Next Article in Journal
PD-L1 Expression on Circulating Tumour Cells May Be Predictive of Response to Regorafenib in Patients Diagnosed with Chemorefractory Metastatic Colorectal Cancer
Next Article in Special Issue
Genome Wide Analysis Points towards Subtype-Specific Diseases in Different Genetic Forms of Amyotrophic Lateral Sclerosis
Previous Article in Journal
Novel Highly Soluble Chimeric Recombinant Spidroins with High Yield
Previous Article in Special Issue
SCH23390 Reduces Methamphetamine Self-Administration and Prevents Methamphetamine-Induced Striatal LTD
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Signalling Pathways Implicated in Alzheimer′s Disease Neurodegeneration in Individuals with and without Down Syndrome

by
Carmen Martínez-Cué
* and
Noemí Rueda
Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(18), 6906; https://doi.org/10.3390/ijms21186906
Submission received: 31 August 2020 / Revised: 17 September 2020 / Accepted: 18 September 2020 / Published: 20 September 2020
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)

Abstract

:
Down syndrome (DS), the most common cause of intellectual disability of genetic origin, is characterized by alterations in central nervous system morphology and function that appear from early prenatal stages. However, by the fourth decade of life, all individuals with DS develop neuropathology identical to that found in sporadic Alzheimer’s disease (AD), including the development of amyloid plaques and neurofibrillary tangles due to hyperphosphorylation of tau protein, loss of neurons and synapses, reduced neurogenesis, enhanced oxidative stress, and mitochondrial dysfunction and neuroinflammation. It has been proposed that DS could be a useful model for studying the etiopathology of AD and to search for therapeutic targets. There is increasing evidence that the neuropathological events associated with AD are interrelated and that many of them not only are implicated in the onset of this pathology but are also a consequence of other alterations. Thus, a feedback mechanism exists between them. In this review, we summarize the signalling pathways implicated in each of the main neuropathological aspects of AD in individuals with and without DS as well as the interrelation of these pathways.

1. Introduction

Alzheimer’s disease (AD), the most common form of dementia, affects 44 million people worldwide [1]. The progressive loss of cognitive abilities in this condition is associated with neuropathological changes, including the accumulation of amyloid plaques comprising β-amyloid (Aβ) peptides and neurofibrillary tangles (NFTs) formed by insoluble deposits of abnormally hyperphosphorylated tau, and synapse and neuron loss.
Down syndrome (DS), caused by a partial or complete triplication of human chromosome 21 (Hsa21), affects more than 6 million persons globally [2]. The cognitive alterations found in DS are primarily caused by prenatal changes in central nervous system (CNS) growth and differentiation [3,4]. However, by the fourth decade of their lives, all individuals with DS develop AD neuropathology identical to that found in individuals with sporadic AD [3,5,6,7,8,9]. In the DS population, AD is likely to arise due to the genetic imbalance of several trisomic genes and the interplay of these triplicated genes with other diploid genes. However, in sporadic AD, the main genetic risk factor is the presence of the E4 allele of apolipoprotein E (ApoE) [10]. Despite the different genetic etiopathology of AD in both conditions, the aforementioned fact that all DS individuals develop AD neuropathology suggests that common downstream signalling pathways are affected in both disorders. Thus, the study of the mechanisms implicated in the early onset and high prevalence of AD in DS could be extremely useful in understanding the etiopathology of neurodegeneration and related dementia in sporadic AD.
Some pathological events that appear years before the appearance of amyloid plaques and NFTs play an important role in the onset of the main neuropathological characteristics of AD. These alterations include neuroinflammation, cellular senescence, altered proteostasis, oxidative stress, and reduced neurogenesis [11,12,13,14,15]. Numerous studies have demonstrated the role of these early alterations in the increase in Aβ burden, tau hyperphosphorylation, neuronal death, and accelerated cognitive decline [16,17,18,19,20,21,22,23]. Several signalling pathways are implicated in the onset and aggravation of the aforementioned pathological changes characteristic of AD in individuals with or without DS. This review summarizes the implication of these pathways and their interplay on the most relevant aspects of this disease, including amyloid plaques, NFTs, cholinergic degeneration, oxidative stress, mitochondrial dysfunction, disturbed energy metabolism, cellular senescence, neuroinflammation, altered neurogenesis, and impaired neurotransmission.
Also, some signalling pathways play a role in numerous neuropathological aspects of AD. Because several feedback loops exist between them, their interplay aggravates AD pathology. Thus, we have emphasized the alterations of their function in different aspects of AD as well as their interactions, especially in amyloid plaque and NFT formation, oxidative stress, energy metabolism, neuroinflammation, neurotransmitter release, and synaptic dysfunction. Among these pathways are those controlled by the Dual Specificity Tyrosine-Regulated Protein Kinase 1 (DYRK1A), the Regulator of Calcineurin (RCAN1), neurotrophins, and the Mammalian Target of Rapamycin (mTOR). Finally, this review also describes the role of other pathways that are altered in specific AD signs (e.g., Superoxide Dismutase (SOD1) in oxidative stress or insulin signalling, glucose transport, and metabolism in altered energy metabolism, among others).
The purpose of this review is to provide an overview of the role of the most relevant signalling pathways implicated in the onset and progression of AD in individuals with and without DS.

2. Amyloid Plaques

AD is characterized by altered proteostasis since many of its pathological characteristics are due to changes in the balance and function of different proteins and peptides [15]. In particular, the accumulation of Aβ in plaques is produced by alterations in the synthesis, folding, and clearance of these peptides.
In AD brains, one of the causes of the accumulation of Aβ aggregates is their defective clearance from the brain, a process normally facilitated by ApoE. Indeed, the major genetic risk factor for sporadic AD is a polymorphism of ApoE [10,24,25]. ApoE contributes to the maintenance of brain homeostasis through numerous pathways, including the regulation of cholesterol, glucose metabolism, synaptic plasticity, neurogenesis, inflammatory responses, and Aβ metabolism [26,27,28]. In the AD population, the presence of the ApoE4 isoform correlates with a higher probability of developing dementia and an earlier onset of cognitive decline [26].
The APOE genotype has also been found to modulate the age of onset and progression of AD in DS. DS carriers of the E4 allele have a greater risk of developing AD and an earlier onset of the disease when compared to carriers of other alleles [25,29].
Several Hsa21 genes are implicated in the altered proteostasis that leads to the changes in Aβ aggregation and clearance in AD. Aβ oligomers are the proteolytic products of the Amyloid Precursor Protein (APP) [30]. Because the gene that encodes APP maps to Hsa21, its overexpression was proposed to be responsible for the accumulation of Aβ in AD in individuals with and without DS [31]. However, compelling evidence demonstrates that other Hsa21 genes are key players in the development of AD neuropathology. Some of them encode kinases and phosphatases with multiple targets in different signalling pathways.
One of the genes that has received increased attention is DYRK1A, which encodes a serine-threonine protein kinase [32] and has been associated with the cognitive impairment found in DS [33,34,35,36]. This gene plays a role in the amyloid pathology found in AD and DS. Individuals with AD display enhanced levels of DYRK1A mRNA [37]. DYRK1A phosphorylates APP and enhances its cleavage by β- and γ-secretases [38]. Also, DYRK1A phosphorylates presenilin (PS), the catalytic subunit of the γ-secretase complex [39]. Both phosphorylations promote APP-processing by the amyloidogenic pathway, increasing the formation of the peptides Aβ40 and Aβ42. In turn, these peptides increase DYRK1A transcription, leading to high levels of expression of this kinase in sporadic AD [40].
Another Hsa21-encoded gene that has been implicated in amyloid plaque accumulation is RCAN1, which encodes a calcium-activated serine/threonine protein phosphatase [41]. RCAN1 mediates Aβ-induced neuronal death by enhancing oxidative stress and by disrupting cellular calcium homeostasis in the AD brain [42]. RCAN1 expression is regulated by the calcineurin-Nuclear Factor of Activated T cells (NFAT) transcription factor signalling pathway [43]. In turn, RCAN1 overexpression inhibits different signalling pathways that are controlled by NFAT [44,45]. Thus, the chronic overexpression of RCAN1 that occurs in DS and AD [46] inhibits calcineurin and dysregulates the NFAT pathways. Lower levels of calcineurin and hyperphosphorylation of NFAT are found in the brains of these individuals [42,45,47,48]. The alterations in NFAT signalling promotes Aβ production through different mechanisms, including modulation of the expression of the β-site APP cleaving enzyme 1 (BACE1) gene implicated in Aβ production [49].
In addition, RCAN1 and DYRK1A act synergistically to control the phosphorylation of cytoplasmatic NFAT (NFATc). NFATc may be phosphorylated by DYRK1A, decreasing gene transcription activity [42,48].
Another kinase that has a central role in AD neuropathology is mTOR, a serine/threonine protein kinase. mTOR is involved in the regulation of the proteostasis network due to its ability to inhibit autophagy, a specialized degradative system for the removal of aggregated proteins [50]. In physiological conditions, mTOR inhibits the accumulation of toxic protein aggregates such as Aβ [51]. However, a role for altered mTOR signalling in amyloid pathology has been proposed [52]. mTOR is regulated and interacts with 5′AMP activated protein kinase (AMPK), Phosphoinositide 3 Kinase (PI3K)/AKT, glycogen synthase kinase (GSK3), the extracellular signal-regulated protein kinases (ERK1/2), and insulin/insulin growth factor (IGF) [51,53,54]. The PI3K/Akt/mTOR axis is hyperactivated in DS and AD [55,56,57,58,59,60,61,62,63] and contributes to the altered Aβ generation, deposition, and clearance found in these conditions [51,64,65,66,67,68,69,70,71,72]. In turn, Aβ activates the PI3K/Akt/mTOR signalling pathway [62,67,73,74,75], generating a feedback loop that further aggravates the amyloid pathology in individuals with AD with or without DS.
Another signalling pathway that has been implicated in AD neuropathology is the transcription factor cAMP response element-binding protein (CREB) [76]. CREB is phosphorylated and activated by PI3K/AKT, protein kinase A (PKA), and protein kinase C (PKC) [77]. However, GSK3β inactivates CREB [78,79], and since Aβ peptides activate GSK3β, their overproduction in AD reduces CREB activity [80,81,82]. In agreement with these data, patients with AD show decreased CREB phosphorylation due to alterations in cAMP/PKA signaling [82,83,84]. The disturbances in cAMP/PKA-dependent CREB signalling have been demonstrated to be responsible for Aβ-induced synaptic loss and cognitive impairments [82,85,86].
CREB also regulates several neurotrophins that play a crucial role in cognition such as Brain-Derived Neurotrophic Factor (BDNF) and Nerve Growth Factor (NGF) [87,88,89,90,91]. CREB-regulated BDNF is reduced in AD [88,91,92,93,94] and DS [95,96], and the magnitude of this reduction correlates with cognitive alterations [97]. Regarding NGF, the levels of this neurotrophin are reduced in DS and AD [98,99]. Dysfunction of NGF signalling induces the accumulation of APP C-terminal fragments and Aβ aggregation [100]. Aβ downregulates CREB-mediated transcription [101,102], resulting in synaptic loss and neurodegeneration [92,103,104]. Thus, BDNF/NGF and CREB downregulation could be one of the mechanisms implicated in the cognitive decline observed in AD [82,92,105,106,107].

3. Neurofibrillary Tangles

The accumulation of NFTs composed of hyperphosphorylated tau protein is one of the most characteristic neuropathological characteristics of AD in individuals with or without DS, and it also results from altered proteostasis [108,109,110]. Tau is an axonal phosphoprotein that promotes the self- assembly of tubulin into microtubules and its stabilization in neurons. Tau phosphorylation plays a physiological role in microtubule dynamics. However, hyperphosphorylation of this protein hampers its ability to bind to microtubules, leading to self-assembly and aggregation into NFTs [111,112]. This aberrant process impairs neurotransmission and increases cognitive decline. In fact, hyperphosphorylation of tau, even in the absence of Aβ aggregates, induces cognitive deficits [113].
Individuals with DS and murine DS models display aberrant tau phosphorylation earlier than normal subjects [114,115,116]. Alterations in different signalling pathways in AD individuals with or without DS are responsible for this pathological process. Similarly to what was previously described for amyloid plaque formation, kinases and phosphatases play a crucial role in tau hyperphosphorylation.
DYRK1A kinase can alter tau functioning by enhancing its phosphorylation and by altering tau splicing. DYRK1A phosphorylates tau at different residues [113,117,118], which alters microtubule assembly and stability in the brains of DS individuals and DS mouse models [115,119,120]. Also, DYRK1A phosphorylates NFAT [47] and provokes its inactivation [121,122]. The functional consequences of NFAT dysfunction and its relevance to AD neuropathology have been described in the previous section.
Moreover, alternative splicing of tau produces six different isoforms of this protein [123]. Two of them, 3R-tau (with three microtubule binding repeats) and 4R-tau (with four microtubule binding repeats) are generated by alternative splicing of tau at exon 10. In normal human brains, similar levels of both isoforms are expressed. However, in AD brains, 3R-tau is overexpressed and its levels correlate with aggravation of the disease. Also, the expression of this isoform is modulated by Aβ and by DYRK1A overexpression, which further enhances 3R-tau levels and increases the 3R-tau/4R-tau ratio [124,125].
Regarding another Hsa21-encoded kinase RCAN1, it has been demonstrated that increased levels of RCAN1.1 inhibit calcineurin activity. Calcineurin inhibition prevents the degradation of tau and enhances tau hyperphosphorylation [126,127]. As mentioned above, in DS, the calcineurin-NFAT signalling pathway is altered due to the overexpression of DYRK1A and RCAN1 [47], and a synergic effect exists between both kinases. DYRK1A phosphorylates RCAN1, increasing the ability of RCAN1 to inhibit calcineurin, leading to reduced NFAT transcriptional activity and enhanced tau phosphorylation [41].
In AD, the upregulation of the tau kinase GSK3β by RCAN1 can play a role in tau hyperphosphorylation and aggregation in NFTs [128,129,130]. Interestingly, both RCAN1.1 and GSK3β levels are elevated in the brains of AD patients, and these increases correlate with tau hyperphosphorylation [131] and Aβ production [132].
Other kinases and phosphatases not encoded by Hsa21 control tau phosphorylation. The brains of DS mouse models display increased levels of cyclin-dependent kinase 5 (CDK5) and decreased activity of the serine/threonine phosphatase 2A (PP2A) [133]. CDK5 is implicated in tau phosphorylation in AD [134,135] and DS brains [136]. Also, PP2A is involved in tau hyperphosphorylation in these conditions [137,138]. Thus, the downregulation of PP2A could be partially responsible for the abnormal tau phosphorylation in AD and DS [48,136,139,140,141,142].
mTOR signalling has also been demonstrated to be implicated in tau pathology in AD and DS. Individuals with DS show hyperactivation of mTOR signalling, which correlates with tau hyperphosphorylation, suggesting a role of this pathway’s dysregulation in tau neuropathology in AD and DS [56].
Sirtuin 1 (SIRT1) also interacts with mTOR and regulates mTOR phosphorylation. Reduced levels of SIRT1 are found in DS and AD [143] and mouse models of DS [144]. Furthermore, SIRT1 is a substrate of DYRK1A, which can promote tau accumulation by controlling its deacetylation process [144,145]. Thus, SIRT1 alteration might participate in the NFT deposition induced by aberrant mTOR signaling in DS.
Finally, basal forebrain cholinergic neurons present tau pathology in AD patients [146,147,148], since these neurons show an enhanced 3R-tau/4Rtau ratio [149]. Tau pathology in the basal forebrain cholinergic system occurs in the early stages of AD and is aggravated as the disease progresses [150,151,152], suggesting that tau pathology plays a role in cholinergic degeneration [153]. Acetylcholine receptors play an important role in aberrant tau phosphorylation in AD. While the activation of α7 nicotinic acetylcholine receptors (α7nAChR) facilitates tau phosphorylation, the activation of M1 muscarinic acetylcholine receptors (mAChR) prevents its phosphorylation [154,155,156,157,158]. In addition, nicotine induces tau phosphorylation in AD through the activation of nAChRs [157].

4. Cholinergic Neurodegeneration

One of the most relevant characteristics of AD neuropathology in individuals with and without DS is the degeneration of the basal forebrain cholinergic system [159,160]. The cholinergic system plays a critical role in different components of cognitive function such as attention, information processing, learning, and memory [159]. Altered cholinergic neurotransmission is one of the main determinants of dementia in AD [see 159] and the neuropathological sign that better correlates with the cognitive decline in this disorder [161,162].
AD brains lose increasing numbers of cholinergic neurons as the disease progresses [163,164]. Also, the main components of cholinergic signalling are affected in AD. The levels of the enzyme that catalyze the synthesis of acetylcholine (ACh) and choline acetyltransferase (ChAT) and of the enzymes that degrade ACh (i.e., acetylcholinesterase (AChE)), the vesicular acetylcholine transporter (VAChT) that transports ACh into the vesicles of mAChR and nAChR, are lower in AD and DS than in the normal population. ACh binding to these receptors is also decreased in both conditions [155,165,166,167,168,169].
Several mechanisms play a role in cholinergic neuron loss. Reduced expression of the neurotrophic factor NGF, its precursor proNGF, and their receptors TrkA and p75NTR are found in DS and AD [170]. These alterations can affect cholinergic neuron survival and ACh release [171].
A positive feedback mechanism between the degeneration of this population of neurons and other neuropathological characteristics of AD has been demonstrated. First, as mentioned above, this degeneration has a relevant role in tau pathology in AD. Second, Aβ peptides induce neurodegenerative changes at cholinergic terminals and can alter cholinergic activity, affecting NGF signalling and the consequent tau phosphorylation [172,173]. In turn, cholinergic neuropathology can aggravate Aβ pathology in AD [174].

5. Changes in Energy Consumption and Accumulation: Oxidative Stress, Mitochondrial Alterations, and Energy Metabolism

5.1. Oxidative Stress

Oxidative stress (OS) is one of the most important mechanisms implicated in the neuronal alterations found in DS and AD [175,176]. This process is involved in cellular redox homeostasis, synaptic plasticity, vesicle-mediated transport, neuroinflammation, protein folding and degradation, and signal transduction [177].
In DS, redox imbalance is caused by the enhanced production of reactive oxygen species (ROS) and the inhibition of antioxidant defense mechanisms [175,178]. The overexpression of different Hsa21 genes, which encode proteins that promote ROS production, plays an important role in the enhanced OS found in this syndrome [179,180,181]. One of these genes is SOD1, which encodes the enzyme that catalyzes the transformation of superoxide anions into molecular oxygen and hydrogen peroxide (H2O2). The increased activity of this enzyme in DS leads to the formation of high levels of H2O2 which are not adequately neutralized by the activity of the antioxidant enzymes catalase and glutathione peroxidase, which contributes to the redox imbalance [182].
The Hsa21 RCAN1 gene also plays a role in OS in DS and AD, mainly through the regulation of mitochondrial function [183]. The brains of individuals with sporadic AD present an enhanced expression of RCAN1 [46]. OS induces the expression of RCAN1 via a calcineurin–NFAT-dependent mechanism [184], which inhibits calcineurin activity [185,186] and increases the stress response [183,187]. Also, Aβ enhances RCAN1 protein expression, reducing calcineurin through the induction of OS [46,186]. Altogether, these data suggest that a positive feedback mechanism exists between RCAN expression, OS, and Aβ pathology.
Another Hsa21 gene that plays an important role in oxidative stress is APP. As mentioned in previous sections, abnormal processing of the APP protein leads to enhanced levels of Aβ. These oligomers induce OS by increasing protein, lipid, DNA, and RNA oxidation [187,188], which leads to alterations in different biochemical and metabolic pathways implicated in AD neuropathology [188].
OS also contributes to alteration of the function of two neurotransmitter systems in AD, which are the targets of current pharmacological treatments of this disease. First, OS plays an important role in glutamate-mediated excitotoxicity in which excessive Ca2+ causes cell death [189]. AD is characterized by increased levels of HNE (a product of lipid peroxidation) bound to the glutamate transporter (GLT-1), which prevents the effective removal of glutamate from the synapse, thus promoting excitotoxicity [190]. Regarding the cholinergic system, the levels of HNE-bound ChAT are significantly increased by Aβ42 in AD [191]. Thus, OS can also contribute to alterations in ChAT activity in this disorder.
Finally, ROS modifies the function of the mTOR pathway, which in turn can affect different components of OS. A feedback loop exists between OS and the mTOR pathway [192]. The role of OS in the altered function of the mTOR/autophagy axis in AD and DS has been demonstrated [193,194,195,196]. In the DS brain and DS mouse models, a link between protein oxidative damage and altered mTOR function has been demonstrated [194,195,196]. Because of the aforementioned role of mTOR in the regulation of proteostasis [197], the alteration of this system leads to Aβ and tau pathology in DS and AD [181]. Enhanced ROS, characteristic of these conditions, alters the regulation of autophagy. In turn, altered mTOR activity and reduced autophagy increase ROS production and oxidative damage in DS [195,196,198], thereby facilitating AD neuropathology.

