesterol handling.
- Early synapse damage and loss—exactly what you’d expect if rafts can’t stabilize “winner” connections.
- Many risk factors that raise AD risk (aging, some viral infections, insulin resistance/diabetes in the brain, blood vessel problems, stress, inflammation, poor sleep) are known to disturb lipid rafts, cholesterol handling, or the switch from step 1 to step 2.
- Familial (genetic) AD fits too: mutations that change how APP is cut can disrupt both the building (sAPPα) and clean‑up (Aβ) tools, harming plasticity earlier in life.
Why this matters
This theory connects many scattered facts into one simple story: cholesterol delivery to neuron membranes controls whether the brain can “lock in” memories. When that system falters, short‑term memory can’t become long‑term, tau goes wrong, and Aβ builds up—not as the original cause, but as part of a plasticity system stuck in the wrong mode.
What this could mean for the future
Here are some directions this theory suggests:
- Treatments might work better if they restore raft formation and healthy cholesterol handling in the brain. That could mean:
- Improving astrocyte-to-neuron cholesterol delivery (ApoE function, ABCA1/ABCG1 transporters).
- Carefully supporting SREBP2 and insulin/Akt signaling in the brain.
- Protecting or rebuilding lipid rafts and the lipids that form them.
- Be cautious with medicines that reduce brain cholesterol too much or disturb rafts.
- Combine Aβ-targeting drugs with therapies that fix the underlying raft/cholesterol problem, rather than focusing on Aβ alone.
- Prevention strategies that help rafts and plasticity: good sleep, exercise, treating insulin resistance, managing stress and inflammation, protecting brain blood vessels, and addressing infections.
- New diagnostics could look for early signs of raft and cholesterol imbalance in brain fluid or blood, or by imaging cholesterol distribution.
Final takeaway
Think of Alzheimer’s as a problem with the brain’s ability to finish the job of learning. It starts building new routes (short‑term memory) but can’t switch over to solid, stable roads (long‑term memory) because the tiny membrane “platforms” (lipid rafts) don’t form properly. That single failure—often tied to how the brain moves cholesterol—can explain the memory symptoms, tau tangles, Aβ plaques, and many known risks. This gives scientists a clearer target: restore healthy raft formation and cholesterol handling so the brain can complete the learning process.
Collections
Sign up for free to add this paper to one or more collections.