Abstract
β‐Amyloid (Aβ) is a specific pathological hallmark of Alzheimer's disease (AD). Because of its neurotoxicity, AD patients exhibit multiple brain dysfunctions. Disease‐modifying therapy (DMT) is the central concept in the development of AD therapeutics today, and most DMT drugs that are currently in clinical trials are anti‐Aβ drugs, such as aducanumab and lecanemab. Therefore, understanding Aβ's neurotoxic mechanism is crucial for Aβ‐targeted drug development. Despite its total length of only a few dozen amino acids, Aβ is incredibly diverse. In addition to the well‐known Aβ1‐42, N‐terminally truncated, glutaminyl cyclase (QC) catalyzed, and pyroglutamate‐modified Aβ (pEAβ) is also highly amyloidogenic and far more cytotoxic. The extracellular monomeric Aβx‐42 (x = 1–11) initiates the aggregation to form fibrils and plaques and causes many abnormal cellular responses through cell membrane receptors and receptor‐coupled signal pathways. These signal cascades further influence many cellular metabolism‐related processes, such as gene expression, cell cycle, and cell fate, and ultimately cause severe neural cell damage. However, endogenous cellular anti‐Aβ defense processes always accompany the Aβ‐induced microenvironment alterations. Aβ‐cleaving endopeptidases, Aβ‐degrading ubiquitin‐proteasome system (UPS), and Aβ‐engulfing glial cell immune responses are all essential self‐defense mechanisms that we can leverage to develop new drugs. This review discusses some of the most recent advances in understanding Aβ‐centric AD mechanisms and suggests prospects for promising anti‐Aβ strategies.
Keywords: Alzheimer's disease (AD), astrocytes, endopeptidase, glutaminyl cyclase (QC), microglia, p75 neurotrophin receptor (p75NTR), proteolysis targeting chimeras (PROTACs), β‐Amyloid (Aβ)
Many variants of Aβ exist, created by a combination of N‐terminal truncates and C‐terminal truncates. QC enzyme catalyzed pEAβx‐42, plays the role of “ignition” as a seed to cause Aβ to aggregate into plaques. Extracellular Aβ activates various Aβ receptors on cell membranes, triggering numerous downstream cell signaling cascades, ultimately impacting cell metabolism, gene expression, cell cycle, and differentiation. On the other hand, Aβ activates endogenous anti‐Aβ mechanisms, and these self‐protection mechanisms are promising targets for developing future AD drugs.

1. ALZHEIMER'S DISEASE AND AMYLOID HYPOTHESIS
Alzheimer's disease (AD) is a chronic neurodegenerative disease hallmarked by cognitive defects, specifically episodic memory impairment. 1 The predominant risk factor for AD is aging since AD appears predominantly in late adulthood. The risk of developing the disease rises exponentially after the sixth decade of life. 2 The most critical in vivo pathological hallmarks of the disease are extracellular β‐amyloid (Aβ) accumulation 3 and intracellular tau propagation. 4 Early deficits of AD typically exhibit as mild and can slowly progress for as long as several decades. The aggregation of Aβ peptide and microtubule‐associated protein tau begins early and initiates the disease, marking the biochemical phase. 5 Later, neuroinflammation and immune responses mediated by excessive proliferation of microglia and astrocytes 6 , 7 arise during the cellular phase. 5 MRI changes, hippocampal shrinkage, memory loss, and dementia appear when the disease advances to the clinical phase. 5
Amyloid‐containing tissue was first documented in 1639 with a description of the appearance of the lardaceous liver and white stone‐containing spleen tissue, and characterization of the protein has continued over the subsequent four centuries. 8 Over time, the concept of “amyloidosis” was established, and Aβ found in the AD brain was added to the growing list of substances triggering amyloidosis. The “amyloid hypothesis” implies that increases in amyloid plaques or soluble Aβ are the primary events causing the onset and progression of the disease. 3 Aβ pathology directly involves accumulation of Aβ oligomers and Aβ plaque, as well as indirectly leading to cellular metabolic disorders (e.g. ER stress and oxidative stress), 8 , 9 synaptic deterioration, 10 and neuronal apoptosis 11 (Figure 1). This toxicity mechanism is conserved across a wide range of species, as supported by the observation that increased Aβ levels can lead to diffused amyloid deposits in the brain and severe cognitive defects such as memory deficits in mice and fruit flies. 12 , 13
FIGURE 1.

Aβ pathology constitutes an essential mechanism for AD. Aβ oligomers, or Aβ plaques, exert their neurotoxic effects by causing various cellular metabolic disorders, such as ER stress, oxidative stress, and cell cycle disturbances. These neural function disruptions lead to further synaptic deterioration and neuronal apoptosis. This figure was created with BioRender.com with the publication agreement number BZ24RS5K5N.