5.2. Mitochondria

Mitochondria are highly metabolic organelles necessary for the maintenance of physiological redox signalling and neuronal activity [199]. Alterations in mitochondrial integrity increase ROS formation [200,201]. In turn, enhanced ROS levels also affect proper mitochondrial function [201]. Altered mitochondrial function plays a role in AD neuropathology including synapse and neuronal loss [177].
The oxidative phosphorylation (OXPHOS) system is the main energy provider to power the activity of mature neurons [202]. According to the “mitochondrial cascade hypothesis”, the origin of AD is a defect in the OXPHOS system [203]. Bioenergetics and Aβ are closely related. Aβ can reduce OXPHOS function and OXPHOS deficiency can increase Aβ production [204]. An OXPHOS defect has been reported in AD [14,205], which secondarily affects de novo pyrimidine biosynthesis and the plasma membrane remodeling of these patients [14]. This might explain the alterations in the composition and structure of neuronal membranes linked to the loss of synapses, which precedes neuronal loss in individuals with AD [206].
DS is also characterized by alterations in OXPHOS function. A reduction in the mRNA levels of several subunits of OXPHOS complexes has been found in DS brains [207,208,209,210,211]. This decline in mRNA levels was accompanied by a lower transcription of mtDNA-encoded genes [11,12]. The quantity of protein subunits for OXPHOS complexes is also reduced in DS brains [211,212,213,214,215]. A decrease in oxygen consumption [210,215,216,217,218,219] and a reduction in mitochondrial inner membrane potential are also characteristic of DS [12,216,218,219,220]. These alterations lead to a reduction in mitochondrial energy production and a lower mitochondrial function [12,215,217,218,219,220].
Some Hsa21 genes play a role in the OXPHOS function. For example, the overexpression of DYRK1A represses a transcriptional coactivator, peroxisome proliferator-activated receptor gamma coactivator 1 α (PGC-1α) that is a key modulator of mitochondrial biogenesis and OXPHOS function [221]. Also, in DS, overexpression of a transcriptional corepressor gene mapping to Hsa21, the nuclear receptor-interacting protein 1 (NRIP1), represses PGC-1α and decreases the mRNA levels of several OXPHOS-related genes [222].
The raptor–mTOR complex plays a role in mitochondrial activity and metabolism [223]. The activation of this complex stimulates the production of ATP by oxidative phosphorylation. In turn, the mTOR pathway is regulated by the redox status of the cell [224]. Thus, again, a positive feedback loop exists between mitochondrial redox status and mTOR activity [197]. As mentioned in previous sections, this pathway is dysregulated in AD and DS and the consequences of this dysregulation play different roles in neurodegeneration.

5.3. Energy Metabolism

One of the mechanisms proposed to be implicated in AD neurodegeneration is the impairment in energy metabolism [197,225]. The risk of developing AD is higher in individuals with obesity [177,226,227,228,229], type II diabetes [230,231,232,233], impaired glucose metabolism [227,233], and hyperlipidemia [234,235]. All of these conditions can cause impairments of brain cells and aggregation of Aβ [236,237]. Some authors have proposed that AD can be considered type III diabetes [238].
Brain insulin resistance is “the failure of brain cells to respond to insulin as they normally would, resulting in impairments in synaptic, metabolic, and immune response functions” [239]. Systemic insulin resistance is a crucial aspect of type II diabetes that contributes to inflammation and oxidative stress. However, brain insulin resistance can occur in the absence of systemic insulin resistance and type II diabetes. Also, so far, it has not been clarified whether systemic and brain insulin resistance affects cognition through the same mechanisms [239].
Alterations of the brain’s insulin resistance pathway have been associated with the development of AD [240]. AD and DS brains show reduced expression of insulin receptors (IR) and increased serine phosphorylation (inhibitory) of insulin receptor substrate 1 (IRS1) [240,241,242,243]. These changes produce alterations in neuronal survival and plasticity, protein synthesis and expression, cell differentiation, and synapse formation [244]. However, normal insulin levels can protect against Aβ toxicity and OS [240].
Also, AD patients display altered glucose transport due to the decreased levels of the glucose transporters GLUT2 and GLUT3 [245] and impaired glucose metabolism [246,247,248]. Indeed, decreased glucose catabolism is found in AD [249,250]. In addition to the production of ATP, the glucose metabolism provides energy and precursors for the biosynthesis of neurotransmitters such as GABA and glutamate and plays a role in autophagy [251,252]. Thus, alterations in glucose metabolism can affect neurotransmission and autophagy.
Different signalling pathways have been implicated in these alterations in energy metabolism in the brains of AD patients. First, the PI3-K/Akt/mTOR axis plays a role in the regulation of energy balance by modulating the response to insulin growth factors (IGFs) and epidermal-derived growth factors (EGFRs). Hyperactivity of the mTOR pathway produces insulin resistance [253] in the brains of AD individuals and mouse models of this disorder [67,73,253], playing a role in the aforementioned AD neuropathology. mTORC1 regulates protein synthesis, autophagy, mitochondrial function, lipogenesis, ketogenesis, and glucose homeostasis through the activation of IGF and EGFR [51]. Growth factors also activate mTORC1 through the Ras signaling pathway effectors ERK1/2 [254]. Also, mTORC2 activates Akt, while Akt modulates mTORC1 [197].
Another kinase implicated in the altered function of the mTOR pathway is AMPK, which regulates cellular metabolism in response to decreased intracellular ATP levels. AMPK and mTOR regulate autophagy [255]. While AMPK activates autophagy, mTOR reduces it. The induction of autophagy by AMPK reduces Aβ levels [66], while the activation of mTOR increases the levels of these peptides [256]. These data provide a further link between the mTOR pathway and altered metabolism in DS and AD.

6. Cellular Senescence

Cellular senescence, a homeostatic process which reduces proliferation and helps to prevent the propagation of damaged cells [257,258], is implicated in the neurodegenerative processes found in AD in individuals with or without DS [15].
Senescent cells are characterized by permanent arrest of the cell cycle [259,260], an increase in the synthesis and release of proinflammatory cytokines, (also called senescence-associated secretory phenotype (SASP)) [261], alterations in mitochondrial function, OS [262], changes in cellular metabolism [263], accumulation of DNA damage [264], changes in nuclear morphology and gene expression [265], and altered proteostasis [266]. As discussed in this review, all these changes can contribute to AD neurodegeneration.
Enhanced senescence has been found in AD and DS brains [15], and it has been proposed to play an important role in the onset and aggravation of AD neuropathology, including Aβ deposition [267], tau phosphorylation [268], increased release of proinflammatory cytokines [269,270] (see Section 7), and alterations in mitochondrial function and OS [176]. A positive feedback loop between cellular senescence and neurodegeneration has been proposed [15].
Additionally, in DS, the overexpression of some genes also induces cellular senescence. The triplication of the Ubiquitin-Specific Peptidase 16 (USP16) gene that encodes a histone H2-specific deubiquitinase plays a role in the enhanced senescence in DS [271]. In a mouse model of DS, the overexpression of this gene downregulates the Wingless and Int-1 (Wnt) signalling pathway, reducing stem cell renewal. USP16 activates Cdkn2a, which acts as a negative regulator of the Wnt signalling pathway. In turn, Wnt plays a crucial role in cellular senescence and aging in various tissues [272,273]. Besides, the USP16 enzyme regulates DNA damage repair by controlling the ubiquitination state of histone H2A. Overexpression of USP16 may induce excessive DNA damage accumulation, leading to acquisition of prematurely senescent phenotypes in different DS cell types [272,274].

7. Immune Response/Inflammation

Years before the appearance of Aβ plaques and NFTs, prominent neuroinflammation was present in the brains of individuals with AD and DS [7,275]. This enhanced neuroinflammation has been demonstrated to play a crucial role in the onset of neurodegeneration in these disorders. Neuroinflammation in DS and AD enhances the production of ROS and aggravates synaptic dysfunction, and Aβ and tau pathology [276,277], while amyloids aggregate NFTs and increase neuroinflammation [278,279].
The brains of AD and DS individuals and of mouse models of these conditions have higher levels of neuroinflammation due to microglia activation, which enhances the release of pro-inflammatory cytokines [7,270,275,280]. Among the inflammatory mediators that have been shown to have a role in neurodegeneration are interleukin-1 (IL-1), IL-6, and IL-17, which are upregulated in DS and AD [270,281,282]. Individuals with AD and mouse models of AD present increased activity of p38 Mitogen-Activated Protein Kinase (p38MAPK), a regulator of the release of cytokines [283]. p38MAPK increases the levels of a number of these cytokines in AD brains, including IL-6, IL-1, and Tumor necrosis factor-α (TNF-α) [281,282,284,285].
Among the mechanisms by which enhanced cytokine release aggravates neuroinflammation is their ability to enhance the expression of APP, the formation of Aβ oligomers, tau hyperphosphorylation, and ROS production [279,286]. However, as previously mentioned, neuroinflammation is not only a cause of neurodegeneration but also a consequence of it. In AD brains, Aβ and APP activate glial cells [287,288], which induces the release of proinflammatory mediators, including IL-1 and Interferon γ (IFNγ) [289]. Because of the high levels of these cytokines, the cells accumulate excessive levels of Aβ that are more likely to be aggregated [7]. Also, IL-1β can exacerbate Aβ expression by increasing BACE [290,291]. IL-1 also affects the activity of the Hypothalamic-Pituitary-Adrenal (HPA) axis, yielding an enhanced release of glucocorticoids [292]. Individuals with AD display hypercortisolism due to alterations in HPA regulation [293]. These high levels of glucocorticoids play a role in other alterations found in AD such as energy deficits [294], insulin resistance [295], and enhanced OS [296].
Cytokines are released by activated microglia, which can also regulate Aβ deposition by phagocytosis [297,298]. When Aβ induction is increased, these cells release inflammatory factors, which results in further activation of microglia and the enhanced release of cytokines and other neurotoxic factors [299,300]. In these circumstances, microglia migrate to Aβ and tau, surrounding them through special pathways and receptors such as CD14 and CD36 [301,302,303], further enhancing the production of pro-inflammatory factors, which damage healthy neurons.
One of the signalling pathways that is implicated in microglia activation is Wnt. This pathway is also implicated in tau hyperphosphorylation and synaptic loss [304]. Both the noncanonical (Wnt5a) and the canonical (Wnt3a) Wnts pathways are implicated in neuroinflammation in AD [304].

8. Changes in Cell Proliferation/Differentiation and Migration

AD is also characterized by reduced neurogenesis [9]. In DS brains, deficits in cell proliferation and differentiation into neurons are found from the early developmental stages and throughout the entire lifespan of the individual [305,306]. Because of the massive loss of different populations of neurons, the reduced regenerative capacity of the brains of individuals with AD with or without DS aggravates the progression of the disease.
Different signalling pathways are implicated in this deficient neurogenesis. As mentioned in Section 6, cellular senescence produces cell cycle arrest in AD and DS. In addition, several kinases and phosphatases encoded in Hsa21 play an important role in neurogenesis defects. First, the Hsa21 gene product DYRK1A plays a role in the altered cell proliferation, differentiation, and survival found in AD and DS through its interaction with different signalling pathways [305,306]. DYRK1A is a negative regulator of cell cycle progression because its overexpression promotes cell cycle exit [307]. Overexpression of DYRK1A also induces premature neuronal differentiation of neuronal progenitors, resulting in a depletion of mature neurons [307]. These altered proliferation and differentiation states induced by DYRK1A are due to its action on different signalling pathways.
One of the downstream DYRK1A pathways that has been implicated in cell cycle arrest is DREAM, a multisubunit complex that regulates quiescence [308]. DREAM complex formation occurs in the G0 phase after DYRK1A phosphorylation [309], which leads to an inhibition of cell proliferation. Also, the phosphorylation of cyclin D1 by DYRK1 inhibits neural cell proliferation and promotes premature differentiation by preventing entry into the S phase [310,311].
DYRK1A inhibits notch signalling, a pathway that controls neurogenesis by maintaining a pool of neuronal progenitor cells (NPCs) in the brain [312]. Thus, it might be implicated in the altered neurogenesis found after DYRK1A overexpression. Notch is overexpressed in the brains of AD and DS individuals [313,314]. The notch signalling pathway is also involved in promoting gliogenesis [315]. DS individuals exhibit an increased number of astrocytes and a reduced number of neurons when compared to the normal population, which provides support for the involvement of the notch signalling pathway in the neurogenic-to-gliogenic shift in DS brains.
Another DYRK1A target is NFAT, for which transcription is inhibited by DYRK1A [47]. Overexpression of DYRK1A and RCAN1 delays neurogenesis by their synergic action on the NFAT pathway [316].
One of the functions of the mTOR signalling pathway is the modulation of cell proliferation and survival [317]. Hyperactivation of this pathway can produce the apoptotic death of NPCs [318]. Thus, the aforementioned alterations in mTOR signaling in DS and AD brains might also be implicated in the neurogenesis defects found in these conditions.
Neurotrophins regulate neuronal survival, differentiation, and migration [319,320]. Among the downstream signalling pathways activated by neurotrophins are the MAPK, PI3K, and phospholipase C-γ (PLCγ) pathways [321,322]. BDNF, the most widely distributed neurotrophic growth factor in the CNS, is essential for the growth, differentiation, and survival of neurons [319,322]. Aβ decreases BDNF by lowering phosphorylated CREB protein. Reduced expression of BDNF is found in AD and DS brains, and it is thought to play a crucial role in the progression of this disease [92]. One of the mechanisms by which this reduced expression may operate is through the impairment of cell proliferation and differentiation.
In DS brains, the mitogenic Sonic Hedgehog (Shh) pathway plays a prominent role in neurogenesis impairment since alterations to this pathway reduce the proliferation of NPCs in different brain areas [323,324,325]. The APP gene plays an important role in cell cycle regulation [326] and is implicated in the altered Shh signalling found in DS [325]. The amyloid precursor protein intracellular domain (AICD) is a cleave product of APP. In DS, APP overexpression produces excessive levels of AICD, which upregulates transcription of the Shh receptor Ptch1 (Patched1). This receptor maintains the Shh pathway in a repressed state [324,325], impairing neurogenesis and aggravating neurodegeneration in DS. Thus, impairment of the Shh pathway due to APP-AICD-dependent Ptch1 overexpression may be a key mechanism that underlies the reduced proliferation and impaired maturation of neuronal precursors in DS and possibly in AD [327].

9. Alterations in Intercellular Signalling: Neurotransmitter Release, Synapses, and Receptors

9.1. Neurotransmitter Release

AD and DS are also characterized by alterations in cellular signalling due to multiple mechanisms. One of the most relevant implicates the DYRK1A and RCAN1 kinases, which have been demonstrated to impair neurotransmitter release. Overexpression of DYRK1A, as occurs in DS and AD, induces alterations in the serotoninergic, dopaminergic, and noradrenergic systems [328]. Because serotonergic transmission is related to GABA synthesis and the glutamatergic and monoaminergic systems interact [329], DYRK1A overexpression can participate in the widespread altered transmission seen in these conditions [330].
Additionally, one of the roles of RCAN1 is the control of neurotransmitter release [331]. Overexpression of this kinase reduces neurotransmitter secretion by impairing the outflow from vesicles [331]. These effects are likely to be due to the inhibitory activity that RCAN1 exerts over calcineurin activity [332], which regulates exocytosis and vesicle recycling [333]. Thus, it is likely that the altered expression of both DYRK1A and RCAN1 is implicated in the impaired intercellular signalling found in AD brains with or without DS.

9.2. Synapses

Years before the appearance of amyloid plaques and NFTs, a massive loss of synapses was evident in the brains of AD patients with or without DS. Alterations in multiple signalling pathways were implicated in this event. First, DYRK1A plays a critical role in synaptic dysfunction [334]. This kinase controls synaptogenesis through axon guidance [335] and the development and maintenance of neurites and dendritic spines [115,336], which are the first stages of synaptic formation. DYRK1A overexpression reduces neuronal dendritic growth and complexity [337] and inhibits the formation of dendritic spines [338].
DYRK1A also regulates synaptic vesicle formation. Overexpression of DYRK1A inhibits endocytosis [339] as well as the production of synaptic components implicated in synapse formation and maintenance [340] such as neuroligin 1 [341] and dynamin [342]. Finally, DYRK1A is also involved in synaptic transmission. Overexpression of this kinase impairs this process [338,343] partially through the modulation of CREB that is implicated in signal transduction pathways responsible for synaptic plasticity [307]. Thus, DYRK1A overexpression is also involved in the alterations of synapse formation, maintenance, and function found in these conditions.
RCAN1 also plays an important role in the synaptic dysfunction found in DS and AD brains. Similar to DYRK1A, RCAN1 mediates axon outgrowth by modulating the actin dynamics of the growth cone [344]. Overexpression of RCAN1 modifies the localization of synaptic proteins such as synaptophysin [345] and decreased phosphorylation of proteins necessary for synaptic plasticity such as CaMKII and ERK1/2 [346]. Calcineurin plays a crucial role in synaptic plasticity and endocytosis through the activation of its downstream targets NFATc, dynamin, and the Hsa21 encoded protein synaptojanin [347,348,349]. Thus, alterations in calcineurin because of RCAN overexpression in DS and AD are implicated in the synaptic dysfunctions found in these conditions.
Another Hsa21 gene that has been demonstrated to be implicated in synaptic alterations in DS and AD is the Down syndrome Cell Adhesion Molecule (DSCAM). This gene plays an important role in dendritic patterning, axon guidance and branching, and synaptic formation. Overexpression of DSCAM in mouse models and DS individuals inhibits dendritic branching [350,351] and synapse formation [352].
Intersectins (ITSNs) are a family of multi-domain adaptor proteins that regulate endocytosis, vesicle recycling, and cell signalling [353]. ITSNs regulate multiple signalling pathways including receptor tyrosine kinases (RTKs), GTPases, and phosphatidylinositol 3-kinase Class 2beta (PI3KC2β). The ITSN1 gene is encoded in Hsa21 [354]. mRNA and protein levels are enhanced in DS and AD [355,356], and this gene is one of the most highly induced genes in AD brains [356]. Increasing evidence supports a role for this protein in the synaptic alterations found in these conditions. Both DS and AD are characterized by enlargement of the early endosomal compartment [357], a sign of altered endocytotic trafficking. This alteration leads to a reduced number of synaptic vesicles and their recycling, which resembles the effects of ITSN1 overexpression. Finally, ITSN1 is also involved in dendritic spine development through the regulation of different proteins [358].
The Wnt signalling pathway protects microglial synapse function and promotes the maturation of neuronal circuits [304]. However, under pathological conditions, such as AD, the canonical Wnt pathway is inhibited, leading to alterations in synapse number and function [359]. In AD, hyperphosphorylation of tau modifies synaptic function through modifications in the Wnt signalling pathway. Synapses damaged by Aβ are eliminated by microglia. However, at the same time, microglia release proinflammatory cytokines that can damage synapses [360,361] either directly or through activation of the Wnt receptor FZD [362]. In DS brains, these pro-inflammatory cytokines can alter the protein expression of synaptic markers (i.e., synapsin-1, PSD95, and GAD65/67) [363].
The AKT/mTOR pathway plays an important role in dendrite and spine morphogenesis, and synaptic transmission [57], partly through the release of cytokines [364], which in turn can activate the mTOR pathway. These events are implicated in the loss of synapses seen in DS and AD. Finally, the PI3K/AKT pathway, which can be induced by growth factors acting on their tyrosine kinase receptors, plays an important role in synaptic development [365,366].
In summary, the abnormal activation of multiple pathways and their synergic actions seem to be responsible for the early synaptic dysfunction found in DS and AD.

