You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Interacting Models of Amyloid-β and Tau Proteins: An Approach to Identify Drug Targets in Alzheimer’s Disease

Abstract

Alzheimer’s disease (AD) is the primary cause of dementia affecting millions each year across the world, though still remains incurable. This might be attributed to the lack of knowledge about the associated proteins, their cellular and molecular mechanisms, and the genesis of the disease. The discovery of drugs that earlier revolved around targeting the amyloid-β cascade has now been reformed with the upgraded knowledge of the cross-seeding ability of tau protein which opens new gateways for therapeutic targets. This article provides a comprehensive review of various direct and indirect connecting pathways between the two main proteins involved in development and progression of AD, enabling us to further expand our repertoire of information regarding the etiology of AD. The current review indicates the need for extensive research in this niche, thus considerable advances can be made in understanding AD which eventually helps to improve the current therapeutics against AD.

INTRODUCTION

Alzheimer’s disease, named after its discoverer Dr. Alois Alzheimer, is a neurodegenerative disorder characterized by the formation of extracellular insoluble plaques of amyloid-β (Aβ) protein and intraneuronal tangles of tau protein (τP) developed in the neuronal cells [1]. Amyloid-β protein precursor (AβPP) is a transmembrane protein of neurons which is involved in the growth and repair mechanism [2]. These proteins are cleaved by two enzymes, α-secretase and γ-secretase, producing a soluble peptide which will be degraded; however, in the presence of another enzyme, β-secretase, along with γ-secretase, it produces an insoluble sticky Aβ monomer which aggregates outside neuronal cells [3, 4]. The plaques formed are localized between synapses disrupting signals and further affecting memory [5, 6]. Neurons are held together by cytoskeleton made up of microtubules, which are bound together with the help of τP which when phosphorylated tend to change their shape and cluster together forming neurofibrillary tangles inside the cells [7–9], driving it toward apoptosis [10]. The symptoms begin with short term memory loss, then as the accumulation of proteins proceed, the loss of motor skills and language are also observed [11, 12].

There exist two classes of AD, namely sporadic (late-onset) AD and familial (early-onset) AD [13]. The manifestation of the former one is associated with genetic and environmental factors, which is also affected by the succession of age [14]. An apolipoprotein (APOE) gene, which is affected, whose product binds to Aβ polypeptide, plays a major role [15]. This gene has 3 major alleles (E2-E4) among which the less effective one is E4 and therefore its presence makes the individual more susceptible to acquiring AD [16]. In familial AD, genes that happen to be mutated are presenilin 1 (PSEN1) and presenilin 2 (PSEN2), which constitute the catalytic subunits of γ-secretase or APP gene, resulting in the accumulation of Aβ polypeptides [17].

Aggregation of Aβ and τP is invariably observed in both forms of AD, which drives the downfall of the central nervous system and into developing the characteristic symptoms of AD.

This is already an established theory that aggregation of Aβ proteins starts about 20 years [18] and that of tau about 15 years prior to the occurrence of the first observed symptom, indicating a possible connection between the two proteins [19]. However, the already present Aβ protein has been found to induce the phosphorylation of τP proteins [20, 21], further causing their detachment from the microtubules of the axon and forming tangles. In the race of discovering a cure to AD, the scientists have targeted Aβ peptide to prevent its aggregation in the neuronal cells; however the outcome of clinical trials carried out for these drugs have not been promising [14]. Presently, scientists have changed their focus from targeting Aβ plaque aggregates to the interaction between the two proteins, the role it plays in inducing phosphorylation of tau, in order to reduce the rate of deterioration of neuronal cells. On the other hand, some studies have also revealed the role of τP in inducing Aβ toxicity in cells [22], generating a vicious cycle of pathogenesis, which gets established in AD.

Here in this review, we discuss the direct and indirect interacting models of Aβ and τP proteins that have been put forward by various researchers, to understand the feedback loop of the two proteins involved, and, moreover, continuously explore new potential targets for therapeutic drugs.