Because of the neurotoxic effects of Aβ, AD patients can exhibit a variety of cognitive dysfunctions, such as slow response times, impaired short‐term memory, repetitive speech, and decreased comprehension and expression capabilities. To date, there is still no effective drug for AD. In the past few decades, pharmaceutical companies have invested heavily in R&D to develop drugs that target Aβ, many of which have successfully entered clinical trials. Unfortunately, all have failed in clinical trials. 10 So why are Aβ‐targeted drugs ineffective? Is the amyloid hypothesis wrong? One possible explanation is that due to the insidious onset of AD and the prolonged course of the disease, Aβ may indeed initiate the disease at the very initial stage, but it also causes changes in other molecules in the cell, such as phosphorylation of tau protein at the synaptic site, 14 and gradually continues to affect to more downstream molecules. When the clinical symptoms of AD have begun to appear, the progression of the disease is likely no longer dependent solely on Aβ, so trying to reduce the level of Aβ is not enough to significantly improve the patient's cognitive impairment. 14 Even so, scientists keep developing drugs that target Aβ using a new mainstream concept, disease‐modifying therapy (DMT). 15 DMT is based on direct action of the drug on the pathophysiological mechanism of the disease, that is, treatment of the cause, but not the symptom. Currently, the DMT drugs that account for the most significant proportion of clinical trials are anti‐Aβ drugs, such as aducanumab and lecanemab, which demonstrates academic acceptance of the amyloid hypothesis. Therefore, understanding Aβ's neurotoxic mechanism is critical for Aβ‐targeted drug development.
2. Aβ PRODUCTION AND AGGREGATION
Aβ consists of peptides of 37–49 amino acids produced by sequential proteolytic cleavage of amyloid precursor protein (APP) by β‐secretase and γ‐secretase. 16 The γ‐secretase can cleave at different C‐terminal positions, creating Aβ fragments of different lengths. 17 Aβ plaques also contain N‐terminally truncated Aβ peptides, giving Aβ species even more variation in sequence and length. 18 Among all these Aβ species, Aβ1‐42, Aβ4‐42, and Aβ11‐42 are proven to be the most amyloidogenic forms of the peptide. 19 Aβ molecules can aggregate to form soluble oligomers, which can further misfold to form, first, Aβ fibrils and, ultimately, Aβ plaques. 8 Many neurodegenerative diseases, including AD, involve prion mechanisms. Protein conformational disorders (PCD) derive from protein misfolding, leading to conformational change which might promote toxicity and subsequent disease. The misfolded protein can self‐associate and becomes deposited in Aβ deposits. The critical events in Aβ PCD are the turn formations and β‐pleated‐sheet conformations 20 , 21 , 22 that stimulate this aggregation in AD. 23 Importantly, the turn position plays a pivotal role in the ability of Aβ to induce aggregative and cytotoxicity. The difference in cytotoxicity between Aβ42 and Aβ40 can be explained by the difference in the turn position in the aggregates; the former has a turn at positions 22 and 23, 20 and the latter has a turn at positions 25 and 26 21 due to the difference in the salt bridges between Lys28/Ala42 24 and Asp23/Lys28. 21
Pyroglutamate (pGlu) modified N‐terminally truncated Aβ (pEAβ) likely initiates aggregation, called “seeding”, because the Aβ plaque core mainly contains pEAβx‐42 (x = 2–11). 25 Further, the researchers discovered that pEAβ3‐42 can co‐oligomerize with excess Aβ1‐42 to form misfolded metastable low‐n oligomers (LNOs) that are far more cytotoxic to cultured neurons than comparable LNOs made from Aβ1‐42 alone and can facilitate the seeding of new LNOs. 26 Because of its seeding capability and convincing preclinical trial evidence, pEAβ has recently become a new potential drug target in AD. 27 In addition, because glutaminyl cyclase (QC) catalyzes the formation of pEAβ, 28 developing drugs targeting QC may also be promising, as supported by a series of pilot investigations. For example, studies using QC knockout transgenic mice 29 or QC inhibitors 30 showed that decreasing QC levels can efficiently rescue the AD phenotype.
While Aβ exists both intracellularly and extracellularly, most is present in the extracellular environment. 31 In the first instance, Aβ, a secreted protein, is cleaved from APP and secreted into the extracellular space. 9 Extracellular Aβ can be taken up into the cell via endocytosis. 8 , 32 Extracellular Aβ aggregates, but intracellular Aβ does not because Aβ aggregation requires a seeding event mediated by Aβ and cell‐surface interactions, 33 and membrane lipid‐mediated seeding takes place only in the extracellular space. 34 , 35 Specifically, gangliosides are located in the outer leaflet of the plasma membrane. 36 Monosialotetrahexosyl ganglioside (GM1), which is implicated in raft structure, 36 binds tightly to Aβ40 37 and Aβ42 38 and facilitates their aggregation. 37 , 39 Lipid raft‐associated Aβ can serve as a platform and acts as an adaptor to initiate extracellular Aβ aggregation into Aβ plaques. 34
3. RECEPTOR‐COUPLED SIGNAL PATHWAYS THAT MEDIATE Aβ CYTOTOXICITY
The soluble extracellular Aβ dimers or oligomers can activate numerous cellular signal transduction pathways coupled to multiple types of well‐recognized Aβ receptors, causing pleiotropic downstream effects. 9 So far, many Aβ receptors have been discovered, including receptor of advanced glycation endproducts (RAGE), 40 p75 neurotrophin receptor (p75NTR), 41 and nicotinic acetylcholine receptors (nAChRs), 42 , 43 among others. In particular, p75NTR, also known as nerve growth factor receptor (NGFR), functions at the molecular nexus of cell death, survival, and differentiation and thus serves as a new therapeutic candidate for intervention in many diseases, including AD. 44 Blocking Aβ binding to p75NTR can reduce Aβ‐induced neurodegeneration, neuroinflammation, and cognitive deficits. 44 Drug developers have carried out animal studies in in vivo AD models to test LM11A‐31, a p75NTR inhibitor, and observed its excellent performance in reducing Aβ oligomer‐induced dendritic loss and synaptic failure. 45 Clinical randomized controlled trials (RCT) of LM11A‐31 in AD are currently ongoing and hopefully will give us positive outcomes soon.