9.3. Receptors

Several lines of research demonstrate the important role of G-protein coupled receptors (GPCRs) in the altered signaling pathways found in AD and DS. In these conditions, GPCRs are implicated in tau hyperphosphorylation through several downstream kinases including GSK3β, CDK-5, and ERK signaling cascades [367]. An imbalance in tau phosphorylation mediated by GPCR-mediated kinases occurs in AD [368]. Several GPCRs have been associated with this imbalance including i) muscarinic ACh receptors [158], for which the number is reduced in AD, leading to enhanced phosphorylation of tau; ii) the CXCR2 and CC3 chemokine receptors, for which activation is implicated in the inflammatory response [367] and tau phosphorylation [369] and which are upregulated in AD [370]; and iii) the metabotropic glutamate receptor 2 (mGluR2) that activates the ERK pathway [371] and is overexpressed in AD leading to tau phosphorylation [367,372]. However, other receptors also play important roles in AD neuropathology [see 367].
In summary, in AD, in individuals with or without DS, alterations in intercellular signalling, including inhibited neurotransmitter release, a reduced number of synapses and alterations in their function, and altered expression of different GPCRs, interfere with neurotransmission, synaptic plasticity, and cognitive function and play important roles in the onset and aggravation of AD pathology.

10. Therapies Targeting Different Pathways Implicated in AD Pathology

Currently, there is no effective treatment to prevent or delay AD in individuals with or without DS, and the only approved drugs, AChE inhibitors and memantine, exert limited symptomatic benefits. Thus, a great effort is being made to search for strategies that prevent or delay the course of the disease. Because of the complex pathology that appears sequentially or simultaneously in AD, a great number and diverse types of therapeutic strategies that target the different alterations found in this disorder are being tested. One of the main problems encountered has been the inability to replicate in humans the efficacy of the different strategies demonstrated in preclinical studies [373]. This section summarizes the current state of the most relevant therapies that target different pathways mentioned in this review.
First, to prevent the formation of amyloid plaques, active and passive immunotherapies that avoid the formation of Aβ by inhibiting BACE that reduce the aggregation of these oligomers into plaques or that facilitate the clearance of Aβ peptides have been developed [374]. Although many of the clinical trials failed due to severe side effects or to its inefficacy, numerous new immunotherapies are currently being tested [374].
Also, several inhibitors of the DYRK1A kinase have been demonstrated to reduce the neuropathology and to improve the cognitive abilities of mouse models of DS and AD [334]. One of them, (-)-Epigallocatechin gallate (EGCG), has been approved for use in the young-adult DS population. However, its ability to enhance the cognitive abilities of these individuals is very controversial, and so far, this molecule has not been tested in individuals with DS and AD. Besides, concerns have been raised about the safety of chronically inhibiting DYRK1A because of its multiple roles in numerous signalling pathways [334].
Although accumulated evidence indicates that RCAN1 might be a potential target for the treatment of AD and DS, so far, a drug able to inhibit RCAN1 has not been developed. However, Zmijewski et al. [375] demonstrated that fish oil supplementation reduced the levels of this protein in mice. Nonetheless, an important issue to take into account with compounds that inhibit calcineurin is that they are immunosuppressive. The therapeutic use for organ transplantation of calcineurin/NFAT inhibitors is associated with severe side effects [376].
Another potential therapeutic target for AD is the mTOR pathway. Numerous studies have demonstrated the ability of mTOR inhibitors, including rapamycin and its analogs, to reduce Aβ load, tau pathology, and cognitive decline in mouse models of AD. However, because these inhibitors induce adverse effects due to the role of mTOR in cell growth and proliferation, metabolism, and protein synthesis, they have not been tested in humans [377].
As explained in previous sections, CREB activation is reduced in AD, resulting in a synaptic and memory impairment. Thus, different strategies to enhance CREB activity have been tested in AD models. Among them, the phosphodiesterase 4 inhibitor rolipram [378] and dietary supplementation with different procyanidins, the main group of flavonoids, have been demonstrated to rescue different neuropathological characteristics of AD and to enhance cognition in animal models of this disorder [379]. However, because of the great number of roles that CREB plays in many tissues, chronic CREB activation could induce important adverse effects [378].
Among the most promising strategies to treat AD are the ones that target tau pathology. Different drugs have been developed to reduce tau translation, posttranslational modifications, aggregations, and impairments in clearance (see [380]). Besides the toxic effects that tau exerts on cells, it is also a mediator of Aβ toxicity; thus, reducing tau pathology could also help to minimize the main hallmark of AD. Despite the efficacy of some strategies that reduce tau expression such as small interfering mRNAs (siRNA) in preclinical models, no clinical trials have been performed in the AD population. Among the drugs that target tau protein modifications are (i) phosphatase inhibitors, such as the NMDA receptor antagonist memantine that produces benefits in AD patients and sodium selenate that increases PP2A activity and is currently been evaluated in a phase II clinical trial; (ii) kinase inhibitors, such as the CDK5 inhibitors flavopiridol and roscovitine that has not been tested in clinical trials in AD patients, tideglusib that does not produce improvements in the AD population and lithium chloride, and a GSK3β inhibitor, which stabilized the cognitive symptoms in AD patients; (iii) drugs that inhibit tau acetylation such a salsalate, a small-molecule NSAIDs; (iv) drugs that inhibit tau deglycosylation such as MK-8719; and (v) molecules that inhibit tau truncation. However, the efficacy of the last three strategies has not been demonstrated in clinical trials. Inhibitors of tau aggregation such as methylene blue and curcumin did not produce any clinical benefits in the AD population [380].
Finally, different tau active and passive immunotherapies have been developed. Similar to what was describe in the case of amyloid immunotherapies, although the results of preclinical studies were very promising, important side effects and low efficacy prevented its use in the AD population. However, several clinical trials trying to overcome these issues are being performed [380].
Growth factors such as NGF and BDNF have been proposed as an aid to prevent cholinergic neurodegeneration and other symptoms of AD [381]. Intranasal administration of NGF reduced cholinergic loss and improved cognition in animal models of AD and a clinical trial are currently being performed to assess its efficacy in humans with this condition. Regarding BDNF, preclinical studies in mouse models of DS have also proven to reduce cholinergic loss as well as other AD-related alterations. Interestingly, some of the approved drugs for the symptomatic treatment of AD such as memantine and donepezil increase BDNF levels [381].
Regarding oxidative stress, similar to what has been described with other therapeutic strategies, different preclinical studies have demonstrated the efficacy of numerous antioxidants (e.g., melatonin, Vitamin E, folinic acid, and different mixtures of vitamins and minerals) and clinical trials performed in individuals with AD with or without DS failed to find any benefit on the cognitive status of these patients or in their neuropathological status [186,382]. It is possible that, in the case of DS individuals, because oxidative stress is present from early developmental stages, the administration of antioxidants in later life stages is not able to rescue other well-established neuropathological signs such as amyloid plaques, NFTs, or synapse loss.
Several compounds that target the mitochondrial alterations found in AD such as Mito Q, Skulachev (SkQ1), melatonin, and Sezto–Shiller (SS) tetrapeptide SS31 reduce the neurodegenerative characteristics of mouse models of this disorder and are good candidates to be tested in clinical trials [383].
Antidiabetic drugs have been demonstrated to rescue most of the alterations found in mouse models of AD [384]. Both hypoglycemic agents (including insulin, sulphonylureas, and glinides) and antihyperglycemic agents (including metformin, thiazolidinediones, dipeptidyl peptidase (DPP) IV inhibitors, Glucagon-like peptide-1 (GLP-1) analogs, GLP-1 receptor agonists, and Sodium-Glucose co-transporters (SGLT)-2 inhibitors) reduce protein aggregation, neuroinflammation, and oxidative stress and enhanced neurogenesis, synaptic plasticity, and cognition in AD rodents [384]. Furthermore, clinical trials have demonstrated that intranasal insulin, sulphonylureas metformin, and the GLP-1 analog liraglutide enhance cognition in AD patients [384]. Thus, antidiabetes drugs are currently one of the most promising strategies to treat AD.
The reduction of neuroinflammation in AD patients has been proposed to be a promising stragegy to treat this disease. Chronic nonsteroidal anti-inflammatory drug (NSAID) consumption has been consistently associated with reduced risk for AD [385], and chronic ibuprofen or naproxen consumption delays the progression from mild cognitive impairment to AD. However, short-term treatments with NSAIDs do not reduce the risk of developing AD. Although buprofen treatment reduces amyloid accumulation and tau in these patients, NSAIDs might only be effective in ApoE4 carriers [385].
Another strategy to reduce neuroinflammation in AD brains is to convert microglia from an inflammatory to a phagocytic phenotype that can enhance the clearance of Aβ. One of the drugs that exerts this effect in mouse models of AD is jujuboside A [386]. However, more evidence of its effects must be obtained in preclinical studies before performing clinical trials.
Finally, it has been proposed that a combination of therapeutic approaches targeting different pathological aspects of the disease would be more effective. Various clinical trials combining different disease-modifying therapies and symptomatic therapies are being performed in individuals with AD [387]. This strategy has been extremely useful in the treatment of other complex diseases such as HIV.

11. Concluding Remarks

Two of the main neuropathological characteristics of AD are the accumulation of amyloid plaques and NFTs. However, numerous mechanisms that appear years earlier than these alterations play a crucial role in the onset and aggravation of this disease. These earlier events include neuronal and synaptic loss and dysfunction, enhanced OS, mitochondrial dysfunction, altered energy metabolism, cellular senescence and neuroinflammation, reduced neurogenesis, and altered neurotransmission. For these reasons, AD has been proposed to be a disease with a complex etiology in which these earlier alterations participate in the appearance and accumulation of plaques and tangles, which in turn aggravate the earlier pathological events in a positive feedforward loop. Also, as described in this review, these pathological mechanisms are interrelated. Numerous signalling pathways that regulate these events are altered in AD. Interestingly, many of these pathways are implicated in multiple AD-related neuropathologies. Besides, in many cases, a synergic effect and/or an interaction between these pathways exist. Among the most relevant examples are the numerous adverse effects found in AD brains due to the overexpression of the Hsa21-encoded DYRK1A and RCAN1 kinases or the mTOR pathway and their interactions. Table 1 summarizes the main signalling pathways implicated in each of the neuropathological characteristics of AD mentioned in this review, and Figure 1 depicts these pathways as well as their interconnections. The complex scenario of AD etiopathology suggests that the development of therapies designed to treat this disorder should target the molecular pathways implicated in multiple altered events of the disease. Finally, because of the high prevalence and early appearance of AD in the DS population and the multiple common mechanisms found in both conditions, DS can be considered a useful model to study AD etiopathology and to search for new therapeutic strategies.

12. Key Summary Points

  • AD, in individuals with or without DS, is a disease with a complex set of neuropathological signs.
  • Numerous signalling pathways are implicated in the onset and aggravation of this neuropathology.
  • The same signalling pathway often plays a role in the appearance or progression of different signs of AD.
  • In many cases, synergic effects and feedback loops exist between these pathways.
  • Because of the complex etiopathology of AD and the interrelation between the factors responsible for the symptoms of the disease, therapeutic approaches should combine different targets.

Author Contributions

Writing—review and editing, C.M.-C. and N.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Institute of Research Valdecilla (IDIVAL), grant number: NVAL 19/23, approval date: 1 January 2020.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the writing of the manuscript or in the decision to publish it.