DIRECT INTERACTIVE PATHOGENESIS

Neurodegenerative diseases like AD and Parkinson’s disease, where aggregation of prion-like proteins is caused due to cross-seeding, have shown to produce prominent pathological implications [23]. In vitro studies have presented with evidence of the direct interaction between Aβ and τP proteins forming stable insoluble forms, further inducing toxicity. The strength of association to Aβ protein is dictated by the nature of τP, i.e., whether it is in phosphorylated or dephosphorylated form [24]. The phosphorylation of serine and threonine amino acid residues of τP has been shown to decrease its affinity toward Aβ protein, with the help of techniques such as Enzyme linked immunosorbent assay and western blotting [25]. This dependency indicated the direct role of Aβ in the phosphorylation of τP proteins, and alongside, in vivo demonstration of the co-localization of proteins was displayed by immunofluorescence assay [25].

Moreover, the researchers were able to discover the interactive sites on these proteins by utilizing peptide membrane arrays, exons 7 and 9 of τP, and middle to c-terminal region of Aβ were discovered to be the interacting sites [25]. This common interactive site occurs to be exploited in identifying a therapeutic drug target to reduce prion aggregation [26].

INDIRECT INTERACTIVE MODELS

Here presented are the brief outlines of various models describing the indirect interactions between Aβ and τP proteins that invariably occur through cell signaling pathways.

GSK3: One of the culprits

Glycogen synthase kinase 3 (GSK3) phosphorylates tau protein [27], which causes its detachment from microtubules and then their aggregation. The Aβ plaques hinder both insulin [28] and Wnt signaling pathways [29], creating an abundance of activated GSK3 in the neuronal cells. In the insulin pathway, Aβ acts as an antagonist to insulin, inhibiting phosphoinositide 3-kinase (PI3), and Akt (serine/threonine-specific protein kinase) activation, as shown in Fig. 1. Akt kinase phosphorylates two isoforms of GSK3, i.e., GSK3α and GSK3β, causing it to lose its kinase property and to be able to phosphorylate τP. However, if Akt is inhibited, the GSK3 is not phosphorylated and is available with its active phosphorylating property [28].

Fig. 1

A schematic representation of how Aβ oligomers interfere with the insulin and Wnt pathways, leading to an increase in the levels of activated GSK3 in the cell which ultimately leads to neuron degeneration [30].

A schematic representation of how Aβ oligomers interfere with the insulin and Wnt pathways, leading to an increase in the levels of activated GSK3 in the cell which ultimately leads to neuron degeneration [30].

The Wnt pathway in one of its parts is responsible for the inactivation of GSK3; on the other hand, the presence of Aβ protein does not allow it to do that, and hence activated GSK3 peptides are present in the neuronal cells. The above-stated deviations of the cellular mechanisms are known to occur in familial AD [30].

CDK5: Another culprit

Cyclin-dependent kinase 5 (CDK5), a known kinase of tau protein, structurally similar to another kinase GSK3, is involved in regulating the cell cycle and several different cellular mechanisms in neurons [31]. The p35 and p39 forms are the activators of CDK5, which are involved in the proteasome degradation of CDK5, upon phosphorylation [32]; however, it gets cleaved into more stable p25 and p29 by calcium-dependent protease calpain. It was activated in presence of a high concentration of Ca2 + ions in the cell, an outcome of Aβ toxicity [33]. Hence, in the presence of stable activators, CDK5 remains active for a prolonged time in the cell causing phosphorylation of τP proteins [33] as shown in Fig. 2.

Fig. 2

A schematic representation of both CDK5 and GSK3β induced phosphorylation of tau protein, activated by calpain cleaved p25. Protein phosphatases 1 and 2A which dephosphorylates tau protein also activate GSK3β leading to more phosphorylation than dephosphorylation. Thick black arrow indicates more affinity of p25 toward GSK3β than CDK5, blue and red arrows indicate phosphorylation and dephosphorylation activity respectively [33].

A schematic representation of both CDK5 and GSK3β induced phosphorylation of tau protein, activated by calpain cleaved p25. Protein phosphatases 1 and 2A which dephosphorylates tau protein also activate GSK3β leading to more phosphorylation than dephosphorylation. Thick black arrow indicates more affinity of p25 toward GSK3β than CDK5, blue and red arrows indicate phosphorylation and dephosphorylation activity respectively [33].