Aβ can not only alter cellular signal transduction pathways by directly binding Aβ receptors but can also interfere with the binding of other ligands and receptors. New studies have shown that before insoluble Aβ plaque formation, soluble Aβ dimers cause glutamate excitotoxicity by blocking glutamate reuptake in the synaptic cleft. 46 , 47 The excessive glutamate stimulation of multiple types of glutamate receptors and their downstream cell signaling transduction cascades, such as CamKII 48 and MAPK/ERK, 49 , 50 can cause neurotoxicity. 51 Another study has shown that Aβ can cause insulin receptor redistribution, leading to insulin resistance, and disturbing ERK/MAPK, PI3K/AKT, and GSK3 pathways. 52
To a large extent, the signal transduction pathways disrupted by Aβ overlap with AD‐related neurodegeneration cellular networks, such as the PKC pathway, 53 , 54 the NF‐κB pathway, 55 and the Ras/MAPK/ERK pathway, 56 , 57 etc. These dysregulated cellular signal transduction pathways can potentially alter gene transcription by phosphorylating transcription factors and their coactivators, 58 , 59 , 60 nuclear receptors, 61 and histones 58 (Figure 1). A crucial focus of AD research is neurogenesis in the brain, which may help development of pro‐neurogenic therapies. 62 Although Aβ has been proven to inhibit cell proliferation 63 , 64 due to disruption of the mitotic spindle and mitotic microtubule motors, 65 it seems to positively enhance the neuronal differentiation phenotype 66 , 67 , 68 , 69 (Figure 1). The underlying mechanism likely involves the Ras GTPases‐MAPK molecular network induced by Aβ. The small GTPases play essential roles in neurogenesis and cell differentiation and are implicated in AD pathogenesis. 57 Studies have shown the positive relationship between Ras superfamily members and Aβ42 production, 70 supporting observations of increased neuronal differentiation and the parallel increase in Aβ plaques observed in human AD. 71 As expected, inhibiting these Ras GTPases decreases Aβ42 levels and improves cognition in AD mouse models, 72 making the Ras/MAPK/ERK pathway a promising therapeutic target for AD. Notably, although it plays a neuroprotective role in fighting amyloid pathology by changing APP processing and reducing Aβ42 production, 73 excess neuronal differentiation has other side effects, such as increased neurogenic‐to‐gliogenic fate switch, e.g. astrogenesis 73 , 74 and increased APP expression. 73
4. CELLULAR ANTI‐Aβ DEFENSE MECHANISMS
In response to excessive production of Aβ, cellular defense mechanisms are initiated. Leveraging these endogenous anti‐Aβ mechanisms can hopefully help to develop new drugs with different mechanisms for treating AD in the future (Figure 2). Many endopeptidases that exist in cells are also Aβ‐degrading proteases (Figure 2A). Endopeptidases directly cleave Aβ 75 , 76 , 77 or reduce processing of APP to Aβ, 78 playing a crucial role in Aβ clearance. Accumulating studies have reported many such endopeptidases. Neprilysin (NEP), a metalloendopeptidase, can directly cleave Aβ, 79 , 80 , 81 and its mRNA and protein levels in the hippocampus and temporal gyrus in AD patients are significantly lower than those in the control group. 82 Similarly, in the cerebral cortex of transgenic Tg2576 mice with AD‐like pathology, NEP proteins levels were decreased. 83 Genetic linkage analyses on chromosome 10q have been used to find a strong association between the insulin‐degrading enzyme (IDE) gene and late onset AD, 84 and IDE can also degrade Aβ in the brain. 81 , 85 Matriptase, a member of the type 2 transmembrane serine protease (TTSP) family that is encoded by the suppression of tumorigenicity‐14 (ST14) gene, cleaves APP and reduces its processing to Aβ. 78
FIGURE 2.

Research evidence reveals endogenous anti‐Aβ mechanisms, which provide alternative drug development approaches to suppress Aβ pathologies. These mechanisms primarily include, but are not limited to, endopeptidases, the ubiquitin‐proteasome system (UPS), and glial cell‐mediated immune responses. (A) Many endopeptidases can directly cleave Aβ. (B) The UPS can utilize a sizeable enzymatic system to precisely degrade unwanted proteins. The new proteolysis targeting chimeras (PROTACs) technique has become a promising way to fight AD pathologies. (C) Glial cells are more than glue. They can internalize and clear Aβ just like scavengers. This figure was created with BioRender.com with the publication agreement number RQ24RS581W.