References

  1. Alzheimer’s Association Home Page. Available online: http://Alzh.org (accessed on 18 September 2020).
  2. Antonarakis, S.E.; Skotko, B.G.; Rafii, M.S.; Strydom, A.; Pape, S.E.; Bianchi, D.W.; Sherman, S.L.; Reeves, R.H. Down syndrome. Nat. Rev. Dis. Primers 2020, 6, 9. [Google Scholar] [CrossRef] [PubMed]
  3. Lott, I.T. Neurological phenotypes for Down syndrome across the life span. Prog. Brain Res. 2012, 197, 101–121. [Google Scholar] [PubMed] [Green Version]
  4. Haydar, T.F.; Reeves, R.H. Trisomy 21 and early brain development. Trends Neurosci. 2012, 35, 81–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Teipel, S.J.; Hampel, H. Neuroanatomy of Down syndrome in vivo: A model of preclinical Alzheimer´s disease. Behav. Genet. 2006, 36, 405–415. [Google Scholar] [CrossRef]
  6. Cenini, G.; Dowling, A.L.; Beckett, T.L.; Barone, E.; Mancuso, C.; Murphy, M.P.; LeVine III, H.; Lott, I.T.; Schmitt, F.A.; Butterfield, D.A.; et al. Association between frontal cortex oxidative damage and beta-amyloid as a function of age in Down syndrome. Biochim. Biophys. Acta 2012, 1822, 130–138. [Google Scholar] [CrossRef]
  7. Wilcock, D.M.; Griffin, W.S. Down’s syndrome, neuroinflammation, and Alzheimer neuropathogenesis. J. Neuroinflammation 2013, 10, 84. [Google Scholar] [CrossRef] [Green Version]
  8. Sabbagh, M.N.; Fleisher, A.; Chen, K.; Rogers, J.; Berk, C.; Reiman, E.; Pontecorvo, M.; Mintun, M.; Skovronsky, D.; Jacobson, S.A.; et al. Positron emission tomography and neuropathologic estimates of fibrillar amyloid-β in a patient with Down syndrome and Alzheimer disease. Arch. Neurol. 2011, 68, 1461–1466. [Google Scholar] [CrossRef]
  9. Moreno-Jiménez, E.P.; Flor-García, M.; Terreros-Roncal, J.; Rábano, A.; Cafini, F.; Pallas-Bazarra, N.; Ávila, J.; Llorens-Martín, M. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat. Med. 2019, 25, 554–560. [Google Scholar] [CrossRef]
  10. Kim, J.; Basak, J.M.; Holtzman, D.M. The role of apolipoprotein E in Alzheimer’s disease. Neuron 2009, 63, 287–303. [Google Scholar] [CrossRef] [Green Version]
  11. Chang, K.T.; Min, K.T. Drosophila melanogaster homolog of Down syndrome critical region 1 is critical for mitochondrial function. Nat. Neurosci. 2005, 8, 1577–1585. [Google Scholar] [CrossRef]
  12. Coskun, P.E.; Wyrembak, J.; Derbereva, O.; Melkonian, G.; Doran, E.; Lott, I.T.; Head, E.; Cotman, C.W.; Wallace, D.C. Systemic mitochondrial dysfunction and the etiology of Alzheimer’s disease and down syndrome dementia. J. Alzheimers Dis. 2010, 20, 293–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Butterfield, D.A.; Di Domenico, F.; Swomley, A.M.; Head, E.; Perluigi, M. Redox proteomics analysis to decipher the neurobiology of Alzheimer-like neurodegeneration: Overlaps in Down’s syndrome and Alzheimer’s disease brain. Biochem. J. 2014, 463, 177–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Pesini, A.; Iglesias, E.; Bayona-Bafaluy, M.P.; Garrido-Pérez, N.; Meade, P.; Gaudó, P.; Jiménez-Salvador, I.; Andrés-Benito, P.; Montoya, J.; Ferrer, I.; et al. Brain pyrimidine nucleotide synthesys and Alzheimer disease. Aging 2019, 11, 1–30. [Google Scholar] [CrossRef] [PubMed]
  15. Martínez-Cué, C.; Rueda, N. Cellular senescence in neurodegenerative diseases. Front. Cell. Neurosci. 2020, 14, 16. [Google Scholar] [CrossRef]
  16. Hard, J. Has the amyloid cascade hypothesis for Alzheimer’s disease been proved? Curr. Alzheimer Res. 2006, 3, 71–73. [Google Scholar] [CrossRef]
  17. Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef]
  18. Gitter, B.D.; Cox, L.M.; Rydel, R.E.; May, P.C. Amyloid beta peptide potentiates cytokine secretion by interleukin-1 beta-activated human astrocytoma cells. Proc. Natl. Acad. Sci. USA 1995, 92, 10738–10741. [Google Scholar] [CrossRef] [Green Version]
  19. Chong, Y. Effect of a carboxy-terminal fragment of the Alzheimer’s amyloid precursor protein on expression of proinflammatory cytokines in rat glial cells. Life Sci. 1997, 61, 2323–2333. [Google Scholar] [CrossRef]
  20. Weldon, D.T.; Rogers, S.D.; Ghilardi, J.R.; Finke, M.P.; Cleary, J.P.; O’Hare, E.; Esler, W.P.; Maggio, J.E.; Mantyh, P.W. Fibrillar beta-amyloid induces microglial phagocytosis, expression of inducible nitric oxide synthase, and loss of a select population of neurons in the rat CNS in vivo. J. Neurosci. 1998, 18, 2161–2173. [Google Scholar] [CrossRef] [Green Version]
  21. Eikelenboom, P.; Veerhuis, R.; Scheper, W.; Rozemuller, A.J.M.; van Gool, W.A.; Hoozemans, J.J.M. He significance of neuroinflammation in understanding Alzheimer’s disease. J. Neural. Transm. 2006, 113, 1685–1695. [Google Scholar] [CrossRef]
  22. Sipos, E.; Kurunczi, A.; Kasza, A.; Horváth, J.; Felszeghy, K.; Laroche, S.; Toldi, J.; Párducz, A.; Penke, B.; Penke-Verdier, Z. Beta-amyloid pathology in the entorhinal cortex of rats induces memory deficits: Implications for Alzheimer’s disease. Neuroscience 2007, 147, 28–36. [Google Scholar] [CrossRef]
  23. Wilcock, D.M. Neuroinflammation in the aging Down syndrome brain; Lessons from Alzheimer’s disease. Curr. Gerontol. Geriatr. Res. 2012, 51, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ritchie, S.J.; Hill, W.D.; Marioni., R.E.; Davies, G.; Hagenaars, S.P.; Harris, S.E.; Cox, S.R.; Taylor, A.M.; Corley, J.; Pattie, A.; et al. Polygenic predictors of age-related decline in cognitive ability. Mol. Psychiatry 2019, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Thomas, M.S.C.; Ojinaga Alfageme, O.; D’Souza, H.; Patkee, P.A.; Rutherford, M.A.; Mok, K.Y.; Hardy, J.; Karmiloff-Smith, A.; the LonDownS Consortium. A multi-level developmental approach to exploring individual differences in Down syndrome: Genes, brain, behaviour, and environment. Res. Dev. Disabil. 2020, 104, 103638. [Google Scholar] [CrossRef] [PubMed]
  26. Bu, G. Apolipoprotein E and its receptors in Alzheimer’s disease: Pathways, pathogenesis and therapy. Nat. Rev. Neurosci. 2009, 10, 333–344. [Google Scholar] [CrossRef] [Green Version]
  27. Huang, Y.; Mucke, L. Alzheimer mechanisms and therapeutic strategies. Cell 2012, 148, 1204–1222. [Google Scholar] [CrossRef] [Green Version]
  28. Liu, C.C.; Kanekiyo, T.; Xu, H.; Bu, G. Apolipoprotein E and Alzheimer disease: Risk, mechanisms and therapy. Nat. Rev. Neurol. 2013, 9, 106–118. [Google Scholar] [CrossRef] [Green Version]
  29. Prasher, V.P.; Sajith, S.G.; Rees, S.D.; Patel, A.; Tewari, S.; Schupf, N.; Zigman, W.B. Significant effect of APOE epsilon 4 genotype on the risk of dementia in Alzheimer’s disease and mortality in persons with Down syndrome. Int. J. Geriatr. Psychiatry 2008, 23, 1134–1140. [Google Scholar] [CrossRef] [Green Version]
  30. Tan, C.C.; Yu, J.T.; Tan, M.S.; Jiang, T.; Zhu, X.C.; Tan, L. Autophagy in aging and neurodegenerative diseases: Implications for pathogenesis and therapy. Neurobiol. Aging 2014, 35, 941–957. [Google Scholar] [CrossRef]
  31. Ryoo, S.R.; Cho, H.J.; Lee, H.W.; Jeong, H.K.; Radnaabazar, C.; Kim, Y.S.; Kim, M.-J.; Son, M.-Y.; Seo, H.; Chung, S.-H.; et al. Dual-specificity tyrosine(Y)-phosphorylation regulated kinase 1A-mediated phosphorylation of amyloid precursor protein: Evidence for a functional link between Down syndrome and Alzheimer’s disease. J. Neurochem. 2008, 104, 1333–1344. [Google Scholar] [CrossRef]
  32. Kentrup, H.; Becker, W.; Heukelbach, J.; Wilmes, A.; Schurmann, A.; Huppertz, C.; Kainulainen, K.; Joost, H.G. Dyrk, a dual specificity protein kinase with unique structural features whose activity is dependent on tyrosine residues between subdomains VII and VIII. J. Biol. Chem. 1996, 271, 3488–3495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Smith, D.J.; Zhu, Y.; Zhang, J.; Cheng, J.F.; Rubin, E.M. Construction of a panel of transgenic mice containing a contiguous 2-Mb set of YAC/P1 clones from human chromosome 21q22.2. Genomics 1995, 27, 425–434. [Google Scholar] [CrossRef] [PubMed]
  34. Smith, D.J.; Stevens, M.E.; Sudanagunta, S.P.; Bronson, R.T.; Makhinson, M.; Watabe, A.M.; O’Dell, T.J.; Fung, J.; Weier, H.U.; Cheng, J.F.; et al. Functional screening of 2Mb of human chromosome 21q22.2 in transgenic mice implicates minibrain in learning defects associated with Down syndrome. Nat. Genet. 1997, 16, 28–36. [Google Scholar] [CrossRef] [PubMed]
  35. Altafaj, X.; Dierssen, M.; Baamonde, C.; Martí, E.; Visa, J.; Guimerà, J.; Oset, M.; Gonzáles, J.R.; Flórez, J.; Fillat, C.; et al. Neurodevelopmental delay, motor abnormalities and cognitive deficits in transgenic mice overexpressing Dyrk1A (minibrain), a murine model of Down’s syndrome. Hum. Mol. Genet. 2001, 10, 1915–1923. [Google Scholar] [CrossRef] [Green Version]
  36. Ahn, K.J.; Jeong, H.K.; Choi, H.S.; Ryoo, S.R.; Kim, Y.J.; Goo, J.S.; Choi, S.-Y.; Han, J.-S.; Ha, I.; Song, W.J. DYRK1A BAC transgenic mice show altered synaptic plasticity with learning and memory defects. Neurobiol. Dis. 2006, 22, 463–472. [Google Scholar] [CrossRef]
  37. Kimura, R.; Kamino, K.; Yamamoto, M.; Nuripa, A.; Kida, T.; Kazui, H.; Hashimoto, R.; Tanaka, T.; Kudo, T.; Yamagata, H.; et al. The DYRK1A gene, encoded in chromosome 21 Down syndrome critical region, bridges between beta-amyloid production and tau phosphorylation in Alzheimer disease. Hum. Mol. Genet. 2007, 16, 15–23. [Google Scholar] [CrossRef] [Green Version]
  38. Wegiel, J.; Gong, C.-X.; Hwang, Y.-W. The role of DYRK1A in neurodegenerative diseases. FEBS J. 2011, 278, 236–245. [Google Scholar] [CrossRef]
  39. Ryu, Y.S.; Park, S.Y.; Jung, M.S.; Yoon, S.H.; Kwen, M.Y.; Lee, S.Y.; Choi, S.-H.; Radnaabazar, C.; Kim, M.-K.; Kim, H.; et al. Dyrk1A-mediated phosphorylation of Presenilin 1: A functional link between Down syndrome and Alzheimer’s disease. J. Neurochem. 2010, 115, 574–584. [Google Scholar] [CrossRef]
  40. Ferrer, I.; Gomez-Isla, T.; Puig, B.; Freixes, M.; Ribe, E.; Dalfo, E.; Avila, J. Current advances on different kinases involved in tau phosphorylation, and implications in Alzheimer’s disease and tauopathies. Curr. Alzheimer Res. 2005, 2, 3–18. [Google Scholar] [CrossRef]
  41. Jung, M.S.; Park, J.H.; Ryu, Y.S.; Choi, S.H.; Yoon, S.H.; Kwen, M.Y.; Oh, J.Y.; Song, W.J.; Chung, S.H. Regulation of RCAN1 protein activity by Dyrk1A protein-mediated phosphorylation. J. Biol. Chem. 2011, 286, 40401–40412. [Google Scholar] [CrossRef] [Green Version]
  42. Mattson, M.P. Oxidative stress, perturbed calcium homeostasis, and immune dysfunction in Alzheimer’s disease. J. Neurovirol. 2002, 8, 539–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Asai, M.; Kinjo, A.; Kimura, S.; Mori, R.; Kawajubo, T.; Shirotani, K.; Yagishita, S.; Maruyama, K.; Iwata, N. Perturbed calcineurin-NFAT signaling is associated with the development of Alzheimer’s disease. Biol. Pharm. Bull. 2016, 39, 1646–1652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Davies, K.J.; Ermak, G.; Rothermel, B.A.; Pritchard, M.; Heitman, J.; Ahnn, J.; Henrique-Silva, F.; Crawford, D.; Canaider, S.; Strippoli, P.; et al. Renaming the DSCR1/Adapt78 gene family as RCAN: Regulators of calcineurin. FASEB J. 2007, 21, 3023–3028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Abdul, H.M.; Furman, J.L.; Sama, M.A.; Mathis, D.M.; Norris, C.M. NFATs and Alzheimer’s Disease. Mol. Cell Pharm. 2010, 2, 7–14. [Google Scholar]
  46. Ermak, G.; Morgan, T.E.; Davies, K.J. Chronic overexpression of the calcineurin inhibitory gene DSCR1 (Adapt78) is associated with Alzheimer’s disease. J. Biol. Chem. 2001, 276, 38787–38794. [Google Scholar] [CrossRef] [Green Version]
  47. Arron, J.R.; Winslow, M.M.; Polleri, A.; Chang, C.P.; Wu, H.; Gao, X.; Neilson, J.R.; Chen, L.; Heit, J.J.; Kim, S.K.; et al. NFAT dysregulation by increased dosage of DSCR1 and DYRK1A on chromosome 21. Nature 2006, 441, 595–600. [Google Scholar] [CrossRef]
  48. Di Domenico, F.; Tramutola, A.; Foppoli, C.; Head, E.; Perluigi, M.; Butterfield, A. mTOR in Down syndrome: Role in Aβ and tau neuropathology and transition to Alzheimer disease-like dementia. Free Radic. Biol. Med. 2018, 114, 94–101. [Google Scholar] [CrossRef]
  49. Cho, H.J.; Jin, S.M.; Youn, H.D.; Huh, K.; Mook-Jung, I. Disrupted intracellular calcium regulates BACE1 gene expression via nuclear factor of activated T cells 1 (NFAT 1) signaling. Aging Cell. 2008, 7, 137–147. [Google Scholar] [CrossRef]
  50. Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef] [Green Version]
  51. O’Neill, C. PI3-kinase/Akt/mTOR signaling: Impaired on/off switches in aging, cognitive decline and Alzheimer’s disease. Exp. Gerontol. 2013, 48, 647–653. [Google Scholar] [CrossRef]
  52. Holtzman, D.M.; Morris, J.C.; Goate, A.M. Alzheimer’s disease: The challenge of the second century. Sci. Transl. Med. 2011, 3, 77sr1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Pei, J.J.; Braak, H.; An, W.L.; Winblad, B.; Cowburn, R.F.; Iqbal, K.; Grundke-Iqbal, I. Up-regulation ofmitogen-activated protein kinases ERK1/2 andMEK1/2 is associated with the progression of neurofibrillary degeneration in Alzheimer’s disease. Mol. Brain Res. 2002, 109, 45–55. [Google Scholar] [CrossRef]
  54. Swatton, J.E.; Sellers, L.A.; Faull, R.L.M.; Holland, A.; Iritani, S.; Bahn, S. Increased MAP kinase activity in Alzheimer’s and Down syndrome but not in schizophrenia human brain. Eur. J. Neurosci. 2004, 19, 2711–2719. [Google Scholar] [CrossRef] [PubMed]
  55. Ma, T.; Hoeffer, C.A.; Capetillo-Zarate, E.; Yu, F.; Wong, H.; Lin, M.T.; Tampellini, D.; Klann, E.; Blitzer, R.D.; Gouras, G.K. Dysregulation of the mTOR pathway mediates impairment of synaptic plasticity in a mouse model of Alzheimer’s disease. PLoS ONE 2010, 5. [Google Scholar] [CrossRef] [PubMed]
  56. Iyer, A.M.; van Scheppingen, J.; Milenkovic, I.; Anink, J.J.; Adle- Biassette, H.; Kovacs, G.G.; Aronica, E. mTOR Hyperactivation in down syndrome hippocampus appears early during development. J. Neuropathol. Exp. Neurol. 2014, 73, 671–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Troca-Marin, J.A.; Casanas, J.J.; Benito, I.; Montesinos, M.L. The Akt-mTOR pathway in Down’s syndrome: The potential use of rapamycin/rapalogs for treating cognitive deficits. CNS Neurol. Disord.-Drug Targets 2014, 13, 34–40. [Google Scholar] [CrossRef]
  58. Griffin, R.J.; Moloney, A.; Kelliher, M.; Johnston, J.A.; Ravid, R.; Dockery, P.; O’Connor, R.; O’Neill, C. Activation of Akt/PKB, increased phosphorylation of Akt substrates and loss and altered distribution of Akt and PTEN are features of Alzheimer’s disease pathology. J. Neurochem. 2005, 93, 105–117. [Google Scholar] [CrossRef]
  59. Li, X.; Alafuzoff, I.; Soininen, H.; Winblad, B.; Pei, J.J. Levels of mTOR and its downstream targets 4E-BP1, eEF2, and eEF2 kinase in relationships with tau in Alzheimer’s disease brain. FEBS J. 2005, 272, 4211–4220. [Google Scholar] [CrossRef]
  60. Peil, J.J.; Hugon, J. mTOR-dependent signalling in Alzheimer’s disease. J. Cell Mol. Med. 2008, 12, 2525–2532. [Google Scholar]
  61. Sun, Y.X.; Ji, X.; Mao, X.; Xie, L.; Jia, J.; Galvan, V.; Greenberg, D.A.; Jin, K. Differential activation of mTOR complex 1 signaling in human brain with mild to severe Alzheimer’s disease. J. Alzheimers Dis. 2014, 38, 437–444. [Google Scholar] [CrossRef]
  62. Martin, D.; Salinas, M.; Lopez-Valdaliso, R.; Serrano, E.; Recuero, M.; Cuadrado, A. Effect of the Alzheimer amyloid fragment Abeta(25-35) on Akt/PKB kinase and survival of PC12 cells. J. Neurochem. 2001, 78, 1000–1008. [Google Scholar] [CrossRef] [PubMed]
  63. Wei, W.L.; Norton, D.D.; Wang, X.T.; Kusiak, J.W. A beta 17-42 in Alzheimer’s disease activates JNK and caspase-8 leading to neuronal apoptosis. Brain 2002, 125, 2036–2043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Perluigi, M.; Pupo, G.; Tramutola, A.; Cini, C.; Coccia, R.; Barone, E.; Head, E.; Butterfield, D.A.; Di Domenico, F. Neuropathological role of PI3K/Akt/mTOR axis in Down syndrome brain. Biochim. Biophys. Acta 2014, 1842, 1144–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Cai, Z.; Zhao, B.; Li, K.; Zhang, L.; Li, C.; Quazi, S.H.; Tan, Y. Mammalian target of rapamycin: A valid therapeutic target through the autophagy pathway for Alzheimer’s disease? J. Neurosci. Res. 2012, 90, 1105–1118. [Google Scholar] [CrossRef] [PubMed]
  66. Cai, Z.; Chen, G.; He, W.; Xiao, M.; Yan, L.J. Activation of mTOR: A culprit of Alzheimer’s disease? Neuropsychiatr. Dis. Treat. 2015, 11, 1015–1030. [Google Scholar] [CrossRef] [Green Version]
  67. Caccamo, A.; Majumder, S.; Richardson, A.; Strong, R.; Oddo, S. Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: Effects on cognitive impairments. J. Biol. Chem. 2010, 285, 13107–13120. [Google Scholar] [CrossRef] [Green Version]