Protein phosphatases 1 and 2A are the main dephosphorylating agents of tau protein [34]. Protein phosphatases 1 is involved in activating GSK3β [35] and protein phosphatases 2A dephosphorylates Akt and thus inhibiting its activity [36]. The p25 has been shown to have a higher affinity toward GSK3β than CDK5, and therefore it binds to its C-terminus domain, causing an alternation in its activity, leading to an increase in phosphorylation of tau proteins [37].

Oxidative stress-mediated tau phosphorylation

Aβ oligomers interact with the neuronal membrane through receptors such as N-methyl-D-aspartate receptor (NMDAR), Ephrin type-A receptor 4 precursor [38], cellular prion protein [39], or can even get embedded into the lipid bilayer, leading to peroxidation of bilayer molecules that causing the damage to proteins, and hence taking our prime defense down [40].

Inside the cell, Aβ can induce tau phosphorylation through a plethora of pathways, for some of them the working mechanism is known (Fig. 3)

Fig. 3

A schematic representation of Aβ-mediated tau phosphorylation, generation of ROS, and cell death through oxidative stress. Blue circle, red triangle, and yellow circle in the mitochondria indicates complex I and IV of electron transport chain, SOD2, and NADPH oxidase, respectively [38, 41, 44, 46, 49, 50].

A schematic representation of Aβ-mediated tau phosphorylation, generation of ROS, and cell death through oxidative stress. Blue circle, red triangle, and yellow circle in the mitochondria indicates complex I and IV of electron transport chain, SOD2, and NADPH oxidase, respectively [38, 41, 44, 46, 49, 50].
  • AGE-RAGE Stress [41] - Advanced glycation end products (AGE) and its receptor (RAGE) play a role in the etiology of AD. RAGE is a multi-ligand, and once Aβ transacts, it induces activation of Nuclear factor Kappa B (NF-κB), which further upregulates the level of various cytokines in the cell, and thus cause cell death [42]. Its activation also increases the expression of RAGE and makes cells more prone to external stress [42]. Their transact also activates NADPH oxidase [43], which leads to an increase in the concentration of reactive oxygen species (ROS) [44]. Cytokines are also known to upregulate this oxidase [45]. ROS further increases the production of Aβ oligomers [46].

  • Aβ hinders the activity of both complex I and IV of the electron transport chain [47] in mitochondria, by directly associating with subunit 1 of cytochrome c oxidase, the major site for the production of ROS and hence causing its upregulation in the cell [40].

  • Aβ induces higher expression of Regulator of calcineurin gene (RCAN1) through oxidative stress as it responds to it [48], which further inhibits the phosphatase activity of calcineurin, an enzyme that dephosphorylates tau protein. It also increases levels of GSK3β, a kinase known to phosphorylate tau [49].

  • The presence of Aβ has also been shown to decrease the levels of superoxide dismutase 2 (SOD2), which provide protection against the production of ROS, especially superoxides, and such a downregulation leads to their accumulation and increases in phosphorylation of τP. A plausible explanation can be due to the depletion of microtubule dependent protein 2 (MAP2), a protein required for the association of tau with microtubules, whose reduction has been linked to hyperphosphorylation of tau [50].

Acetylation of tau: associate of phosphorylation

Tau protein, similar to any other protein, can undergo various post-translational modifications including phosphorylation, acetylation [51], nitration [52], glycation [53], etc.

Aβ oligomers have been shown to induce nitric oxide (NO) production, nitrosylated glyceraldehyde 3-phosphate dehydrogenase (GAPDH) which acetylates p300 acetyltransferase, causing its activation and then acetylation of tau protein [54] as shown in Fig. 4. Nitrosylated GAPDH causes deactivation by nitrosylation of Sirtuin1 (SIRT1), a deacetylase of tau, further increasing the possibility of tau acetylation [55]. Its acetylation sets off its aggregation through phosphorylation by proline-directed protein kinases (PDPKs) [56].

Fig. 4

A Schematic representation of how Aβ dictates tau phosphorylation via a GAPDH nitrosylated dictated pathway which acetylates p300 and nitrosylates SIRT1 promoting tau acetylation which furthermore causes its phosphorylation by PDKs [55].