Another Aβ degrading mechanism is the ubiquitin‐proteasome system (UPS), which degrades unwanted or damaged proteins in the cytoplasm (Figure 2B). Ubiquitination first acts as a signal for protein degradation, mediated by three different enzymes: the ubiquitin‐activating enzyme (E1), the ubiquitin‐conjugating enzyme (E2), and the ubiquitin‐ligase enzyme (E3). 86 Then ubiquitin‐tagged proteins are directed to the 26S proteasome for degradation. 86 Studies have proved that UPS can degrade Aβ, 87 and UPS defects are evident in AD. 88 A study using postmortem human cerebral cortex revealed that the trypsin‐like proteolytic activity of the 26S proteasome was reduced, and E1 and E2 activity was significant decreased in an AD sample. 89 Another mass spectrometry‐coupled liquid chromatography investigation found that ubiquitin C (UBC) is a key protein that interacts with a variety of pathophysiological molecular factors associated with AD and suggested that the reduction in UPS is one of the causative factors of AD. 90 A recently developed technique, proteolysis targeting chimeras (PROTACs), 91 can utilize UPS to efficiently and precisely degrade target proteins, such as phosphorylated tau, 92 to battle tauopathies. Whether this technique can also efficiently reduce Aβ pathologies by targeting Aβ and Aβ‐associated molecules is a fascinating question for future research.
Neural immune responses mediated by brain immune cells – microglia and astrocytes – in response to Aβ‐induced inflammation play a crucial role in AD development. 6 , 93 A recent transcriptomic study utilizing laser capture microscopy (LCM) discovered the upregulation of microglia‐related genes in the Aβ plaque tissue compared to non‐Aβ plaque tissue in the APP mouse, suggesting a robust immune response at the molecular level under the omic scale. 94 Both microglia 93 and astrocytes 95 can take up amyloid (Figure 2C), which primarily implicates Aβ‐binding scavenger receptors. The microglial phagocytosis‐mediated clearance of Aβ 7 helps to alleviate Aβ pathology. Class B scavenger receptors such as CD36, expressed on microglia, can recognize and bind Aβ and mediate macrophage recognition of Aβ. 96 , 97 In addition, class A scavenger receptors can mediate Aβ phagocytosis by astrocytes and lead to Aβ clearance. 95 Based on these neuroprotective functions, researchers anticipate that glial cells may become new therapeutic targets in AD treatment. Still, neuroinflammation is a double‐edged sword because the hyperactivation of glial cells can also be detrimental. In most cases, Aβ can disrupt the ability of microglia to clear the protein and cause microglia hyperactivation. 98 In addition, senescent astrocytes showed increased secretion of senescence‐associated secretory phenotype (SASP) factors in AD, reducing the glial cells' ability to clear Aβ. 99
5. SUMMARY AND FUTURE DIRECTIONS
Currently, the main approaches to AD drug development include Aβ‐targeted drugs, tau‐targeted drugs, and drugs using other mechanisms. Over the past 20 years, hundreds of drugs, including Aβ‐targeted medicines, have been effective in animal experiments, but most have failed in clinical trials. Therefore, developing new powerful drugs for treating AD is urgent and requires a more profound understanding of AD mechanisms. This review has discussed some of the most recent advances in understanding AD mechanisms and how this further knowledge helps contribute to new drug development. Future work will soon investigate these Aβ‐centric mechanisms, such as the endogenous Aβ degrading system, in more depth and bring us more effective treatment methods for AD.
AUTHOR CONTRIBUTIONS
HZ, FE, and JW conceived and drafted the manuscript. HZ, XL, and JX designed and made the graphics. XL and XW helped find, collect, and sort references.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest. Haolin Zhang is an Editorial Board member of AMEM and a co‐author of this article. To minimize bias, he was excluded from all editorial decision‐making related to the acceptance of this article for publication.
ACKNOWLEDGMENTS
We thank the National Natural Science Foundation of China (NSFC) (grant numbers 91854115 and 31970044) for the funds to Juan Wang, the Beijing University of Technology Faculty of Environment and Life Seed Funding (fund number 049000513202) to Haolin Zhang, and the National Institute of Neurological Disorders and Stroke/National Institutes of Health (NINDS/NIH) of the United States of America (grant number 2RF1NS095799) for the funds to Felice Elefant.
Zhang H, Li X, Wang X, Xu J, Elefant F, Wang J. Cellular response to β‐amyloid neurotoxicity in Alzheimer's disease and implications in new therapeutics. Anim Models Exp Med. 2023;6:3‐9. doi: 10.1002/ame2.12313
Felice Elefant and Juan Wang contributed equally to this work as corresponding authors.