  68. Di Domenico, F.; Head, E.; Butterfield, D.A.; Perluigi, M. Oxidative Stress and Proteostasis Network: Culprit and Casualty of Alzheimer’s-Like Neurodegeneration. Adv. Geriatr. 2014, 14. [Google Scholar] [CrossRef] [Green Version]
  69. Funderburk, S.F.; Wang, Q.J.; Yue, Z. The Beclin 1–VPS34 complex–at the crossroads of autophagy and beyond. Trends Cell. Biol. 2010, 20, 355–362. [Google Scholar] [CrossRef] [Green Version]
  70. Mizushima, N.; Levine, B.; Cuervo, A.M.; Klionsky, D.J. Autophagy fights disease through cellular self-digestion. Nature 2008, 451, 1069–1075. [Google Scholar] [CrossRef] [Green Version]
  71. Nixon, R.A. Autophagy, amyloidogenesis and Alzheimer disease. J. Cell Sci. 2007, 120, 4081–4091. [Google Scholar] [CrossRef] [Green Version]
  72. Salminen, A.; Kaarniranta, K.; Kauppinen, A.; Ojala, J.; Haapasalo, A.; Soininen, H.; Hiltunen, M. Impaired autophagy and APP processing in Alzheimer’s disease: The potential role of Beclin 1 interactome. Prog. Neurobiol. 2013, 106–107, 33–54. [Google Scholar] [CrossRef] [PubMed]
  73. Caccamo, A.; Maldonado, M.A.; Majumder, S.; Medina, D.X.; Holbein, W.; Magri, A.; Oddo, S. Naturally secreted amyloid-beta increases mammalian target of rapamycin (mTOR) activity via a PRAS40-mediated mechanism. J. Biol. Chem. 2011, 286, 8924–8932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Majumder, S.; Richardson, A.; Strong, R.; Oddo, S. Inducing autophagy by rapamycin before, but not after, the formation of plaques and tangles ameliorates cognitive deficits. PLoS ONE 2011, 6, e25416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Zhao, W.Q.; De Felice, F.G.; Fernandez, S.; Chen, H.; Lambert, M.P.; Quon, M.J.; Krafft, G.A.; Klein, W.L. Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J. 2008, 22, 246–260. [Google Scholar] [CrossRef] [Green Version]
  76. Pláteník, J.; Fišar, Z.; Buchal, R.; Jirák, R.; Kitzlerová, E.; Zvěřová, M.; Raboch, J. GSK3β, CREB, and BDNF in peripheral blood of patients with Alzheimer’s disease and depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2014, 50, 83–93. [Google Scholar] [CrossRef]
  77. Walton, M.R.; Dragunow, M. Is CREB a key to neuronal survival? Trends Neurosci. 2000, 23, 48–53. [Google Scholar] [CrossRef]
  78. Grimes, C.A.; Jope, R.S. CREB DNA binding activity is inhibited by glycogen synthase kinase-3β and facilitated by lithium. J. Neurochem. 2001, 78, 1219–1232. [Google Scholar] [CrossRef] [Green Version]
  79. Wang, Q.M.; Roach, P.J.; Fiol, C.J. Use of a synthetic peptide as a selective substrate for glycogen synthase kinase 3. Anal. Biochem. 1994, 220, 397–402. [Google Scholar] [CrossRef]
  80. DaRocha-Souto, B.; Coma, M.; Perez-Nievas, B.; Scotton, T.; Siao, M.; Sánchez-Ferrer, P.; Hashimoto, T.; Fan, Z.; Hudry, E.; Barroeta, I. Activation of glycogen synthase kinase-3 beta mediates β-amyloid induced neuritic damage in Alzheimer’s disease. Neurobiol. Dis. 2012, 45, 425–437. [Google Scholar] [CrossRef] [Green Version]
  81. Tong, T.; Thornton, P.L.; Balazs, R.; Cotman, C.W. β-amyloid-(1–42) impairs activity-dependent cAMPresponse element-binding protein signaling in neurons at concentrations in which cell survival is not compromised. J. Biol. Chem. 2001, 276, 17301–17306. [Google Scholar] [CrossRef] [Green Version]
  82. Vitolo, O.V.; Sant’Angelo, A.; Costanzo, V.; Battaglia, F.; Arancio, O.; Shelanski, M. Amyloid β-peptide inhibition of the PKA/CREB pathway and long-term potentiation: Reversibility by drugs that enhance cAMP signaling. Proc. Natl. Acad. Sci. USA 2002, 99, 13217–13221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Cowburn, R.F.; O’Neill, C.; Ravid, R.; Alafuzoff, I.; Winblad, B.; Fowler, C.J. Adenylyl cyclase activity in postmortem human brain: Evidence of altered G protein mediation in Alzheimer’s disease. J. Neurochem. 1992, 58, 1409–1419. [Google Scholar] [CrossRef] [PubMed]
  84. Schnecko, A.; Witte, K.; Bohl, J.; Ohm, T.; Lemmer, B. Adenylyl cyclase activity in Alzheimer’s disease brain: Stimulatory and inhibitory signal transduction pathways are differently affected. Brain Res. 1994, 644, 291–296. [Google Scholar] [CrossRef]
  85. Gong, B.; Cao, Z.; Zheng, P.; Vitolo, O.V.; Liu, S.; Staniszewski, A.; Moolman, D.; Zhang, H.; Shelanski, M.; Arancio, O. Ubiquitin hydrolase Uch-L1 rescues β-amyloid-induced decreases in synaptic function and contextual memory. Cell 2006, 126, 775–788. [Google Scholar] [CrossRef] [Green Version]
  86. Smith, D.L.; Pozueta, J.; Gong, B.; Arancio, O.; Shelanski, M. Reversal of long-term dendritic spine alterations in Alzheimer disease models. Proc. Natl. Acad. Sci. USA 2009, 106, 16877–16882. [Google Scholar] [CrossRef] [Green Version]
  87. Rosa, E.; Fahnestock, M. CREB expression mediates amyloid β-induced basal BDNF downregulation. Neurobiol. Aging 2015, 36, 2406–2413. [Google Scholar] [CrossRef]
  88. Connor, B.; Young, D.; Yan, Q.; Faull, R.; Synek, B.; Dragunow, M. Brain-derived neurotrophic factor is reduced in Alzheimer’s disease. Mol. Brain Res. 1997, 49, 71–81. [Google Scholar] [CrossRef]
  89. Hock, C.; Heese, K.; Hulette, C.; Rosenberg, C.; Otten, U. Region-specific neurotrophin imbalances in Alzheimer disease: Decreased levels of brain-derived neurotrophic factor and increased levels of nerve growth factor in hippocampus and cortical areas. Arch. Neurol. 2000, 57, 846–851. [Google Scholar] [CrossRef] [Green Version]
  90. Lee, H.-G.; Zhu, X.; Casadesus, G.; Pallas, M.; Camins, A.; O’Neill, M.J.; Nakanishi, S.; Perry, G.; Smith, M.A. The effect of mGluR2 activation on signal transduction pathways and neuronal cell survival. Brain Res. 2009, 1249, 244–250. [Google Scholar] [CrossRef] [Green Version]
  91. Cowansage, K.K.; LeDoux, J.E.; Monfils, M.-H. Brain-derived neurotrophic factor: A dynamic gatekeeper of neural plasticity. Curr. Mol. Pharmacol. 2010, 3, 12–29. [Google Scholar] [CrossRef]
  92. Fahnestock, M. Brain-derived neurotrophic factor: The link between amyloid-β and memory loss. Future Neurol. 2011, 6, 627–639. [Google Scholar] [CrossRef]
  93. Peng, S.; Wuu, J.; Mufson, E.J.; Fahnestock, M. Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer’s disease. J. Neurochem. 2005, 93, 1412–1421. [Google Scholar] [CrossRef] [PubMed]
  94. Garzon, D.J.; Fahnestock, M. Oligomeric amyloid decreases basal levels of brain-derived neurotrophic factor (BDNF) mRNA via specific downregulation of BDNF transcripts IV and V in differentiated human neuroblastoma cells. J. Neurosci. 2007, 27, 2628–2635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Chaves, J.C.S.; Machado, F.T.; Almeida, M.F.; Bacovsky, T.B.; Ferrari, M.F.R. microRNAs expression correlates with levels of APP, DYRK1A, hyperphosphorylated Tau and BDNF in the hippocampus of a mouse model for Down syndrome during ageing. Neurosci. Lett. 2020, 714, 134541. [Google Scholar] [CrossRef]
  96. Andrade-Talavera, Y.; Benito, I.; Casañas, J.J.; Rodríguez-Moreno, A.; Montesinos, M.L. Rapamycin restores BDNF-LTP and the persistence of long-term memory in a model of Down’s syndrome. Neurobiol. Dis. 2015, 82, 16–525. [Google Scholar] [CrossRef] [PubMed]
  97. Budni, J.; Bellettini-Santos, T.; Mina, F.; Garcez, M.L.; Zugno, A.I. The involvement of BDNF, NGF and GDNF in aging and Alzheimer’s disease. Aging Dis. 2015, 6, 331. [Google Scholar] [CrossRef] [Green Version]
  98. Bruno, M.A.; Mufson, E.J.; Wuu, J.; Cuello, A.C. Increased matrix metalloproteinase 9 activity in mild cognitive impairment. J. Neuropathol. Exp. Neurol. 2009, 68, 1309–1318. [Google Scholar] [CrossRef]
  99. Iulita, M.F.; Do Carmo, S.; Ower, A.K.; Fortress, A.M.; Flores Aguilar, L.; Hanna, M.; Wisniewski, T.; Granholm, A.C.; Buhusi, M.; Busciglio, J.; et al. Nerve growth factor metabolic dysfunction in Down’s syndrome brains. Brain 2014, 137, 860–872. [Google Scholar] [CrossRef] [Green Version]
  100. Matrone, C.; Ciotti, M.T.; Mercanti, D.; Marolda, R.; Calissano, P. NGF and BDNF signaling control amyloidogenic route and Abeta production in hippocampal neurons. Proc. Natl. Acad. Sci. USA 2008, 105, 13139–13144. [Google Scholar] [CrossRef] [Green Version]
  101. España, J.; Valero, J.; Miñano-Molina, A.J.; Masgrau, R.; Martín, E.; Guardia-Laguarta, C.; Lleó, A.; Giménez-Llort, L.; Rodríguez-Alvarez, J.; Saura, C.A. Amyloid disrupts activity-dependent gene transcription required for memory through the CREB coactivator CRTC1. J. Neurosci. 2010, 30, 9402–9410. [Google Scholar]
  102. Pugazhenthi, S.; Wang, M.; Pham, S.; Sze, C.-I.; Eckman, C.B. Downregulation of CREB expression in Alzheimer’s brain and in Aβ-treated rat hippocampal neurons. Mol. Neurodegener. 2011, 6, 60. [Google Scholar] [CrossRef] [PubMed]
  103. Allen, S.J.; Watson, J.J.; Dawbarn, D. The neurotrophins and their role in Alzheimer’s disease. Curr. Neuropharmacol. 2011, 9, 559–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lu, B. BDNF and activity-dependent synaptic modulation. Learn. Mem. 2003, 10, 86–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Dineley, K.T.; Westerman, M.; Bui, D.; Bell, K.; Ashe, K.H.; Sweatt, J.D. β-Amyloid activates the mitogenactivated protein kinase cascade via hippocampal α7 nicotinic acetylcholine receptors: In vitro and in vivo mechanisms related to Alzheimer’s disease. J. Neurosci. 2001, 21, 4125–4133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Dineley, K.T.; Xia, X.; Bui, D.; Sweatt, J.D.; Zheng, H. Accelerated plaque accumulation, associative learning deficits, and up-regulation of α7 nicotinic receptor protein in transgenic mice co-expressing mutant human presenilin 1 and amyloid precursor proteins. J. Biol. Chem. 2002, 277, 22768–22780. [Google Scholar] [CrossRef] [Green Version]
  107. Gong, B.; Vitolo, O.V.; Trinchese, F.; Liu, S.; Shelanski, M.; Arancio, O. Persistent improvement in synaptic and cognitive functions in an Alzheimer mouse model after rolipram treatment. J. Clin. Investig. 2004, 114, 1624–1634. [Google Scholar] [CrossRef] [Green Version]
  108. Bancher, C.; Grundke-Iqbal, I.; Iqbal, K.; Fried, V.A.; Smith, H.T.; Wisniewski, H.M. Abnormal phosphorylation of tau precedes ubiquitination in neurofibrillary pathology of Alzheimer disease. Brain Res. 1991, 539, 11–18. [Google Scholar] [CrossRef]
  109. Grundke-Iqbal, I.; Iqbal, K.; Tung, Y.C.; Quinlan, M.; Wisniewski, H.M.; Binder, L.I. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. USA 1986, 83, 4913–4917. [Google Scholar] [CrossRef] [Green Version]
  110. Hanger, D.P.; Brion, J.P.; Gallo, J.M.; Cairns, N.J.; Luthert, P.J.; Anderton, B.H. Tau in Alzheimer’s disease and Down’s syndrome is insoluble and abnormally phosphorylated. Biochem. J. 1991, 275, 99–104. [Google Scholar] [CrossRef] [Green Version]
  111. Cardenas, A.M.; Ardiles, A.O.; Barraza, N.; Baez-Matus, X.; Caviedes, P. Role of tau protein in neuronal damage in Alzheimer’s disease and Down syndrome. Arch. Med. Res. 2012, 43, 645–654. [Google Scholar] [CrossRef]
  112. Ksiezak-Reding, H.; Liu, W.-K.; Yen, S.-H. Phosphate analysis and dephosphorylation of modified tau associated with paired helical filaments. Brain Res. 1992, 597, 209–219. [Google Scholar] [CrossRef]
  113. Kimura, T.; Yamashita, S.; Fukuda, T.; Park, J.M.; Murayama, M.; Mizoroki, T.; Yoshiike, Y.; Sahara, N.; Takashima, A. Hyperphosphorylated tau in parahippocampal cortex impairs place learning in aged mice expressing wild-type human tau. EMBO J. 2007, 26, 5143–5152. [Google Scholar] [CrossRef] [Green Version]
  114. Head, E.; Lott, I.T.; Hof, P.R.; Bouras, C.; Su, J.H.; Kim, R.; Haier, R.; Cotman, C.W. Parallel compensatory and pathological events associated with tau pathology in middle aged individuals with Down syndrome. J. Neuropathol. Exp. Neurol. 2003, 62, 917–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Liu, F.; Lian, Z.; Wegiel, J.; Hwang, Y.W.; Iqbal, K.; Grundke-Iqbal, I.; Ramakrishna, N.; Gong, C.-X. Overexpression of Dyrk1A contributes to neurofibrillary degeneration in Down syndrome. FASEB J. 2008, 22, 3224–3233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Shukkur, E.A.; Shimohata, A.; Akagi, T.; Yu, W.; Yamaguchi, M.; Murayama, M.; Chui, D.; Takeuchi, T.; Amano, K.; Subramhanya, K.H.; et al. Mitochondrial dysfunction and tau hyperphosphorylation in Ts1Cje, a mouse model for Down syndrome. Hum. Mol. Genet. 2006, 15, 2752–2762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Woods, Y.L.; Cohen, P.; Becker, W.; Jakes, R.; Goedert, M.; Wang, X.; Proud, C.G. The kinase DYRK phosphorylates protein-synthesis initiation factor eIF2Bepsilon at Ser539 and the microtubule-associated protein tau at Thr212: Potential role for DYRK as a glycogen synthase kinase 3-priming kinase. Biochem. J. 2001, 355, 609–615. [Google Scholar] [CrossRef]
  118. Azorsa, D.O.; Robeson, R.H.; Frost, D.; Meec Hoovet, B.; Brautigam, G.R.; Dickey, C.; Beaudry, C.; Basu, G.D.; Holz, D.R.; Hernandez, J.A.; et al. High-content siRNA screening of the kinome identifies kinases involved in Alzheimer’s disease-related tau hyperphosphorylation. BMC Genom. 2010, 11, 25. [Google Scholar] [CrossRef] [Green Version]
  119. Frost, D.; Meechoovet, B.; Wang, T.; Gately, S.; Giorgetti, M.; Shcherbakova, I.; Dunckley, T. beta-carboline compounds, including harmine, inhibit DYRK1A and tau phosphorylation at multiple Alzheimer’s disease-related sites. PLoS ONE 2011, 6, e19264. [Google Scholar] [CrossRef] [Green Version]
  120. Yin, X.; Jin, N.; Gu, J.; Shi, J.; Zhou, J.; Gong, C.X.; Iqbal, K.; Grundke-Iqbal, I.; Liu, F. Dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1A) modulates serine/arginine-rich protein 55 (SRp55)-promoted Tau exon 10 inclusion. J. Biol. Chem. 2012, 287, 30497–30506. [Google Scholar] [CrossRef] [Green Version]
  121. Mancini, M.; Toker, A. NFAT proteins: Emerging roles in cancer progression. Nat. Rev. Cancer 2009, 9, 810–820. [Google Scholar] [CrossRef] [Green Version]
  122. Wu, H.; Peisley, A.; Graef, I.A.; Crabtree, G.R. NFAT signaling and the invention of vertebrates. Trends Cell Biol. 2007, 17, 251–260. [Google Scholar] [CrossRef] [PubMed]
  123. Goedert, M.; Spillantini, M.G.; Jakes, R.; Rutherford, D.; Crowther, R.A. Multiple isoforms of humanmicrotubule-associated protein tau: Sequences and localization in neurofibrillary tangles of Alzheimer’s disease. Neuron 1989, 3, 519–526. [Google Scholar] [CrossRef]
  124. Shi, J.; Zhang, T.; Zhou, C.; Chohan, M.O.; Gu, X.; Wegiel, J.; Zhou, J.; Hwang, Y.-W.; Iqbal, K.; Drundke-Iqbal, I.; et al. Increased dosage of Dyrk1A alters alternative splicing factor (ASF)-regulated alternative splicing of tau in Down syndrome. J. Biol. Chem. 2008, 283, 28660–28669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Wegiel, J.; Kaczmarski, W.; Barua, M.; Kuchna, I.; Nowicki, K.; Wang, K.C.; Wegiel, J.; Ma, S.Y.; Franckowiak, J.; Mazur-Kolecka, B.; et al. Link between DYRK1A overexpression and several-fold enhancement of neurofibrillary degeneration with 3-repeat tau protein in Down syndrome. J. Neuropathol. Exp. Neurol. 2011, 70, 36–50. [Google Scholar] [CrossRef] [Green Version]
  126. Ermak, G.; Pritchard, M.A.; Dronjak, S.; Niu, B.; Davies, K.J. Do RCAN1 proteins link chronic stress with neurodegeneration? FASEB J. 2011, 25, 3306–3311. [Google Scholar] [CrossRef] [Green Version]
  127. Poppek, D.; Keck, S.; Ermak, G.; Jung, T.; Stolzing, A.; Ullrich, O.; Davies, K.J.A.; Grune, T. Phosphorylation inhibits turnover of the tau protein by the proteasome: Influence of RCAN1 and oxidative stress. Biochem. J. 2006, 400, 511–520. [Google Scholar] [CrossRef] [Green Version]
  128. Lloret, A.; Badia, M.C.; Giraldo, E.; Ermak, G.; Alonso, M.-D.; Pallardó, F.V.; Davies, K.J.A.; Viña, J. Amyloid-beta toxicity and tau hyperphosphorylation are linked via RCAN1 in Alzheimer’s Disease. J. Alzheimers Dis. 2011, 27, 701–709. [Google Scholar] [CrossRef] [Green Version]
  129. Ermak, G.; Harris, C.D.; Battocchio, D.; Davies, K.J. RCAN1 (DSCR1 or Adapt78) stimulates expression of GSK-3beta. FEBS J. 2006, 273, 2100–2109. [Google Scholar] [CrossRef]
  130. Reynolds, C.H.; Betts, J.C.; Blackstock, W.P.; Nebreda, A.R.; Anderton, B.H. Phosphorylation sites on tau identified by nanoelectrospray mass spectrometry: Differences in vitro between the mitogen-activated protein kinases ERK2, c-Jun Nterminal kinase and P38, and glycogen synthase kinase-3b. J. Neurochem. 2000, 74, 1587–1595. [Google Scholar] [CrossRef]
  131. Takashima, A.; Honda, T.; Yasutake, K.; Michel, G.; Murayama, O.; Murayama, M.; Ishiguro, K.; Yamaguchi, H. Activation of tau protein kinaseI/glycogen synthase kinase-3beta by amyloid beta peptide enhances phosphorylation of tau in hippocampal neurons. Neurosci. Res. 1998, 31, 317–323. [Google Scholar] [CrossRef]
  132. Qing, H.; He, G.; Ly, P.T.; Fox, C.J.; Staufenbiel, M.; Cai, F.; Zhang, Z.; Wei, S.; Sun, X.; Chen, C.-H.; et al. Valproic acid inhibits Abeta production, neuritic plaque formation, and behavioral deficits in Alzheimer’s disease mouse models. J. Exp. Med. 2008, 205, 2781–2789. [Google Scholar] [CrossRef] [PubMed]