A Schematic representation of how Aβ dictates tau phosphorylation via a GAPDH nitrosylated dictated pathway which acetylates p300 and nitrosylates SIRT1 promoting tau acetylation which furthermore causes its phosphorylation by PDKs [55].

Dendritic tau-dependent Aβ toxicity

Tau protein, abundantly seen in axon and comparatively less in dendrites [57], upon phosphorylation, detaches from the microtubule and forms intracellular neurofibrillary tangles. These tangles of tau protein show greater affinity towards FYN, a tyrosine-protein kinase that is then localized to the somatodendritic region, further to phosphorylate NMDAR receptor present in the neuronal membrane and facilitates its interaction with postsynaptic density protein (PSD95) [58] (Fig. 5). Once NMDAR receptors are phosphorylated, they become more sensitive to the already existing Aβ plaques, which induce further downstream toxicity effects [22], and it also induces an influx of Ca2 + ions and affects other pathways, ultimately leading to neuronal loss [59].

Fig. 5

A schematic representation of tau influenced Aβ toxicity by enhancing the sensitivity of NMDAR receptor to Aβ peptides through its phosphorylation by FYN kinase [58].

A schematic representation of tau influenced Aβ toxicity by enhancing the sensitivity of NMDAR receptor to Aβ peptides through its phosphorylation by FYN kinase [58].

CONCLUSIONS

The interacting models of Aβ and τP stated in this review show the diverse signaling pathways that are involved in the etiology of AD, revealing the potential drug targets for the prevention or cure of the disease. Apart, there is a need to discover the other AD-associated pathways for a better understanding of the disease.

Earlier drugs used targeted the degradation of Aβ aggregates and failed to provide the desired outcome due to the seeding phenomena associated with the prion-like proteins. Nevertheless, the already phosphorylated tau proteins are sufficient to spread and cause toxicity to the cellular systems. The same situation persisted when τP tangles were targeted. Hence, aiming at only one of the proteins might not provide a cure to AD.

One current study that targets a common epitope involved in cross-seeding of the proteins with peptide-based inhibitors has shown effective results in blocking the pathological forms of Aβ and τP [26]. However, better inhibitors could be used for effective and prolonged inhibition of protein aggregations.

Designing a drug that helps cure AD or at least control its progression would require a complete understanding of the crosstalk between Aβ and τP, molecular mechanisms of other associated signaling cascade proteins, taking into account the pharmacodynamics and pharmacokinetics of the drug. The application of nanotechnology toward the bioavailability of drugs and enhancing its qualities and reducing any downstream toxicity can also be utilized.

ACKNOWLEDGMENTS

PK is thankful to the organizers of the Virtual Internship with Science Leader (#VISL), outcome which is this review article. SJM and SKP thank JSS Academy of Higher Education and Research (JSSAHER), Mysuru, Karnataka, India, for the infrastructure and support provided.

CONFLICT OF INTEREST

The authors have no conflict of interest to report.

REFERENCES

[1] 

Alzheimer A ((1911) ) Über eigenartige Krankheitsfälle des späteren Alters (On certain peculiar diseases of old age) - Translated and with an introduction by H Förstl and R Levy. Zbl Ges Neurol Psych 2: , 71–73.

[2] 

Grimm MOW , Mett J , Grimm HS , Hartmann T ((2017) ) App function and lipids: A bidirectional link. Front Mol Neurosci 10: , 63.

[3] 

Selkoe DJ ((2001) ) Alzheimer’s disease: Genes, proteins, and therapy. Physiol Rev 81: , 741–766.

[4] 

Chen GF , Xu TH , Yan Y , Zhou YR , Jiang Y , Melcher K , Xu HE ((2017) ) Amyloid beta: Structure, biology and structure-based therapeutic development. Acta Pharmacol Sin 38: , 1205–1235.

[5] 

Nitsch RM , Blusztajn JK , Pittas AG , Slack BE , Growdon JH , Wurtman RJ ((1992) ) Evidence for a membrane defect in Alzheimer disease brain. Proc Natl Acad Sci U S A 89: , 1671–1675.

[6] 

Reiss AB , Arain HA , Stecker MM , Siegart NM , Kasselman LJ ((2018) ) Amyloid toxicity in Alzheimer’s disease. Rev Neurosci 29: , 613–627.