REFERENCES
- 1. Dubois B, Feldman HH, Jacova C, et al. Advancing research diagnostic criteria for Alzheimer's disease: the IWG‐2 criteria. Lancet Neurol. 2014;13:614‐629. [DOI] [PubMed] [Google Scholar]
- 2. Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL. Alzheimer's disease. Nat Rev Dis Primers. 2015;1:15056. [DOI] [PubMed] [Google Scholar]
- 3. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297:353‐356. [DOI] [PubMed] [Google Scholar]
- 4. Mamun AA, Uddin MS, Mathew B, Ashraf GM. Toxic tau: structural origins of tau aggregation in Alzheimer's disease. Neural Regen Res. 2020;15:1417‐1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. De Strooper B, Karran E. The cellular phase of Alzheimer's disease. Cell. 2016;164:603‐615. [DOI] [PubMed] [Google Scholar]
- 6. Heneka MT, Carson MJ, El Khoury J, et al. Neuroinflammation in Alzheimer's disease. Lancet Neurol. 2015;14:388‐405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Solito E, Sastre M. Microglia function in Alzheimer's disease. Front Pharmacol. 2012;3:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Iadanza MG, Jackson MP, Hewitt EW, Ranson NA, Radford SE. A new era for understanding amyloid structures and disease. Nat Rev Mol Cell Biol. 2018;19:755‐773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Chen GF, Xu TH, Yan Y, et al. Amyloid beta: structure, biology and structure‐based therapeutic development. Acta Pharmacol Sin. 2017;38:1205‐1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Karisetty BC, Bhatnagar A, Armour EM, Beaver M, Zhang H, Elefant F. Amyloid‐beta peptide impact on synaptic function and Neuroepigenetic gene control reveal new therapeutic strategies for Alzheimer's disease. Front Mol Neurosci. 2020;13:577622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Takada E, Okubo K, Yano Y, et al. Molecular mechanism of apoptosis by amyloid beta‐protein fibrils formed on neuronal cells. ACS Chem Nerosci. 2020;11:796‐805. [DOI] [PubMed] [Google Scholar]
- 12. Hsiao K, Chapman P, Nilsen S, et al. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science. 1996;274:99‐102. [DOI] [PubMed] [Google Scholar]
- 13. Iijima K, Liu HP, Chiang AS, Hearn SA, Konsolaki M, Zhong Y. Dissecting the pathological effects of human Abeta40 and Abeta42 in drosophila: a potential model for Alzheimer's disease. Proc Natl Acad Sci U S A. 2004;101:6623‐6628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Sehar U, Rawat P, Reddy AP, Kopel J, Reddy PH. Amyloid Beta in aging and Alzheimer's disease. Int J Mol Sci. 2022;23:1‐25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Breijyeh Z, Karaman R. Comprehensive review on Alzheimer's disease: causes and treatment. Molecules. 2020;25:5789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Bergmans BA, De Strooper B. Gamma‐secretases: from cell biology to therapeutic strategies. Lancet Neurol. 2010;9:215‐226. [DOI] [PubMed] [Google Scholar]
- 17. van der Kant R, Goldstein LS. Cellular functions of the amyloid precursor protein from development to dementia. Dev Cell. 2015;32:502‐515. [DOI] [PubMed] [Google Scholar]
- 18. Masters CL, Selkoe DJ. Biochemistry of amyloid beta‐protein and amyloid deposits in Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2:a006262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Teplow DB. Structural and kinetic features of amyloid beta‐protein fibrillogenesis. Amyloid. 1998;5:121‐142. [DOI] [PubMed] [Google Scholar]
- 20. Colvin MT, Silvers R, Ni QZ, et al. Atomic resolution structure of monomorphic Abeta42 amyloid fibrils. J Am Chem Soc. 2016;138:9663‐9674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Petkova AT, Ishii Y, Balbach JJ, et al. A structural model for Alzheimer's beta ‐amyloid fibrils based on experimental constraints from solid state NMR. Proc Natl Acad Sci U S A. 2002;99:16742‐16747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Walti MA, Ravotti F, Arai H, et al. Atomic‐resolution structure of a disease‐relevant Abeta(1‐42) amyloid fibril. Proc Natl Acad Sci U S A. 2016;113:E4976‐E4984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Collinge J. Mammalian prions and their wider relevance in neurodegenerative diseases. Nature. 2016;539:217‐226. [DOI] [PubMed] [Google Scholar]
- 24. Xiao Y, Ma B, McElheny D, et al. Abeta(1‐42) fibril structure illuminates self‐recognition and replication of amyloid in Alzheimer's disease. Nat Struct Mol Biol. 2015;22:499‐505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Miller DL, Papayannopoulos IA, Styles J, et al. Peptide compositions of the cerebrovascular and senile plaque core amyloid deposits of Alzheimer's disease. Arch Biochem Biophys. 1993;301:41‐52. [DOI] [PubMed] [Google Scholar]