  133. Pollonini, G.; Gao, V.; Rabe, A.; Palminiello, S.; Albertini, G.; Alberini, C.M. Abnormal expression of synaptic proteins and neurotrophin-3 in the Down syndrome mouse model Ts65Dn. Neuroscience 2008, 156, 99–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Cruz, J.C.; Tseng, H.-C.; Goldman, J.A.; Shih, H.; Tsai, L.-H. Aberrant Cdk5 activation by p25 triggers pathological events leading to neurodegeneration and neurofibrillary tangles. Neuron 2003, 40, 471–483. [Google Scholar] [CrossRef] [Green Version]
  135. Li, T.; Hawkes, C.; Qureshi, H.Y.; Kar, S.; Paudel, H.K. Cyclindependent protein kinase 5 primes microtubule-associated protein tau site-specifically for glycogen synthase kinase 3b. Biochemistry 2006, 45, 3134–3145. [Google Scholar] [CrossRef] [PubMed]
  136. Liang, Z.; Liu, F.; Iqbal, K.; Grundke-Iqbal, I.; Wegiel, J.; Gong, C.X. Decrease of protein phosphatase 2A and its association with accumulation and hyperphosphorylation of tau in Down syndrome. J. Alzheimers Dis. 2008, 13, 295–302. [Google Scholar] [CrossRef] [Green Version]
  137. Sontag, J.M.; Sontag, E. Protein phosphatase 2A dysfunction in Alzheimer’s disease. Front. Mol. Neurosci. 2014, 7, 16. [Google Scholar] [CrossRef]
  138. Chohan, M.O.; Khatoon, S.; Iqbal, I.G.; Iqbal, K. Involvement of I2PP2A in the abnormal hyperphosphorylation of tau and its reversal by Memantine. FEBS Lett. 2006, 580, 3973–3979. [Google Scholar] [CrossRef] [Green Version]
  139. Liu, F.; Grundke-Iqbal, I.; Iqbal, K.; Gong, C.X. Contributions of protein phosphatases PP1, PP2A, PP2B and PP5 to the regulation of tau phosphorylation. Eur. J. Neurosci. 2005, 22, 1942–1950. [Google Scholar] [CrossRef]
  140. Gong, C.X.; Singh, T.J.; Grundkeiqbal, I.; Iqbal, K. Phosphoprotein phosphatase-Activities in alzheimer-disease brain. J. Neurochem. 1993, 61, 921–927. [Google Scholar] [CrossRef]
  141. Gong, C.X.; Shaikh, S.; Wang, J.Z.; Zaidi, T.; Grundkeiqbal, I.; Iqbal, K. Phosphataseactivity toward abnormally phosphorylated-tau - decrease in alzheimer-disease brain. J. Neurochem. 1995, 65, 732–738. [Google Scholar] [CrossRef]
  142. Sun, L.; Liu, S.Y.; Zhou, X.W.; Wang, X.C.; Liu, R.; Wang, Q.; Wang, J.C. Inhibition of protein phosphatase 2A- and protein phosphatase 1-induced tau hyperphosphorylation and impairment of spatial memory retention in rats. Neuroscience 2003, 118, 1175–1182. [Google Scholar] [CrossRef]
  143. Julien, C.; Tremblay, C.; Emond, V.; Lebbadi, M.; Salem, N.; Bennett, D.A.; Calon, F. Sirtuin 1 reduction parallels the accumulation of tau in Alzheimer disease. J. Neuropathol. Exp. Neurol. 2009, 68, 48–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Tramutola, A.; Pupo, G.; Di Domenico, F.; Barone, E.; Arena, A.; Lanzillotta, C.; Brokeaart, D.; Blarzino, C.; Head, E.; Butterfiels, D.A.; et al. Activation of p53 in Down Syndrome and in the Ts65Dn Mouse Brain is Associated with a Pro-Apoptotic Phenotype. J. Alzheimers Dis. 2016, 52, 359–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Guo, X.; Williams, J.G.; Schug, T.T.; Li, X. DYRK1A and DYRK3 promote cell survival through phosphorylation and activation of SIRT1. J. Biol. Chem. 2010, 285, 13223–13232. [Google Scholar] [CrossRef] [Green Version]
  146. Counts, S.E.; Mufson, E.J. The role of nerve growth factor receptors in cholinergic basal forebrain degeneration in prodromal Alzheimer disease. J. Neuropathol. Exp. Neurol. 2005, 64, 263–272. [Google Scholar] [CrossRef] [Green Version]
  147. Forman, M.S.; Mufson, E.J.; Leurgans, S.; Pratico, D.; Joyce, S.; Leight, S.; Lee, M.-Y.; Trojanowski, Q. Cortical biochemistry in MCI and Alzheimer disease: Lack of correlation with clinical diagnosis. Neurology 2007, 68, 757–763. [Google Scholar] [CrossRef]
  148. Ginsberg, S.D.; Che, S.; Counts, S.E.; Mufson, E.J. Single cell gene expression profiling in Alzheimer’s disease. NeuroRx 2006, 3, 302–318. [Google Scholar] [CrossRef]
  149. Ginsberg, S.D.; Che, S.; Counts, S.E.; Mufson, E.J. Shift in the ratio of three-repeat tau and four-repeat tau mRNAs in individual cholinergic basal forebrain neurons in mild cognitive impairment and Alzheimer’s disease. J. Neurochem. 2006, 96, 1401–1408. [Google Scholar] [CrossRef]
  150. Mesulam, M.M. The cholinergic lesion of Alzheimerˇıs disease: Pivotal factor or side show. Learn. Mem. 2004, 11, 43–49. [Google Scholar] [CrossRef] [Green Version]
  151. Geula, C.; Nagykery, N.; Nicholas, A.; Wu, C.K. Cholinergic neuronal and axonal abnormalities are present early in aging and in Alzheimer disease. J. Neuropathol. Exp. Neurol. 2008, 67, 309–318. [Google Scholar] [CrossRef] [Green Version]
  152. Sassin, I.; Schultz, C.; Thal, D.R.; Rüb, U.; Arai, K.; Braak, E. Evolution of Alzheimer’s disease-related cytoskeletal changes in the basal nucleus of Meynert. Acta Neuropathol. 2000, 100, 259–269. [Google Scholar] [CrossRef] [PubMed]
  153. Belarbi, K.; Schindowski, K.; Burnouf, S.; Caillierez, R.; Grosjean, M.E.; Demeyer, D.; Hamdane, M.; Sergeant, N.; Blum, D.; Buée, L. Early Tau pathology involving the septo-hippocampal pathway in a Tau transgenic model: Relevance to Alzheimer’s disease. Curr. Alzheimer Res. 2009, 6, 152–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Hellström-Lindahl, E.; Moore, H.; Nordberg, A. Increased levels of tau protein in SH-SY5Y cells after treatment with cholinesterase inhibitors and nicotinic agonists. J. Neurochem. 2000, 74, 777–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Buckingham, S.D.; Jones, A.K.; Brown, L.A.; Sattelle, D.B. Nicotinic acetylcholine receptor signaling: Roles in Alzheimer’s disease and amyloid neuroprotection. Pharm. Rev. 2009, 61, 39–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Bencherif, M.; Lippiello, P.M. Alpha7 neuronal nicotinic receptors: The missing link to understanding Alzheimer’s etiopathology? Med. Hypotheses 2010, 74, 281–285. [Google Scholar] [CrossRef]
  157. Wang, H.Y.; Li, W.; Benedetti, N.J.; Lee, D.H. α7 nicotinic acetylcholine receptors mediate β-amyloid peptide-induced tau protein phosphorylation. J. Biol. Chem. 2003, 278, 31547–31553. [Google Scholar] [CrossRef] [Green Version]
  158. Caccamo, A.; Oddo, S.; Billings, L.M.; Green, K.N.; Martinez-Coria, H.; Fisher, A.; LaFerla, F.M. M1 receptors play a central role in modulating AD-like pathology in transgenic mice. Neuron 2006, 49, 671–682. [Google Scholar] [CrossRef] [Green Version]
  159. Ferreira-Vieira, T.H.; Guimaraes, M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s disease: Targeting the cholinergic system. Curr. Neuropharmacol. 2016, 14, 101–115. [Google Scholar] [CrossRef] [Green Version]
  160. Goekoop, R.; Scheltens, P.; Barkhoh, F.; Rombouts, S.A.R.B. Cholinergic challenge in Alzheimer patients and mild cognitive impairment differentially affects hippocampal activation – a pharmacological fMRI study. Brain 2006, 129, 141–157. [Google Scholar] [CrossRef] [Green Version]
  161. Bierer, L.M.; Haroutunian, V.; Gabriel, S.; Knott, P.J.; Carlin, L.S.; Purohit, D.P.; Perl, D.P.; Schmeidler, J.; Kanof, P.; Davis, K.L. Neurochemical correlates of dementia severity in Alzheimer’s disease: Relative importance of the cholinergic deficits. J. Neurochem. 1995, 64, 749–760. [Google Scholar] [CrossRef]
  162. Gsell, W.; Jungkunz, G.; Riederer, P. Functional neurochemistry of Alzheimer’s disease. Curr. Pharm. Des. 2004, 10, 265–293. [Google Scholar] [CrossRef] [PubMed]
  163. Contestabile, A.; Ciani, E.; Contestabile, A. The place of choline acetyltransferase activity measurement in the “cholinergic hypothesis” of neurodegenerative diseases. Neurochem. Res. 2008, 33, 318–327. [Google Scholar] [CrossRef] [PubMed]
  164. Mufson, E.J.; Counts, S.E.; Perez, S.E.; Ginsberg, S.D. Cholinergic system during the progression of Alzheimer’s disease: Therapeutic implications. Exp. Rev. Neurother. 2008, 8, 1703–1718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Burghaus, L.; Schütz, U.; Krempel, U.; de Vos, R.A.; Jansen Steur, E.N.; Wevers, A.; Lindstrom, J.; Schröder, H. Quantitative assessment of nicotinic acetylcholine receptor proteins in the cerebral cortex of Alzheimer patients. Mol. Brain Res. 2000, 76, 385–388. [Google Scholar] [CrossRef]
  166. Mousavi, M.; Hellström-Lindahl, E.; Guan, Z.Z.; Shan, K.R.; Ravid, R.; Nordberg, A. Protein and mRNA levels of nicotinic receptors in brain of tobacco using controls and patients with Alzheimer’s disease. Neuroscience 2003, 122, 515–520. [Google Scholar] [CrossRef]
  167. Nordberg, A. Nicotinic receptor abnormalities of Alzheimer’s disease: Therapeutic implications. Biol. Psychiatry 2001, 49, 200–210. [Google Scholar] [CrossRef]
  168. Wilcock, G.K.; Esiri, M.M.; Bowen, D.M.; Smith, C.C. Alzheimer’s disease. Correlation of cortical choline acetyltransferase activity with the severity of dementia and histological abnormalities. J. Neurol. Sci. 1982, 57, 407–417. [Google Scholar] [CrossRef]
  169. Perry, E.K.; Morris, C.M.; Court, J.A.; Cheng, A.; Fairbairn, A.F.; McKeith, I.G.; Irving, D.; Brown, A.; Perry, R.H. Alteration in nicotine binding sites in Parkinson’s disease. Lewy body dementia and Alzheimer’s disease: Possible index of early neuropathology. Neuroscience 1995, 64, 385–395. [Google Scholar] [CrossRef]
  170. Mufson, E.J.; Counts, S.E.; Fahnestock, M.; Ginsberg, S.D. Cholinotrophic molecular substrates of mild cognitive impairment in the elderly. Curr. Alzheimer Res. 2007, 4, 340–350. [Google Scholar] [CrossRef]
  171. Cuello, A.C.; Bruno, M.A.; Bell, K.F. NGF-cholinergic dependency in brain aging. MCI and Alzheimer’s disease. Curr. Alzheimer Res. 2007, 4, 351–358. [Google Scholar] [CrossRef]
  172. Auld, D.S.; Kornecook, T.J.; Bastianetto, S.; Quirion, R. Alzheimer’s disease and the basal forebrain cholinergic system: Relations to β-amyloid peptides, cognition, and treatment strategies. Prog. Neurobiol. 2002, 68, 209–245. [Google Scholar] [CrossRef]
  173. Yan, Z.; Feng, J. Alzheimer’s disease: Interactions between cholinergic functions and β-amyloid. Curr. Alzheimer Res. 2004, 1, 241–248. [Google Scholar] [CrossRef] [PubMed]
  174. Apelt, J.; Kumar, A.; Schliebs, R. Impairment of cholinergic neurotransmission in adult and aged transgenic Tg2576 mouse brain expressing the Swedish mutation of human beta-amyloid precursor protein. Brain Res. 2002, 953, 17–30. [Google Scholar] [CrossRef]
  175. Di Domenico, F.; Tramutola, A.; Butterfield, D.A. Role of 4-hydroxy-2-nonenal (HNE) in the pathogenesis of alzheimer disease and other selected age-related neurodegenerative disorders. Free Radic. Biol. Med. 2017, 111, 253–261. [Google Scholar] [CrossRef] [PubMed]
  176. Rueda, N.; Martínez-Cué, C. Antioxidants in Down Syndrome: From Preclinical Studies to Clinical Trials. Antioxidants 2020, 9, 692. [Google Scholar] [CrossRef] [PubMed]
  177. Butterfield, D.A.; Boyd-Kimball, D. Redox proteomics and amyloid β-peptide: Insights into Alzheimer disease. J. Neurochem. 2019, 151, 459–487. [Google Scholar] [CrossRef] [Green Version]
  178. Di Carlo, M.; Giacomazza, D.; Picone, P.; Nuzzo, D.; San Biagio, P.L. Are oxidative stress and mitochondrial dysfunction the key players in the neurodegenerative diseases? Free Radic. Res. 2012, 46, 1327–1338. [Google Scholar] [CrossRef]
  179. Lott, I.T. Antioxidants in Down syndrome. Biochim. Biophys. Acta 2012, 1822, 657–663. [Google Scholar] [CrossRef] [Green Version]
  180. Perluigi, M.; Butterfield, D.A. Oxidative Stress and Down Syndrome: A Route toward Alzheimer-Like Dementia. Curr. Gerontol. Geriatr. Res. 2012, 724904. [Google Scholar] [CrossRef] [Green Version]
  181. Wiseman, F.K.; Al-Janabi, T.; Hardy, J.; Karmiloff-Smith, A.; Nizetic, D.; Tybulewicz, V.L.; Fisher, E.M.; Strydom, A. A genetic cause of Alzheimer disease: Mechanistic insights from Down syndrome. Nat. Rev. Neurosci. 2015, 16, 564–574. [Google Scholar] [CrossRef] [Green Version]
  182. De Haan, J.B.; Cristiano, F.; Iannello, R.C.; Kola, I. Cu/Zn-superoxide dismutase and glutathione peroxidase during aging. Biochem. Mol. Biol. Int. 1995, 35, 1281–1297. [Google Scholar] [PubMed]
  183. Ermak, G.; Sojitra, S.; Yin, F.; Cadenas, E.; Cuervo, A.M.; Davies, K.J. Chronic expression of RCAN1-1L protein induces mitochondrial autophagy and metabolic shift from oxidative phosphorylation to glycolysis in neuronal cells. J. Biol. Chem. 2012, 287, 14088–14098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Sun, X.; Wu, Y.; Herculano, B.; Song, W. RCAN1 overexpression exacerbates calcium overloading-induced neuronal apoptosis. PLoS ONE 2014, 9, e95471. [Google Scholar] [CrossRef] [PubMed]
  185. Lee, J.E.; Jang, H.; Cho, E.J.; Youn, H.D. Down syndrome critical region 1 enhances the proteolytic cleavage of calcineurin. Exp. Mol. Med. 2009, 41, 471–477. [Google Scholar] [CrossRef]
  186. Celsi, F.; Svedberg, M.; Unger, C.; Cotman, C.W.; Carrì, M.T.; Ottersen, O.P.; Nordberg, A.; Torp, R. Beta-amyloid causes downregulation of calcineurin in neurons through induction of oxidative stress. Neurobiol. Dis. 2007, 26, 342–352. [Google Scholar] [CrossRef]
  187. Crawford, D.R.; Leahy, K.P.; Abramova, N.; Lan, L.; Wang, Y.; Davies, K.J. Hamster adapt78 mRNA is a Down syndrome critical region homologue that is inducible by oxidative stress. Arch. Biochem. Biophys. 1997, 342, 6–12. [Google Scholar] [CrossRef]
  188. Butterfield, D.A.; Boyd-Kimball, D. Oxidative stress, amyloid-b peptide, and altered key molecular pathways in the pathogenesis and progression of Alzheimer’s disease. J. Alzheimers Dis. 2018, 62, 1345–1367. [Google Scholar] [CrossRef] [Green Version]
  189. Olney, J.W. Excitotoxicity: An overview. Can. Dis. Wkly. Rep. 1990, 16, S47–S57. [Google Scholar]
  190. Butterfield, D.A.; Pocernich, C.B. The glutamatergic system and Alzheimer’s disease: Therapeutic implications. CNS Drugs 2003, 17, 641–652. [Google Scholar] [CrossRef]
  191. Butterfield, D.A.; Lauderback, C.M. Lipid peroxidation and protein oxidation in Alzheimer’s disease brain: Potential causes and consequences involving amyloid b-peptide-associated free radical oxidative stress. Free Radic. Biol. Med. 2002, 32, 1050–1060. [Google Scholar] [CrossRef]
  192. Di Domenico, F.; Barone, E.; Perluigi, M.; Butterfield, D.A. The triangle of death in Alzheimer’s disease brain: The aberrant cross-talk among energy metabolism, mammalian target of rapamycin signaling, and protein homeostasis revealed by redox proteomics. Antioxid. Redox Signal 2017, 26, 364–387. [Google Scholar] [CrossRef] [PubMed]
  193. Di Domenico, F.; Tramutola, A.; Barone, E.; Lanzillotta, C.; Defever, O.; Arena, A.; Zuliani, I.; Foppoli, C.; Iavarone, F.; Vincenzoni, F.; et al. Restoration of aberrant mTOR signaling by intranasal rapamycing reduces oxidative damage: Focus on HNE-modified proteins in a mouse model of Down syndrome. Redox Biol. 2019, 23, 101162. [Google Scholar] [CrossRef] [PubMed]
  194. D i Domenico, F.; Coccia, R.; Cocciolo, A.; Murphy, M.P.; Cenini, G.; Head, E.; Butterfield, D.A.; Giorgi, A.; Schinina, M.E.; Mancuso, C.; et al. Impairment of proteostasis network in Down syndrome prior to the development of Alzheimer’s disease neuropathology: Redox proteomics analysis of human brain. Biochim. Biophys. Acta 2013, 1832, 1249–1259. [Google Scholar] [CrossRef]
  195. Di Domenico, F.; Pupo, G.; Tramutola, A.; Giorgi, A.; Schinina, M.E.; Coccia, R.; Head, E.; Butterfield, D.A.; Perluigi, M. Redox proteomics analysis of HNE-modified proteins in Down syndrome brain: Clues for understanding the development of Alzheimer disease. Free Radic. Biol. Med. 2014, 71, 270–280. [Google Scholar] [CrossRef] [Green Version]
  196. Tramutola, A.; Lanzillotta, C.; Arena, A.; Barone, E.; Perluigi, M.; Di Domenico, F. Increased Mammalian Target of Rapamycin Signaling Contributes to the Accumulation of Protein Oxidative Damage in a Mouse Model of Down’s Syndrome. Neurodegener. Dis. 2016, 16, 62–68. [Google Scholar] [CrossRef] [PubMed]
  197. Perluigi, M.; Di Domenico, F.; Butterfield, D.A. mTOR signaling in aging and neurodegeneration: At the crossroad between metabolism dysfunction and impairment of autophagy. Neurobiol. Dis. 2015, 84, 39–49. [Google Scholar] [CrossRef]
  198. Tramutola, A.; Lanzillotta, C.; Barone, E.; Arena, A.; Zuliani, I.; Mosca, L.; Blarzino, C.; Butterfield, D.A.; Perluigi, M.; Di Domenico, F. Intranasal rapamycin ameliorates Alzheimer-like cognitive decline in a mouse model of Down syndrome. Transl. Neurodegener. 2018, 7, 28. [Google Scholar] [CrossRef] [Green Version]
  199. Godoy, J.A.; Rios, J.A.; Zolezzi, J.M.; Braidy, N.; Inestrosa, N.C. Signaling pathway cross talk in Alzheimer’s disease. Cell Commun. Signal. 2014, 12, 23. [Google Scholar] [CrossRef] [Green Version]