[7] 

Goedert M , Spillantini MG , Cairns NJ , Crowther RA ((1992) ) Tau proteins of Alzheimer paired helical filaments: Abnormal phosphorylation of all six brain isoforms. Neuron 8: , 159–168.

[8] 

Vinters HV ((2015) ) Emerging concepts in Alzheimer’s disease. Annu Rev Pathol Mech Dis 10: , 291–319.

[9] 

Graham WV , Bonito-Oliva A , Sakmar TP ((2017) ) Update on Alzheimer’s disease therapy and prevention strategies. Annu Rev Med 68: , 413–430.

[10] 

Dickson DW ((2004) ) Apoptotic mechanisms in Alzheimer neurofibrillary degeneration: Cause or effect? J Clin Invest 114: , 23–27.

[11] 

Merriam AE , Aronson MK , Gaston P , Wey S-L , Katz I ((1988) ) The psychiatric symptoms of Alzheimer’s disease. J Am Geriatr Soc 36: , 7–12.

[12] 

Bature F , Guinn BA , Pang D , Pappas Y ((2017) ) Signs and symptoms preceding the diagnosis of Alzheimer’s disease: A systematic scoping review of literature from 1937 to 2016. BMJ Open 7: , e015746.

[13] 

Piaceri I , Nacmias B , Sorbi S ((2013) ) Genetics of familial and sporadic Alzheimer’s disease. Front Biosci (Elite Ed) 5: , 167–177.

[14] 

Tiwari S , Atluri V , Kaushik A , Yndart A , Nair M ((2019) ) Alzheimer’s disease: Pathogenesis, diagnostics, and therapeutics. Int J Nanomedicine 14: , 5541–5554.

[15] 

Avramopoulos D ((2009) ) Genetics of Alzheimer’s disease: Recent advances. Genome Med 1: , 34.

[16] 

Singh PP , Singh M , Mastana SS ((2006) ) APOE distribution in world populations with new data from India and the UK. Ann Hum Biol 33: , 279–308.

[17] 

Shen J , Kelleher RJ ((2007) ) The presenilin hypothesis of Alzheimer’s disease: Evidence for a loss-of-function pathogenic mechanism. Proc Natl Acad Sci U S A 104: , 403–409.

[18] 

Holtzman DM , Morris JC , Goate AM ((2011) ) Alzheimer’s disease: The challenge of the second century. Sci Transl Med 3: , 77sr1.

[19] 

Bloom GS ((2014) ) Amyloid-β and tau: The trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol 71: , 505–508.

[20] 

Alvarez G , Muñoz-Montaño JR , Satrústegui J , Avila J , Bogónez E , Díaz-Nido J ((1999) ) Lithium protects cultured neurons against β-amyloid-induced neurodegeneration. FEBS Lett 453: , 260–264.

[21] 

Busciglio J , Lorenzo A , Yeh J , Yankner BA ((1995) ) β-Amyloid fibrils induce tau phosphorylation and loss of microtubule binding. Neuron 14: , 879–888.

[22] 

Ittner LM , Ke YD , Delerue F , Bi M , Gladbach A , van Eersel J , Wölfing H , Chieng BC , Christie MJ , Napier IA , Eckert A , Staufenbiel M , Hardeman E , Götz J ((2010) ) Dendritic function of tau mediates amyloid-β toxicity in Alzheimer’s disease mouse models. Cell 142: , 387–397.

[23] 

Soto C , Pritzkow S ((2018) ) Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci 21: , 1332–1340.

[24] 

Lasagna-Reeves CA , Castillo-Carranza DL , Guerrero-Muñoz MJ , Jackson GR , Kayed R ((2010) ) Preparation and characterization of neurotoxic tau oligomers. Biochemistry 49: , 10039–10041.

[25] 

Guo JP , Arai T , Miklossy J , McGeer PL ((2006) ) Aβ and tau form soluble complexes that may promote self aggregation of both into the insoluble forms in Alzheimer’s diseases. Proc Natl Acad Sci U S A 103: , 1953–1958.

[26] 

Tripathi T , Khan H ((2020) ) Direct interaction between the β-amyloid core and tau facilitates cross-seeding: A novel target for therapeutic intervention. Biochemistry 59: , 341–342.