- 26. Nussbaum JM, Schilling S, Cynis H, et al. Prion‐like behaviour and tau‐dependent cytotoxicity of pyroglutamylated amyloid‐beta. Nature. 2012;485:651‐655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Bayer TA. Pyroglutamate Abeta cascade as drug target in Alzheimer's disease. Mol Psych. 2022;27:1880‐1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Cynis H, Scheel E, Saido TC, Schilling S, Demuth HU. Amyloidogenic processing of amyloid precursor protein: evidence of a pivotal role of glutaminyl cyclase in generation of pyroglutamate‐modified amyloid‐beta. Biochemistry. 2008;47:7405‐7413. [DOI] [PubMed] [Google Scholar]
- 29. Jawhar S, Wirths O, Schilling S, Graubner S, Demuth HU, Bayer TA. Overexpression of glutaminyl cyclase, the enzyme responsible for pyroglutamate a{beta} formation, induces behavioral deficits, and glutaminyl cyclase knock‐out rescues the behavioral phenotype in 5XFAD mice. J Biol Chem. 2011;286:4454‐4460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Coimbra JR, Sobral PJ, Santos AE, Moreira PI, Salvador JA. An overview of glutaminyl cyclase inhibitors for Alzheimer's disease. Future Med Chem. 2019;11:3179‐3194. [DOI] [PubMed] [Google Scholar]
- 31. Mattson MP. Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol. 2000;1:120‐129. [DOI] [PubMed] [Google Scholar]
- 32. Jin S, Kedia N, Illes‐Toth E, et al. Amyloid‐beta(1‐42) aggregation initiates its cellular uptake and cytotoxicity. J Biol Chem. 2016;291:19590‐19606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Wiatrak B, Piasny J, Kuzniarski A, Gasiorowski K. Interactions of amyloid‐beta with membrane proteins. Int J Mol Sci. 2021;22:6075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Morgado I, Garvey M. Lipids in amyloid‐beta processing, aggregation, and toxicity. Adv Exp Med Biol. 2015;855:67‐94. [DOI] [PubMed] [Google Scholar]
- 35. Niu Z, Zhang Z, Zhao W, Yang J. Interactions between amyloid beta peptide and lipid membranes. Biochim Biophys Acta Biomembr. 2018;1860:1663‐1669. [DOI] [PubMed] [Google Scholar]
- 36. Posse de Chaves E, Sipione S. Sphingolipids and gangliosides of the nervous system in membrane function and dysfunction. FEBS Lett. 2010;584:1748‐1759. [DOI] [PubMed] [Google Scholar]
- 37. Amaro M, Sachl R, Aydogan G, Mikhalyov II, Vacha R, Hof M. GM1 ganglioside inhibits beta‐amyloid oligomerization induced by sphingomyelin. Angew Chem Int ed Engl. 2016;55:9411‐9415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Yanagisawa K, Odaka A, Suzuki N, Ihara Y. GM1 ganglioside‐bound amyloid beta‐protein (a beta): a possible form of preamyloid in Alzheimer's disease. Nat Med. 1995;1:1062‐1066. [DOI] [PubMed] [Google Scholar]
- 39. Ahyayauch H, de la Arada I, Masserini ME, Arrondo JLR, Goni FM, Alonso A. The binding of Abeta42 peptide monomers to sphingomyelin/cholesterol/ganglioside bilayers assayed by density gradient ultracentrifugation. Int J Mol Sci. 2020;21:1674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Schmidt AM, Yan SD, Yan SF, Stern DM. The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses. J Clin Invest. 2001;108:949‐955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Wang YR, Wang J, Liu YH, et al. Cysteine‐rich repeat domains 2 and 4 are amyloid‐beta binding domains of Neurotrophin receptor p75NTR and potential targets to block amyloid‐beta neurotoxicity. J Alzheimers Dis. 2018;63:139‐147. [DOI] [PubMed] [Google Scholar]
- 42. Liu Q, Huang Y, Xue F, et al. A novel nicotinic acetylcholine receptor subtype in basal forebrain cholinergic neurons with high sensitivity to amyloid peptides. J Neurosci. 2009;29:918‐929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Wu J, Kuo YP, George AA, Xu L, Hu J, Lukas RJ. Beta‐amyloid directly inhibits human alpha4beta2‐nicotinic acetylcholine receptors heterologously expressed in human SH‐EP1 cells. J Biol Chem. 2004;279:37842‐37851. [DOI] [PubMed] [Google Scholar]
- 44. Malik SC, Sozmen EG, Baeza‐Raja B, Le Moan N, Akassoglou K, Schachtrup C. In vivo functions of p75(NTR): challenges and opportunities for an emerging therapeutic target. Trends Pharmacol Sci. 2021;42:772‐788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Yang T, Tran KC, Zeng AY, Massa SM, Longo FM. Small molecule modulation of the p75 neurotrophin receptor inhibits multiple amyloid beta‐induced tau pathologies. Sci Rep. 2020;10:20322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Zott B, Simon MM, Hong W, et al. A vicious cycle of beta amyloid‐dependent neuronal hyperactivation. Science. 2019;365:559‐565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Huganir RL, Nicoll RA. AMPARs and synaptic plasticity: the last 25 years. Neuron. 2013;80:704‐717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Gerber U, Gee CE, Benquet P. Metabotropic glutamate receptors: intracellular signaling pathways. Curr Opin Pharmacol. 2007;7:56‐61. [DOI] [PubMed] [Google Scholar]
- 50. Willard SS, Koochekpour S. Glutamate, glutamate receptors, and downstream signaling pathways. Int J Biol Sci. 2013;9:948‐959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Lipton SA, Rosenberg PA. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med. 1994;330:613‐622. [DOI] [PubMed] [Google Scholar]
- 52. Zhao WQ, De Felice FG, Fernandez S, et al. Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J. 2008;22:246‐260. [DOI] [PubMed] [Google Scholar]