  200. Dröge, W.; Schipper, H. Oxidative stress and aberrant signaling in aging and cognitive decline. Aging Cell 2007, 6, 361–370. [Google Scholar] [CrossRef]
  201. Brand, M.D. The role of mitochondria in longevity and healthspan. Longev. Healthspan 2014, 3, 7. [Google Scholar] [CrossRef] [Green Version]
  202. Hall, C.N.; Klein-Flügge, M.C.; Howarth, C.; Attwell, D. Oxidative phosphorylation, not glycolysis, powers presynaptic and postsynaptic mechanisms underlying brain information processing. J. Neurosci. 2012, 32, 8940–8951. [Google Scholar] [CrossRef] [PubMed]
  203. Swerdlow, R.H.; Khan, S.M. A “mitochondrial cascade hypothesis” for sporadic Alzheimer’s disease. Med. Hypotheses 2004, 63, 8–20. [Google Scholar] [CrossRef] [PubMed]
  204. Wilkins, H.M.; Swerdlow, R.H. Amyloid precursor protein processing and bioenergetics. Brain Res. Bull. 2017, 133, 71–79. [Google Scholar] [CrossRef] [PubMed]
  205. Pesini, A.; Iglesias, E.; Garrido, N.; Bayona-Bafaluy, M.P.; Montoya, J.; Ruiz-Pesini, E. OXPHOS, pyrimidine nucleotides, and Alzheimer’s disease: A pharmacogenomics approach. J. Alzheimers Dis. 2014, 42, 87–96. [Google Scholar] [CrossRef] [PubMed]
  206. Selkoe, D.J. Alzheimer’s disease is a synaptic failure. Science 2002, 298, 789–791. [Google Scholar] [CrossRef] [Green Version]
  207. Krapfenbauer, K.; Yoo, B.C.; Cairns, N.; Lubec, G. Differential display reveals deteriorated mRNA levels of NADH3 (complex I) in cerebellum of patients with Down syndrome. J. Neural. Transm. Suppl. 1999, 57, 211–220. [Google Scholar]
  208. Salemi, M.; Barone, C.; Salluzzo, M.G.; Giambirtone, M.; Scillato, F.; Rando, R.G.; Romano, C.; Morale, M.C.; Ridolfi, F.; Romano, C. A polymorphism (rs1042522) in TP53 gene is a risk factor for Down Syndrome in Sicilian mothers. J. Matern. Fetal Neonatal Med. 2017, 30, 2752–2754. [Google Scholar] [CrossRef]
  209. Conti, A.; Fabbrini, F.; D’Agostino, P.; Negri, R.; Greco, D.; Genesio, R.; D’Armiento, M.; Olla, C.; Paladini, D.; Zannini, M.; et al. Altered expression of mitochondrial and extracellular matrix genes in the heart of human fetuses with chromosome 21 trisomy. BMC Genom. 2007, 8, 268. [Google Scholar] [CrossRef] [Green Version]
  210. Piccoli, C.; Izzo, A.; Scrima, R.; Bonfiglio, F.; Manco, R.; Negri, R.; Quarato, G.; Cela, O.; Ripoli, M.; Prisco, M.; et al. Chronic pro-oxidative state and mitocondrial dysfunctions are more pronounced in fibroblasts from Down syndrome foeti with congenital heart defects. Hum. Mol. Genet. 2013, 22, 1218–1232. [Google Scholar] [CrossRef] [Green Version]
  211. Qiu, J.J.; Liu, Y.N.; Ren, Z.R.; Yan, J.B. Dysfunctions of mitochondria in close association with strong perturbation of long noncoding RNAs expression in Down syndrome. Int. J. Biochem. Cell. Biol. 2017, 92, 115–120. [Google Scholar] [CrossRef]
  212. Kim, S.H.; Vlkolinsky, R.; Cairns, N.; Lubec, G. Decreased levels of complex III core protein 1 and complex V beta chain in brains from patients with Alzheimer’s disease and Down syndrome. Cell. Mol. Life Sci. 2000, 57, 1810–1816. [Google Scholar] [CrossRef] [PubMed]
  213. Kim, S.H.; Dierssen, M.; Ferreres, J.C.; Fountoulakis, M.; Lubec, G. Increased protein levels of heterogeneous nuclear ribonucleoprotein A2/B1 in fetal Down syndrome brains. J. Neural Transm. Suppl. 2001, 57, 273–280. [Google Scholar]
  214. Kim, S.H.; Vlkolinsky, R.; Cairns, N.; Fountoulakis, M.; Lubec, G. The reduction of NADH ubiquinone oxidoreductase 24- and 75-kDa subunits in brains of patients with Down syndrome and Alzheimer’s disease. Life Sci. 2001, 68, 2741–2750. [Google Scholar] [CrossRef]
  215. Valenti, D.; Tullo, A.; Caratozzolo, M.F.; Merafina, R.S.; Scartezzini, P.; Marra, E.; Vacca, R.A. Impairment of F1F0-ATPase, adenine nucleotide translocator and adenylate kinase causes mitochondrial energy deficit in human skin fibroblasts with chromosome 21 trisomy. Biochem. J. 2010, 431, 299–310. [Google Scholar] [CrossRef] [PubMed]
  216. Valenti, D.; Manente, G.A.; Moro, L.; Marra, E.; Vacca, R.A. Deficit of complex I activity in human skin fibroblasts with chromosome 21 trisomy and overproduction of reactive oxygen species by mitochondria: Involvement of the cAMP/PKA signalling pathway. Biochem. J. 2011, 435, 679–688. [Google Scholar] [CrossRef] [Green Version]
  217. Valenti, D.; De Rasmo, D.; Signorile, A.; Rossi, L.; de Bari, L.; Scala, I.; Granese, B.; Papa, S.; Vacca, R.A. Epigallocatechin-3-gallate prevents oxidative phosphorylation deficit and promotes mitochondrial biogenesis in human cells from subjects with Down’s syndrome. Biochim. Biophys. Acta 2013, 1832, 542–552. [Google Scholar] [CrossRef] [Green Version]
  218. Izzo, A.; Nitti, M.; Mollo, N.; Paladino, S.; Procaccini, C.; Faicchia, D.; Cali, G.; Genesio, R.; Bonfiglio, F.; Cicatiello, R.; et al. Metformin restores the mitochondrial network and reverses mitochondrial dysfunction in Down syndrome cells. Hum. Mol. Genet. 2017, 26, 1056–1069. [Google Scholar] [CrossRef] [Green Version]
  219. Panagaki, T.; Randi, E.B.; Augsburger, F.; Szabo, C. Overproduction of H2S, generated by CBS, inhibits mitochondrial Complex IV and suppresses oxidative phosphorylation in Down syndrome. Proc. Natl. Acad. Sci. USA 2019, 116, 18769–18771. [Google Scholar] [CrossRef] [Green Version]
  220. Helguera, P.; Seiglie, J.; Rodriguez, J.; Hanna, M.; Helguera, G.; Busciglio, J. Adaptive downregulation of mitochondrial function in Down syndrome. Cell Metab. 2013, 17, 132–140. [Google Scholar] [CrossRef] [Green Version]
  221. Izzo, A.; Mollo, N.; Nitti, M.; Paladino, S.; Calì, G.; Genesio, R.; Bonfiglio, F.; Cicatiello, R.; Barbato, M.; Sarnataro, V.; et al. Mitochondrial dysfunction in down syndrome: Molecular mechanisms and therapeutic targets. Mol. Med. 2018, 24, 2. [Google Scholar] [CrossRef] [Green Version]
  222. Izzo, A.; Manco, R.; Bonfiglio, F.; Calì, G.; De Cristofaro, T.; Patergnani, S.; Cicatiello, R.; Scrima, R.; Zannini, M.; Pinton, P.; et al. NRIP1/RIP140 siRNA-mediated attenuation counteracts mitochondrial dysfunction in Down syndrome. Hum. Mol. Genet. 2014, 23, 4406–4419. [Google Scholar] [CrossRef] [PubMed]
  223. Schieke, S.M.; Finkel, T. Mitochondrial signaling, TOR, and life span. Biol. Chem. 2006, 387, 1357–1361. [Google Scholar] [CrossRef] [PubMed]
  224. Sarbassov, D.D.; Guertin, D.A.; Ali, S.M.; Sabatini, D.M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005, 307, 1098–1101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Cunnane, S.; Nugent, S.; Roy, M.; Courchesne-Loye, A.; Croteau, E.; Tremblay, S.; Castellano, A.; Pifferi, F.; Bocti, C.; Paquet, N.; et al. Brain fuel metabolism, aging, and Alzheimer’s disease. Nutrition 2011, 27, 3–20. [Google Scholar] [CrossRef] [Green Version]
  226. Anstey, K.J.; Cherbuin, N.; Budge, M.; Young, J. Body mass index in midlife and late-life as a risk factor for dementia: A metaanalysis of prospective studies. Obes. Rev. 2011, 12, e426–e437. [Google Scholar] [CrossRef]
  227. Ronnemaa, E.; Zethelius, B.; Lannfelt, L.; Kilander, L. Vascular risk factors and dementia: 40-year follow-up of a population-based cohort. Dement. Geriatr. Cogn. Disord. 2011, 31, 460–466. [Google Scholar] [CrossRef]
  228. Loef, M.; Walach, H. Midlife obesity and dementia: Metaanalysis and adjusted forecast of dementia prevalence in the United States and China. Obesity 2013, 21, E51–E55. [Google Scholar] [CrossRef]
  229. Gottesman, R.F.; Schneider, A.L.; Zhou, Y.; Coresh, J.; Green, E.; Gupta, N.; Knopman, D.S.; Mintz, A.; Rahmim, A.; Scharrett, A.R.; et al. Association between midlife vascular risk factors and estimated brain amyloid deposition. JAMA 2017, 317, 1443–1450. [Google Scholar] [CrossRef]
  230. Hayden, M.R. Type 2 Diabetes Mellitus Increases The Risk of Late-Onset Alzheimer’s Disease: Ultrastructural Remodeling of the Neurovascular Unit and Diabetic Gliopathy. Brain Sci. 2019, 9, 262. [Google Scholar] [CrossRef] [Green Version]
  231. Chatterjee, S.; Mudher, A. Alzheimer’s disease and type 2 diabetes: A critical assessment of the shared pathological traits. Front. Neurosci. 2018, 12, 383. [Google Scholar] [CrossRef] [Green Version]
  232. Tumminia, A.; Vinciguerra, F.; Parisi, M.; Frittitta, L. Type 2 diabetes mellitus and Alzheimer’s disease: Role of insulin signalling and therapeutic implications. Int. J. Mol. Sci. 2018, 19, 3306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Tang, B.L. Glucose, glycolysis, and neurodegenerative diseases. J. Cell. Physiol. 2020, 1–10. [Google Scholar] [CrossRef] [PubMed]
  234. Soloman, A.; Kivipelto, M.; Wolozin, B.; Zhou, J.; Whitmer, R.A. Midlife serum cholesterol and increased risk of Alzheimer’s and vascular dementia three decades later. Dement. Geriatr. Cogn. Disord. 2009, 28, 75–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Meng, X.F.; Yu, J.T.; Wang, H.F.; Tan, M.S.; Wang, C.; Tan, C.C.; Tan, L. Midlife vascular risk factors and the risk of Alzheimer’s disease: A systematic review and meta-analysis. J. Alzheimers Dis. 2014, 42, 1295–1310. [Google Scholar] [CrossRef]
  236. Lee, H.J.; Seo, H.I.; Cha, H.Y.; Yang, Y.J.; Kwon, S.H.; Yang., S.J. Diabetes and Alzheimer’s disease: Mechanisms and nutritional aspects. Clin. Nutr. Res. 2018, 7, 229–240. [Google Scholar] [CrossRef] [Green Version]
  237. Li, W.; Huang, E.; Gao, S. Type 1 diabetes mellitus and cognitive impairments: A systematic review. J. Alzheimers Dis. 2017, 57, 29–36. [Google Scholar] [CrossRef] [Green Version]
  238. Desai, G.S.; Zheng, C.; Geetha, T.; Mathews, S.T.; White, B.D.; Huggins, K.W.; Zizza, C.A.; Broderick, T.L.; Babu, J.R. The pancreas-brain axis: Insight into disrupted mechanisms associating type 2 diabetes and Alzheimer’s disease. J. Alzheimers Dis. 2014, 42, 347–356. [Google Scholar] [CrossRef]
  239. Arnold, S.E.; Arvanitakis, Z.; Macauley-Rambach, S.L.; Koenig, A.M.; Wang, H.-Y.; Ahima, R.S.; Craft, S.; Gandy, S.; Buettner, C.; Stoeckel, L.E.; et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: Concepts and conundrums. Nat. Rev. Neurol. 2018, 14, 168–181. [Google Scholar] [CrossRef]
  240. Tramutola, A.; Lanzillotta, C.; Di Domenico, F.; Head, E.; Butterfield, D.A.; Perluigi, M.; Barone, E. Brain insulin resistance triggers early onset Alzheimer disease in Dowon syndrome. Neurobiol. Dis. 2020, 137, 104772. [Google Scholar] [CrossRef]
  241. Biessels, G.J.; Reagan, L.P. Hippocampal insulin resistance and cognitive dysfunction. Nat. Rev. Neurosci. 2015, 16, 660–671. [Google Scholar] [CrossRef]
  242. De Felice, F.G. Alzheimer’s disease and insulin resistance: Translating basic science into clinical applications. J. Clin. Invest. 2013, 123, 531–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Tramutola, A.; Triplett, J.C.; Di Domenico, F.; Niedowicz, D.M.; Murphy, M.P.; Coccia, R.; Perluigi, M.; Butterfield, D.A. Alteration of mTOR signaling occurs early in the progression of Alzheimer disease (AD): Analysis of brain from subjects with pre-clinical AD, amnestic mild cognitive impairment and late-stage AD. J. Neurochem. 2015, 133, 739–749. [Google Scholar] [CrossRef] [PubMed]
  244. Blázquez, E.; Velázquez, E.; Hurtado-Carneiro, V.; Ruiz-Albusac, J.M. Insulin in the brain: Its pathophysiological implications for States related with central insulin resistance, type 2 diabetes and Alzheimer’s disease. Front. Endocrinol. 2014, 5, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Simpson, I.A.; Chundu, K.R.; Davies-Hill, T.; Honer, W.G.; Davies, P. Decreased concentrations of GLUT1 and GLUT3 glucose transporters in the brains of patients with Alzheimer’s disease. Ann. Neurol. 1994, 35, 546–551. [Google Scholar] [CrossRef] [PubMed]
  246. Bergau, N.; Maul, S.; Rujescu, D.; Simm, A. Reduction of glycolysis intermediate concentrations in the cerebrospinal fluid of Alzheimer’s disease patients. Front. Neurosci. 2019, 13, 871. [Google Scholar] [CrossRef] [PubMed]
  247. Croteau, E.; Castellano, C.A.; Fortier, M.; Bocti, C.; Fulop, T.; Paquet, N.; Cunnane, S.C. A cross-sectional comparison of brain glucose and ketone metabolism in cognitively healthy older adults, mild cognitive impairment and early Alzheimer’s disease. Exp. Gerontol. 2018, 107, 18–26. [Google Scholar] [CrossRef]
  248. Theurey, P.; Connolly, N.M.C.; Fortunati, I.; Basso, E.; Lauwen, S.; Ferrante, C.; Pinho, C.M.; Joselin, A.; Gioran, A.; Bano, D.; et al. Systems biology identifies preserved integrity but impaired metabolism of mitochondria due to a glycolytic defect in Alzheimer’s disease neurons. Aging Cell 2019, 18, e12924. [Google Scholar] [CrossRef] [Green Version]
  249. Yamaguchi, S.; Meguro, K.; Itoh, M.; Hayasaka, C.; Shimada, M.; Yamazaki, H.; Yamadori, A. Decreased cortical glucose metabolism correlates with hippocampal atrophy in Alzheimer’s disease as shown by MRI and PET. J. Neurol. Neurosurg. Psychiatry 1997, 62, 596–600. [Google Scholar] [CrossRef] [Green Version]
  250. Kato, T.; Inui, Y.; Nakamura, A.; Ito, K. Brain fluordeoxyglucose (FDG) PET in dementia. Ageing Res. Rev. 2016, 30, 73–84. [Google Scholar] [CrossRef]
  251. Hertz, L.; Chen, Y. Integration between glycolysis and glutamate-glutamine cycle flux may explain preferential glycolytic increase during brain activation, requiring glutamate. Front. Integr Neurosci. 2017, 11, 18. [Google Scholar] [CrossRef] [Green Version]
  252. Mergenthaler, P.; Lindauer, U.; Dienel, G.A.; Meisel, A. Sugar for the brain: The role of glucose in physiological and pathological brain function. Trends Neurosci. 2013, 36, 587–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Ueno, M.; Carvalheira, J.B.; Tambascia, R.C.; Bezerra, R.M.; Amaral, M.E.; Carneiro, E.M.; Folli, F.; Franchini, K.G.; Saad, M.J. Regulation of insulin signalling by hyperinsulinaemia: Role of IRS-1/2 serine phosphorylation and the mTOR/p70 S6K pathway. Diabetologia 2005, 48, 506–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Alayev, A.; Holz, M.K. mTOR signaling for biological control and cancer. J. Cell Physiol. 2013, 228, 1658–1664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell. Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Spilman, P.; Podlutskaya, N.; Hart, M.J.; Debnath, J.; Gorostiza, O.; Bredesen, D.; Richardson, A.; Strong, R.; Galvan, V. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer’s disease. PLoS ONE 2010, 5, e9979. [Google Scholar] [CrossRef] [Green Version]
  257. Vicencio, J.M.; Galluzzi, L.; Tajeddine, N.; Ortiz, C.; Criollo, A.; Tasdemir, E.; Morselli, E.; Younes, A.B.; Maiuri, M.C.; Lavendero, S.; et al. Senescence, apoptosis or autophagy? When a damaged cell must decide its path-a mini-review. Gerontology 2008, 54, 92–99. [Google Scholar] [CrossRef]
  258. Faragher, R.G.A.; McArdle, A.; Willows, A.; Ostler, E.L. Senescence in the aging process. F1000Res 2017, 6, 1219. [Google Scholar] [CrossRef] [Green Version]
  259. Stein, G.H.; Drullinger, L.F.; Soulard, A.; Dulić, V. Differential roles for cyclin-dependent kinase inhibitors p21 and p16 in the mechanisms of senescence and differentiation in human fibroblasts. Mol. Cell Biol. 1999, 19, 2109–2117. [Google Scholar] [CrossRef] [Green Version]
  260. Krenning, L.; Feringa, F.M.; Shaltiel, I.A.; Van den Berg, J.; Medema, R.H. Transient activation of p53 in G2 phase is sufficient to induce senescence. Mol. Cell 2014, 55, 59–72. [Google Scholar] [CrossRef] [Green Version]
  261. Özcan, S.; Alessio, N.; Acar, M.B.; Mert, E.; Omerli, F.; Peluso, G.; Galderisi, U. Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging 2016, 8, 1316–1329. [Google Scholar] [CrossRef] [Green Version]
  262. Correia-Melo, C.; Passos, J.F. Mitochondria: Are they causal players in cellular senescence? Biochim. Biophys. Acta 2015, 1847, 1373–1379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Weichhart, T. mTOR as regulator of lifespan, aging, and cellular senescence: A mini-review. Gerontology 2018, 64, 127–134. [Google Scholar] [CrossRef] [PubMed]
  264. Nakamura, A.J.; Chiang, Y.J.; Hathcock, K.S.; Horikawa, I.; Sedelnikova, O.A.; Hodes, R.J.; Bonner, W.M. Both telomeric and non-telomeric DNA damage are determinants of mammalian cellular senescence. Epigenetics Chromatin 2008, 1, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Freund, A.; Laberge, R.-M.; Demaria, M.; Campisi, J. Lamin B1 loss is a senescence-associated biomarker. Mol. Biol. Cell. 2012, 23, 2066–2075. [Google Scholar] [CrossRef] [PubMed]
  266. Cormenier, J.; Martin, N.; Deslé, J.; Salazar-Cardozo, C.; Pourtier, A.; Abbadie, C.; Pluquet, O. The ATF6 arm of the Unfolded Protein Response mediates replicative senescence in human fibroblasts through a COX2/prostaglandin E2 intracrine pathway. Mech. Ageing Dev. 2018, 170, 82–91. [Google Scholar] [CrossRef]