[27] 

Jope RS , Johnson GVW ((2004) ) The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci 29: , 95–102.

[28] 

Townsend M , Mehta T , Selkoe DJ ((2007) ) Soluble Aβ inhibits specific signal transduction cascades common to the insulin receptor pathway. J Biol Chem 282: , 33305–33312.

[29] 

Magdesian MH , Carvalho MMVF , Mendes FA , Saraiva LM , Juliano MA , Juliano L , Garcia-Abreu J , Ferreira ST ((2008) ) Amyloid-β binds to the extracellular cysteine-rich domain of frizzled and inhibits Wnt/β-catenin signaling. J Biol Chem 283: , 9359–9368.

[30] 

Hernández F , Gómez de Barreda E , Fuster-Matanzo A , Lucas JJ , Avila J ((2010) ) GSK3: A possible link between beta amyloid peptide and tau protein. Exp Neurol 223: , 322–325.

[31] 

Ip NY , Tsai LH ((2008) ) Cyclin dependent kinase 5 (Cdk5). Springer-Verlag US.

[32] 

Peterson DW , Ando DM , Taketa DA , Zhou H , Dahlquist FW , Lew J ((2010) ) No difference in kinetics of tau or histone phosphorylation by CDK5/p25 versus CDK5/p35 in vitro. Proc Natl Acad Sci U S A 107: , 2884–2889.

[33] 

Kerokoski P , Suuronen T , Salminen A , Soininen H , Pirttilä T ((2004) ) Both N-methyl-D-aspartate (NMDA) and non-NMDA receptors mediate glutamate-induced cleavage of the cyclin-dependent kinase 5 (cdk5) activator p35 in cultured rat hippocampal neurons. Neurosci Lett 368: , 181–185.

[34] 

Liu F , Grundke-Iqbal I , Iqbal K , Gong CX ((2005) ) Contributions of protein phosphatases PP1, PP2A, PP2B and PP5 to the regulation of tau phosphorylation. Eur J Neurosci 22: , 1942–1950.

[35] 

Hernández F , Langa E , Cuadros R , Avila J , Villanueva N ((2010) ) Regulation of GSK3 isoforms by phosphatases PP1 and PP2A. Mol Cell Biochem 344: , 211–215.

[36] 

Mora A , Sabio G , Risco AM , Cuenda A , Alonso JC , Soler G , Centeno F ((2002) ) Lithium blocks the PKB and GSK3 dephosphorylation induced by ceramide through protein phosphatase-2A. Cell Signal 14: , 557–562.

[37] 

Chow HM , Guo D , Zhou JC , Zhang GY , Li HF , Herrup K , Zhang J ((2014) ) CDK5 activator protein p25 preferentially binds and activates GSK3β. Proc Natl Acad Sci U S A 111: , E4887–E4895.

[38] 

Liu Z , Li T , Li P , Wei N , Zhao Z , Liang H , Ji X , Chen W , Xue M , Wei J ((2015) ) The ambiguous relationship of oxidative stress, tau hyperphosphorylation, and autophagy dysfunction in Alzheimer’s disease. Oxid Med Cell Longev 2015: , 352723.

[39] 

Laurén J , Gimbel DA , Nygaard HB , Gilbert JW , Strittmatter SM ((2009) ) Cellular prion protein mediates impairment of synaptic plasticity by amyloid-B oligomers. Nature 457: , 1128–1132.

[40] 

Luque-Contreras D , Carvajal K , Toral-Rios D , Franco-Bocanegra D , Campos-Peña V ((2014) ) Oxidative stress and metabolic syndrome: Cause or consequence of Alzheimer’s disease? Oxid Med Cell Longev 2014: , 497802.

[41] 

Prasad K , Mishra M ((2018) ) AGE-RAGE stress, stressors, and antistressors in health and disease. Int J Angiol 27: , 1–12.

[42] 

Bierhaus A , Humpert PM , Morcos M , Wendt T , Chavakis T , Arnold B , Stern DM , Nawroth PP ((2005) ) Understanding RAGE, the receptor for advanced glycation end products. J Mol Med 83: , 876–886.