- 53. Choi DS, Wang D, Yu GQ, et al. PKCepsilon increases endothelin converting enzyme activity and reduces amyloid plaque pathology in transgenic mice. Proc Natl Acad Sci U S A. 2006;103:8215‐8220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Etcheberrigaray R, Tan M, Dewachter I, et al. Therapeutic effects of PKC activators in Alzheimer's disease transgenic mice. Proc Natl Acad Sci U S A. 2004;101:11141‐11146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Chacon PJ, Rodriguez‐Tebar A. Increased expression of the homologue of enhancer‐of‐split 1 protects neurons from beta amyloid neurotoxicity and hints at an alternative role for transforming growth factor beta1 as a neuroprotector. Alzheimers Res Ther. 2012;4:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Kang S, Lee YH, Lee JE. Metabolism‐centric overview of the pathogenesis of Alzheimer's disease. Yonsei Med J. 2017;58:479‐488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Qu L, Pan C, He SM, et al. The Ras superfamily of small GTPases in non‐neoplastic cerebral diseases. Front Mol Neurosci. 2019;12:121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Chow CW, Davis RJ. Proteins kinases: chromatin‐associated enzymes? Cell. 2006;127:887‐890. [DOI] [PubMed] [Google Scholar]
- 59. De Cesare D, Fimia GM, Sassone‐Corsi P. Signaling routes to CREM and CREB: plasticity in transcriptional activation. Trends Biochem Sci. 1999;24:281‐285. [DOI] [PubMed] [Google Scholar]
- 60. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57‐70. [DOI] [PubMed] [Google Scholar]
- 61. Rochette‐Egly C. Nuclear receptors: integration of multiple signalling pathways through phosphorylation. Cell Signal. 2003;15:355‐366. [DOI] [PubMed] [Google Scholar]
- 62. Zheng J. Hippocampal neurogenesis and pro‐neurogenic therapies for Alzheimer's disease. Animal Model Exp Med. 2022;5:3‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Bernabeu‐Zornoza A, Coronel R, Palmer C, Monteagudo M, Zambrano A, Liste I. Physiological and pathological effects of amyloid‐beta species in neural stem cell biology. Neural Regen Res. 2019;14:2035‐2042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Chatila ZK, Kim E, Berle C, et al. BACE1 regulates proliferation and neuronal differentiation of newborn cells in the adult hippocampus in mice. eNeuro. 2018;5:1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Borysov SI, Granic A, Padmanabhan J, Walczak CE, Potter H. Alzheimer Abeta disrupts the mitotic spindle and directly inhibits mitotic microtubule motors. Cell Cycle. 2011;10:1397‐1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Del Carmen SM, Laura GV, Leonardo OL, Bernabe RG, Antonio MM. Abeta40 oligomers promote survival and early neuronal differentiation of dentate gyrus‐isolated precursor cells through activation of the Akt signaling pathway. Neurotox Res. 2020;38:611‐625. [DOI] [PubMed] [Google Scholar]
- 67. Millet P, Lages CS, Haik S, et al. Amyloid‐beta peptide triggers Fas‐independent apoptosis and differentiation of neural progenitor cells. Neurobiol Dis. 2005;19:57‐65. [DOI] [PubMed] [Google Scholar]
- 68. Piras S, Furfaro AL, Piccini A, et al. Monomeric Abeta1‐42 and RAGE: key players in neuronal differentiation. Neurobiol Aging. 2014;35:1301‐1308. [DOI] [PubMed] [Google Scholar]
- 69. Zhang H, Karisetty BC, Bhatnagar A, et al. Tip60 protects against amyloid‐beta‐induced transcriptomic alterations via different modes of action in early versus late stages of neurodegeneration. Mol Cell Neurosci. 2020;109:103570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Banworth MJ, Li G. Consequences of Rab GTPase dysfunction in genetic or acquired human diseases. Small GTPases. 2018;9:158‐181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Jin K, Peel AL, Mao XO, et al. Increased hippocampal neurogenesis in Alzheimer's disease. Proc Natl Acad Sci U S A. 2004;101:343‐347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Aguilar BJ, Zhu Y, Lu Q. Rho GTPases as therapeutic targets in Alzheimer's disease. Alzheimers Res Ther. 2017;9:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Satir TM, Nazir FH, Vizlin‐Hodzic D, et al. Accelerated neuronal and synaptic maturation by BrainPhys medium increases Abeta secretion and alters Abeta peptide ratios from iPSC‐derived cortical neurons. Sci Rep. 2020;10:601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Paavilainen T, Pelkonen A, Makinen ME, et al. Effect of prolonged differentiation on functional maturation of human pluripotent stem cell‐derived neuronal cultures. Stem Cell Res. 2018;27:151‐161. [DOI] [PubMed] [Google Scholar]
- 75. Leissring MA. The AbetaCs of Abeta‐cleaving proteases. J Biol Chem. 2008;283:29645‐29649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. De Strooper B. Proteases and proteolysis in Alzheimer disease: a multifactorial view on the disease process. Physiol Rev. 2010;90:465‐494. [DOI] [PubMed] [Google Scholar]
- 77. Nalivaeva NN, Beckett C, Belyaev ND, Turner AJ. Are amyloid‐degrading enzymes viable therapeutic targets in Alzheimer's disease? J Neurochem. 2012;120(Suppl 1):167‐185. [DOI] [PubMed] [Google Scholar]
- 78. Lanchec E, Desilets A, Beliveau F, et al. The type II transmembrane serine protease matriptase cleaves the amyloid precursor protein and reduces its processing to beta‐amyloid peptide. J Biol Chem. 2017;292:20669‐20682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Cao W, Song HJ, Gangi T, et al. Identification of novel genes that modify phenotypes induced by Alzheimer's beta‐amyloid overexpression in drosophila. Genetics. 2008;178:1457‐1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Finelli A, Kelkar A, Song HJ, Yang H, Konsolaki M. A model for studying Alzheimer's Abeta42‐induced toxicity in Drosophila melanogaster. Mol Cell Neurosci. 2004;26:365‐375. [DOI] [PubMed] [Google Scholar]
- 81. Leissring MA, Lu A, Condron MM, et al. Kinetics of amyloid beta‐protein degradation determined by novel fluorescence‐ and fluorescence polarization‐based assays. J Biol Chem. 2003;278:37314‐37320. [DOI] [PubMed] [Google Scholar]
- 82. Yasojima K, Akiyama H, McGeer EG, McGeer PL. Reduced neprilysin in high plaque areas of Alzheimer brain: a possible relationship to deficient degradation of beta‐amyloid peptide. Neurosci Lett. 2001;297:97‐100. [DOI] [PubMed] [Google Scholar]
- 83. Apelt J, Ach K, Schliebs R. Aging‐related down‐regulation of neprilysin, a putative beta‐amyloid‐degrading enzyme, in transgenic Tg2576 Alzheimer‐like mouse brain is accompanied by an astroglial upregulation in the vicinity of beta‐amyloid plaques. Neurosci Lett. 2003;339:183‐186. [DOI] [PubMed] [Google Scholar]
- 84. Bertram L, Blacker D, Mullin K, et al. Evidence for genetic linkage of Alzheimer's disease to chromosome 10q. Science. 2000;290:2302‐2303. [DOI] [PubMed] [Google Scholar]
- 85. Chesneau V, Vekrellis K, Rosner MR, Selkoe DJ. Purified recombinant insulin‐degrading enzyme degrades amyloid beta‐protein but does not promote its oligomerization. Biochem J. 2000;351(Pt 2):509‐516. [PMC free article] [PubMed] [Google Scholar]
- 86. Nandi D, Tahiliani P, Kumar A, Chandu D. The ubiquitin‐proteasome system. J Biosci. 2006;31:137‐155. [DOI] [PubMed] [Google Scholar]
- 87. Park HM, Kim JA, Kwak MK. Protection against amyloid beta cytotoxicity by sulforaphane: role of the proteasome. Arch Pharm Res. 2009;32:109‐115. [DOI] [PubMed] [Google Scholar]
- 88. Sulistio YA, Heese K. The ubiquitin‐proteasome system and molecular chaperone deregulation in Alzheimer's disease. Mol Neurobiol. 2016;53:905‐931. [DOI] [PubMed] [Google Scholar]
- 89. Lopez Salon M, Morelli L, Castano EM, Soto EF, Pasquini JM. Defective ubiquitination of cerebral proteins in Alzheimer's disease. J Neurosci Res. 2000;62:302‐310. [DOI] [PubMed] [Google Scholar]
- 90. Manavalan A, Mishra M, Feng L, Sze SK, Akatsu H, Heese K. Brain site‐specific proteome changes in aging‐related dementia. Exp Mol Med. 2013;45:e39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Burslem GM, Crews CM. Proteolysis‐targeting chimeras as therapeutics and tools for biological discovery. Cell. 2020;181:102‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Jangampalli Adi P, Reddy PH. Phosphorylated tau targeted small‐molecule PROTACs for the treatment of Alzheimer's disease and tauopathies. Biochim Biophys Acta Mol Basis Dis. 2021;1867:166162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer's disease. J Cell Biol. 2018;217:459‐472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Rothman SM, Tanis KQ, Gandhi P, et al. Human Alzheimer's disease gene expression signatures and immune profile in APP mouse models: a discrete transcriptomic view of Abeta plaque pathology. J Neuroinflammation. 2018;15:256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Durand D, Turati J, Rudi MJ, et al. Unraveling the beta‐amyloid clearance by astrocytes: involvement of metabotropic glutamate receptor 3, sAPPalpha, and class‐a scavenger receptor. Neurochem Int. 2019;131:104547. [DOI] [PubMed] [Google Scholar]
- 96. Coraci IS, Husemann J, Berman JW, et al. CD36, a class B scavenger receptor, is expressed on microglia in Alzheimer's disease brains and can mediate production of reactive oxygen species in response to beta‐amyloid fibrils. Am J Pathol. 2002;160:101‐112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Moore KJ, El Khoury J, Medeiros LA, et al. A CD36‐initiated signaling cascade mediates inflammatory effects of beta‐amyloid. J Biol Chem. 2002;277:47373‐47379. [DOI] [PubMed] [Google Scholar]
- 98. Pan XD, Zhu YG, Lin N, et al. Microglial phagocytosis induced by fibrillar beta‐amyloid is attenuated by oligomeric beta‐amyloid: implications for Alzheimer's disease. Mol Neurodegener. 2011;6:45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Han X, Zhang T, Liu H, Mi Y, Gou X. Astrocyte senescence and Alzheimer's disease: a review. Front Aging Neurosci. 2020;12:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