[43] 

Yang D , Elner SG , Bian ZM , Till GO , Petty HR , Elner VM ((2007) ) Pro-inflammatory cytokines increase reactive oxygen species through mitochondria and NADPH oxidase in cultured RPE cells. Exp Eye Res 85: , 462–472.

[44] 

Wautier MP , Chappey O , Corda S , Stern DM , Schmidt AM , Wautier JL ((2001) ) Activation of NADPH oxidase by AGE links oxidant stress to altered gene expression via RAGE. Am J Physiol Endocrinol Metab 280: , E685–E694.

[45] 

Mohammed AM , Syeda K , Hadden T , Kowluru A ((2013) ) Upregulation of phagocyte-like NADPH oxidase by cytokines in pancreatic beta-cells: Attenuation of oxidative and nitrosative stress by 2-bromopalmitate. Biochem Pharmacol 85: , 109–114.

[46] 

Prasad K ((2019) ) AGE-RAGE stress: A changing landscape in pathology and treatment of Alzheimer’s disease. Mol Cell Biochem 459: , 95–112.

[47] 

Bobba A , Amadoro G , Valenti D , Corsetti V , Lassandro R , Atlante A ((2013) ) Mitochondrial respiratory chain Complexes I and IV are impaired by β-amyloid via direct interaction and through Complex I-dependent ROS production, respectively. Mitochondrion 13: , 298–311.

[48] 

Crawford DR , Leahy KP , Abramova N , Lan L , Wang Y , Davies KJA ((1997) ) Hamster adapt78 mRNA is a Down syndrome critical region homologue that is inducible by oxidative stress. Arch Biochem Biophys 342: , 6–12.

[49] 

Lloret A , Badia MC , Giraldo E , Ermak G , Alonso MD , Pallardó FV , Davies KJA , Viña J ((2011) ) Amyloid-β toxicity and tau hyperphosphorylation are linked via RCAN1 in Alzheimer’s disease. J Alzheimers Dis 27: , 701–709.

[50] 

Melov S , Adlard PA , Morten K , Johnson F , Golden TR , Hinerfeld D , Schilling B , Mavros C , Masters CL , Volitakis I , Li Q-X , Laughton K , Hubbard A , Cherny RA , Gibson B , Bush AI ((2007) ) Mitochondrial oxidative stress causes hyperphosphorylation of tau. PLoS One 2: , e536.

[51] 

Cohen TJ , Friedmann D , Hwang AW , Marmorstein R , Lee VMY ((2013) ) The microtubule-associated tau protein has intrinsic acetyltransferase activity. Nat Struct Mol Biol 20: , 756–762.

[52] 

Horiguchi T , Uryu K , Giasson BI , Ischiropoulos H , LightFoot R , Bellmann C , Richter-Landsberg C , Lee VMY , Trojanowski JQ ((2003) ) Nitration of tau protein is linked to neurodegeneration in tauopathies. Am J Pathol 163: , 1021–1031.

[53] 

Ledesma MD , Bonay P , Colaço C , Avila J ((1994) ) Analysis of microtubule-associated protein tau glycation in paired helical filaments. J Biol Chem 269: , 21614–21619.

[54] 

Sen T , Saha P , Sen N ((2018) ) Nitrosylation of GAPDH augments pathological tau acetylation upon exposure to amyloid-β. Sci Signal 11: , eaao6765.

[55] 

Saha P , Sen N ((2019) ) Tauopathy: A common mechanism for neurodegeneration and brain aging. Mech Ageing Dev 178: , 72–79.

[56] 

Cohen TJ , Guo JL , Hurtado DE , Kwong LK , Mills IP , Trojanowski JQ , Lee VMY ((2011) ) The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat Commun 2: , 252.

[57] 

Konzack S , Thies E , Marx A , Mandelkow EM , Mandelkow E ((2007) ) Swimming against the tide: Mobility of the microtubule-associated protein tau in neurons. J Neurosci 27: , 9916–9927.

[58] 

Ittner LM , Götz J ((2011) ) Amyloid-β and tau - A toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci 12: , 65–72.

[59] 

Salter MW , Kalia LV ((2004) ) SRC kinases: A hub for NMDA receptor regulation. Nat Rev Neurosci 5: , 317–328.