  267. Zhang, P.; Kishimoto, Y.; Grammatikakis, I.; Gottimukkala, K.; Cutler, R.G.; Zhang, S.; Abdelmohsen, K.; Bohr, V.A.; Sen, J.M.; Gorospe, M.; et al. Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat. Neurosci. 2019, 22, 719–728. [Google Scholar] [CrossRef]
  268. Musi, N.; Valentine, J.M.; Sickora, K.R.; Baeuerle, E.; Thompson, C.S.; Shen, Q.; Orr, M.E. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell. 2018, 17, e12840. [Google Scholar] [CrossRef]
  269. Roberson, R.; Kuddo, T.; Horowitz, K.; Caballero, M.; Spong, C.Y. Cytokine and chemokine alterations in Down syndrome. Am. J. Perinatol. 2012, 29, 705–708. [Google Scholar] [CrossRef]
  270. Rueda, N.; Vidal, V.; García-Cerro, S.; Narcís, J.O.; Llorens-Martín, M.; Corrales, A.; Lantigua, S.; Iglesias, M.; Merino, J.; Merino, R.; et al. Anti-IL17 treatment ameliorates Down syndrome phenotypes in mice. Brain Behav. Immun. 2018, 73, 235–251. [Google Scholar] [CrossRef]
  271. Yang, S.; Wang, J.; Guo, S.; Huang, D.; Bestard Lorigados, I.; Nie, X.; Lou, D.; Li, Y.; Liu, M.; Kang, Y.; et al. Transcriptional activation of UPS16 gene expression by NFkB signaling. Mol. Brain 2019, 12, 120. [Google Scholar] [CrossRef] [Green Version]
  272. Adorno, M.; Sikandar, S.; Mitra, S.S.; Kuo, A.; Di Robilant, B.N.; Haro-Acosta, V.; Ouadah, Y.; Quarta, M.; Rodriguez, J.; Qian, D.; et al. Usp16 contributes to somatic stem-cell defects in Down’s syndrome. Nature 2013, 501, 380–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Adorno, M.; di Robilant, B.N.; Sikandar, S.S.; Haro-Acosta, V.; Antony, J.; Heller, C.H.; Clarke, M.F. Usp16 modulates Wnt signaling in primary tissues through Cdkn2a regulation. Sci. Rep. 2018, 8, 17506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Wang, Y.; Chang, J.; Shao, L.; Feng, W.; Luo, Y.; Chow, M.; Du, W.; Meng, A.; Zhou, D. Hematopoietic Stem Cells from Ts65Dn Mice Are Deficient in the Repair of DNA Double-Strand Breaks. Radiat Res. 2016, 185, 630–637. [Google Scholar] [CrossRef] [PubMed]
  275. Olivieri, F.; Prattichizzo, F.; Grillari, J.; Balistreri, C.R. Cellular senescence and inflammaging in age-related diseases. Med. Inflamm. 2018, 9076485. [Google Scholar] [CrossRef] [PubMed]
  276. Fuster-Matanzo, A.; Llorens-Martín, M.; Hernández, F.; Avila, J. Role of neuroinflammation in adult neurogenesis and Alzheimer disease: Therapeutic approaches. Mediat. Inflamm. 2013, 260925. [Google Scholar] [CrossRef] [Green Version]
  277. Lyman, M.; Lloyd, D.G.; Ji, X.; Vizcaychipi, M.P.; Ma, D. Neuroinflammation: The role and consequences. Neurosci. Res. 2014, 79, 1–12. [Google Scholar] [CrossRef]
  278. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
  279. Guerreiro, R.; Brás, J.; Hardy, J. Snapshot: Genetics of Alzheimer’s disease. Cell 2013, 155, 968. [Google Scholar] [CrossRef] [Green Version]
  280. Rawji, K.S.; Mishra, M.K.; Michaels, N.J.; Rivest, S.; Stys, P.K.; Yong, V.W. Immunosenescence of microglia and macrophages: Impact on the ageing central nervous system. Brain 2016, 139, 653–661. [Google Scholar] [CrossRef] [Green Version]
  281. Bauer, J.; Strauss, S.; Schreiter-Gasser, U.; Ganter, U.; Schlegel, P.; Witt, I.; Yolk, B.; Berger, M. Interleukin-6 and α-2-macroglobulin indicate an acute-phase state in Alzheimer’s disease cortices. FEBS Lett. 1991, 285, 111–114. [Google Scholar] [CrossRef] [Green Version]
  282. Huell, M.; Strauss, S.; Volk, B.; Berger, M.; Bauer, J. Interleukin-6 is present in early stages of plaque formation and is restricted to the brains of Alzheimer’s disease patients. Acta Neuropathol. 1995, 89, 544–551. [Google Scholar] [CrossRef] [PubMed]
  283. Sun, A.; Liu, M.; Nguyen, X.V.; Bing, G. P38 MAP kinase is activated at early stages in Alzheimer’s disease brain. Exp. Neurol. 2003, 183, 394–405. [Google Scholar] [CrossRef]
  284. Lai, K.S.P.; Liu, C.S.; Rau, A.; Lanctôt, K.L.; Köhler, C.A.; Pakosh, M.; Carvalho, A.F.; Herrmann, N. Peripheral inflammatory markers in Alzheimer’s disease: A systematic review and meta-analysis of 175 studies. J. Neurol. Neurosurg. Psychiatry 2017, 88, 876–882. [Google Scholar] [CrossRef] [PubMed]
  285. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and age-related diseases: Role of inflammation triggers and cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef] [PubMed]
  286. Blurton-Jones, M.; Laferla, F.M. Pathways by which Aβ facilitates tau pathology. Curr. Alzheimer Res. 2006, 3, 437–448. [Google Scholar] [CrossRef] [PubMed]
  287. Minter, M.R.; Taylor, J.M.; Crack, P.J. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. J. Neurochem. 2016, 136, 457–474. [Google Scholar] [CrossRef] [PubMed]
  288. Barger, S.W.; Harmon, A.D. Microglial activation by alzhelmer amyloid precursor protein and modulation by apolipoprotein E. Nature 1997, 388, 878–881. [Google Scholar] [CrossRef]
  289. Ho, G.J.; Drego, R.; Hakimian, E.; Masliah, E. Mechanisms of cell signaling and inflammation in Alzheimer’s disease. Curr. Drug Targets Inflamm. Allergy 2004, 4, 247–256. [Google Scholar] [CrossRef]
  290. Sastre, M.; Dewachter, I.; Landreth, G.E.; Willson, T.M.; Klockgether, T.; van Leuven, F.; Heneka, M.T. Nonsteroidal anti-inflammatory drugs and peroxisome proliferator-activated receptor-γ agonists modulate immunostimulated processing of amyloid precursor protein through regulation of β-secretase. J. Neurosci. 2003, 23, 9796–97804. [Google Scholar] [CrossRef] [Green Version]
  291. Sastre, M.; Dewachter, I.; Rossner, S.; Bogdanovic, N.; Rosen, E.; Borghgraef, P.; Evert, B.O.; Dumitrescu-Ozimek, L.; Thal, D.R.; Landreth, G.; et al. Nonsteroidal anti-inflammatory drugs repress β- secretase gene promoter activity by the activation of PPARgamma. Proc. Natl. Acad. Sci. USA 2006, 103, 443–448. [Google Scholar] [CrossRef] [Green Version]
  292. Mrak, R.E.; Griffin, W.S. Potential inflammatory biomarkers in Alzheimer’s disease. J. Alzheimers Dis. 2005, 8, 369–375. [Google Scholar] [CrossRef] [PubMed]
  293. Akiyama, H.; Barger, S.; Barnum, S.; Bradt, B.; Bauer, J.; Cole, G.M.; Cooper, N.R.; Eikelenboom, P.; Emmerling, M.; Fiebich, B.L.; et al. Inflammation and Alzheimer’s disease. Neurobiol. Aging 2000, 21, 383–421. [Google Scholar] [CrossRef]
  294. Sapolsky, R.M. Glucocorticoid toxicity in the hippocampus: Reversal by supplementation with brain fuels. J. Neurosci. 1986, 6, 2240–2244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Qi, D.; Rodrigues, B. Glucocorticoids produce whole body insulin resistance with changes in cardiac metabolism. Am. J. Physiol. Endocrinol. Metab. 2007, 292, 654–667. [Google Scholar] [CrossRef] [Green Version]
  296. Lim, G.P.; Yang, F.; Chu, T.; Gahtan, E.; Ubeda, O.; Beech, W.; Overmier, J.B.; Hsiao Ashe, K.; Frautschy, S.A.; Cole, G.M. Ibuprofen effects on Alzheimer pathology and open field activity in APPsw transgenic mice. Neurobiol. Aging 2001, 22, 983–991. [Google Scholar] [CrossRef]
  297. Vogel, A.; Upadhya, R.; Shetty, A.K. Neural stem cell derived extracellular vesicles: Attributes and prospects for treating neurodegenerative disorders. EBioMedicine 2018, 38, 273–282. [Google Scholar] [CrossRef]
  298. Wheeler, D.L.; Sariol, A.; Meyerholz, D.K.; Perlman, S. Microglia are required for protection against lethal coronavirus encephalitis in mice. J. Clin. Invest. 2018, 128, 931–943. [Google Scholar] [CrossRef]
  299. Halle, A.; Hornung, V.; Petzold, G.C.; Stewart, C.R.; Monks, B.G.; Reinheckel, T.; Gitzgerald, K.A.; Latz, E.; Moore, K.J.; Golenbock, D.T. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat. Immunol. 2008, 9, 857–865. [Google Scholar] [CrossRef] [Green Version]
  300. Johansson, J.U.; Woodling, N.S.; Wang, Q.; Panchal, M.; Liang, X.; Trueba-Saiz, A.; Brown, H.D.; Mhatre, S.D.; Loui, T.; Andreasson, K.I. Prostaglandin signaling suppresses beneficial microglial function in Alzheimer’s disease models. J. Clin. Invest. 2015, 125, 350–364. [Google Scholar] [CrossRef] [Green Version]
  301. Liu, Y.; Walter, S.; Stagi, M.; Cherny, D.; Letiembre, M.; Schulz-Schaeffer, W.; Heine, H.; Penke, B.; Neumann, H.; Fassbender, K. LPS receptor (CD14): A receptor for phagocytosis of Alzheimer’s amyloid peptide. Brain 2005, 128, 1778–1789. [Google Scholar] [CrossRef] [Green Version]
  302. Martin, E.; Boucher, C.; Fontaine, B.; Delarasse, C. Distinct inflammatory phenotypes of microglia and monocyte-derived macrophages in Alzheimer’s disease models: Effects of aging and amyloid pathology. Aging Cell 2017, 16, 27–38. [Google Scholar] [CrossRef] [PubMed]
  303. Stancu, I.C.; Cremers, N.; Vanrusselt, H.; Couturier, J.; Vanoosthuyse, A.; Kessels, S.; Lodder, C.; Brône, B.; Huaux, F.; Octave, J.-N.; et al. Aggregated Tau activates NLRP3-ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol. 2019, 137, 599–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Yang, Y.; Zhang, Z. Microglia and Wnt pathways: Prospects for inflammation in Alzheimer’s disease. Front. Aging Neurosci. 2020, 12, 120. [Google Scholar] [CrossRef]
  305. Contestabile, A.; Fila, T.; Ceccarelli, A.; Bonasoni, P.; Bonapace, L.; Santini, D.; Bartesaghi, R.; Ciani, E. Cell cycle alteration and decreased cell proliferation in the hippocampal dentate gyrus and in the neocortical germinal matrix of fetuses with Down syndrome and in Ts65Dn mice. Hippocampus 2007, 17, 665–678. [Google Scholar] [CrossRef] [PubMed]
  306. Stagni, F.; Giacomini, A.; Emili, M.; Guidi, S.; Bartesaghi, R. Neurogenesis impairment: An early developmental defect in Down syndrome. Free Radic. Biol. Med. 2018, 114, 15–32. [Google Scholar] [CrossRef] [PubMed]
  307. Hammerle, B.; Ulin, E.; Guimera, J.; Becker, W.; Guillemot, F.; Tejedor, F.J. Transient expression ofMnb/Dyrk1a couples cell cycle exit and differentiation of neuronal precursors by inducing p27KIP1 expression and suppressing NOTCH signaling. Development 2011, 138, 2543–2554. [Google Scholar] [CrossRef] [Green Version]
  308. Sadasiva, S.; DeCaprio, J.A. The DREAM complex: Master coordinator of cell cycle-dependent gene expression. Nat. Rev. Cancer 2013, 13, 585–595. [Google Scholar] [CrossRef] [Green Version]
  309. Litovchick, L.; Florens, L.A.; Swanson, S.K.; Washburn, M.P.; DeCaprio, J.A. DYRK1A protein kinase promotes quiescence and senescence through DREAM complex assembly. Genes Dev. 2011, 25, 801–813. [Google Scholar] [CrossRef] [Green Version]
  310. Yabut, O.; Domogauer, J.; D’Arcangelo, G. Dyrk1A overexpression inhibits proliferation and induces premature neuronal differentiation of neural progenitor cells. J. Neurosci. 2010, 30, 4004–4014. [Google Scholar] [CrossRef]
  311. Chen, J.-Y.; Lin, J.-R.; Feng-Chiao, T.; Meyer, T. Dosage of Dyrk1A shifts cells within a p21-cyclin D1 signalling map to control the decision to enter the cell cycle. Mol. Cell 2013, 52, 87–100. [Google Scholar] [CrossRef] [Green Version]
  312. Kageyama, R.; Ohtsuka, T.; Shimojo, H.; Imayoshi, I. Dynamic regulation of Notch signaling in neural progenitor cells. Curr. Opin. Cell. Biol. 2009, 21, 733–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  313. Nagarsheth, M.H.; Viehman, A.; Lippa, S.M.; Lippa, C.F. Notch-1 immunoexpression is increased in Alzheimer’s and Pick’s disease. J. Neurol. Sci. 2006, 244, 111–116. [Google Scholar] [CrossRef] [PubMed]
  314. Fischer, D.F.; van Dijk, R.; Sluijs, J.A.; Nair, S.M.; Racchi, M.; Levelt, C.N.; van Leeuwen, F.W.; Ho, E.M. Activation of the Notch pathway in Down syndrome: Cross-talk of Notch and APP. FASEB J. 2005, 19, 1451–1458. [Google Scholar] [PubMed]
  315. Wang, S.; Barres, B.A. Up a notch: Instructing gliogenesis minireview. Neuron 2000, 27, 197–200. [Google Scholar] [CrossRef] [Green Version]
  316. Kurabayashi, N.; Sanada, K. Increased dosage of DYRK1A and DSCR1 delays neuronal differentiation in neocortical progenitor cells. Genes Dev. 2013, 27, 2708–2721. [Google Scholar] [CrossRef] [Green Version]
  317. Abelaira, H.M.; Reus, G.Z.; Neotti, M.V.; Quevedo, J. The role of mTOR in depression and antidepressant responses. Life Sci. 2014, 101, 10–14. [Google Scholar] [CrossRef]
  318. Kassai, H.; Sugaya, Y.; Noda, S.; Nakao, K.; Maeda, T.; Kano, M.; Aiba, A. Selective activation of mTORC1 signaling recapitulates microcephaly, tuberous sclerosis, and neurodegenerative diseases. Cell Rep. 2014, 7, 1626–1639. [Google Scholar] [CrossRef] [Green Version]
  319. Chao, M.V. Neurotrophins and their receptors: A convergence point for many signalling pathways. Nat. Rev. Neurosci. 2003, 4, 299. [Google Scholar] [CrossRef]
  320. Zweifel, L.S.; Kuruvilla, R.; Ginty, D.D. Functions and mechanisms of retrograde neurotrophin signaling. Nat. Rev. Neurosci. 2005, 6, 615–625. [Google Scholar] [CrossRef]
  321. Chao, M.V.; Rajagopal, R.; Lee, F.S. Neurotrophin signalling in health and disease. Clin. Sci. 2006, 110, 167–173. [Google Scholar] [CrossRef]
  322. Tapia-Arancibia, L.; Aliaga, E.; Silhol, M.; Arancibia, S. New insights into brain BDNF function in normal aging and Alzheimer disease. Brain Res. Rev. 2008, 59, 201–220. [Google Scholar] [CrossRef] [PubMed]
  323. Roper, R.J.; Baxter, L.L.; Saran, N.G.; Klinedinst, D.K.; Beachy, P.A.; Reeves, R.H. Defective cerebellar response to mitogenic Hedgehog signaling in Down syndrome mice. Proc. Natl. Acad. Sci. USA 2006, 103, 1452–1456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  324. Trazzi, S.; Mitrugno, V.M.; Valli, E.; Fuchs, C.; Rizzi, S.; Guidi, S.; Perini, G.; Bartesaghi, R.; Ciani, E. APP-dependent up-regulation of Ptch1 underlies proliferation impairment of neural precursors in Down syndrome. Hum. Mol. Genet. 2011, 20, 1560–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Trazzi, S.; Fuchs, C.; Valli, E.; Perini, G.; Bartesaghi, R.; Ciani, E. The amyloid precursor protein (APP) triplicated gene impairs neuronal precursor differentiation and neurite development through two different domains in the Ts65Dn mouse model for Down syndrome. J. Biol. Chem. 2013, 288, 20817–20829. [Google Scholar] [CrossRef] [Green Version]
  326. Nalivaeva, N.N.; Turner, A.J. The amyloid precursor protein: A biochemical enigma in brain development, function and disease. FEBS Lett. 2013, 587, 2046–2054. [Google Scholar] [CrossRef] [Green Version]
  327. Giacomini, A.; Stagni, F.; Trazzi, S.; Guidi, S.; Emili, M.; Brigham, E.; Ciani, E.; Bartesaghi, R. Inhibition of APP gamma-secretase restores sonic hedgehog signaling and neurogenesis in the Ts65Dn mouse model of Down syndrome. Neurobiol. Dis. 2015, 82, 385–396. [Google Scholar] [CrossRef] [Green Version]
  328. London, J.; Rouch, C.; Bui, L.C.; Assayag, E.; Souchet, B.; Daubigne, F.; Medjaoui, H.; Luquet, S.; Magnan, C.; Delabar, J.M.; et al. Overexpression of the DYRK1A Gene (Dual-Specificity Tyrosine Phosphorylation- Regulated Kinase 1A) Induces Alterations of the Serotoninergic and Dopaminergic Processing in Murine Brain Tissues. Mol. Neurobiol. 2018, 55, 3822–3831. [Google Scholar] [CrossRef]
  329. Yuen, E.Y.; Qin, L.; Wei, J.; Liu, W.; Liu, A.; Yan, Z. Synergistic regulation of glutamatergic transmission by serotonin and norepinephrine reuptake inhibitors in prefrontal cortical neurons. J. Biol. Chem. 2014, 289, 25177–25185. [Google Scholar] [CrossRef] [Green Version]
  330. Xing, B.; Li, Y.C.; Gao, W.J. Norepinephrine versus dopamine and their interaction in modulating synaptic function in the prefrontal cortex. Brain Res. 2016, 1641, 217–233. [Google Scholar] [CrossRef] [Green Version]
  331. Keating, D.J.; Dubach, D.; Zanin, M.P.; Yu, Y.; Martin, K.; Zhao, Y.-F.; Chen, C.; Porta, S.; Arbonés, M.L.; Mittaz, L.; et al. DSCR1/RCAN1 regulates vesicle exocytosis and fusion pore kinetics: Implications for Down syndrome and Alzheimer’s disease. Hum. Mol. Genet. 2008, 1, 1020–1030. [Google Scholar] [CrossRef] [Green Version]
  332. Zanin, M.P.; Mackenzie, K.D.; Peiris, H.; Pritchard, M.A.; Keating, D.J. RCAN1 regulates vesicle recycling and quantal release kinetics via effects on calcineurin activity. J. Neurochem. 2013, 124, 290–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  333. Peiris, H.; Keating, D. The neural and endocrine roles of RCAN1 in health and disease. Clin. Exp. Pharm. Physiol 2018, 45, 377–383. [Google Scholar] [CrossRef] [PubMed]
  334. Arbonés, M.L.; Thomazeau, A.; Nakano-Kobayashi, A.; Hagiwara, M.; Delabar, J.M. DYRK1A and cognition: A lifelong relationship. Pharmacol. Ther. 2019, 194, 199–221. [Google Scholar] [CrossRef] [PubMed]
  335. Aranda, S.; Alvarez, M.; Turro, S.; Laguna, A.; de la Luna, S. Sprouty2-mediated inhibition of fibroblast growth factor signaling is modulated by the protein kinase DYRK1A. Mol. Cell. Biol. 2008, 28, 5899–5911. [Google Scholar] [CrossRef] [Green Version]
  336. Park, J.; Sung, J.Y.; Park, J.; Song, W.J.; Chang, S.; Chung, K.C. Dyrk1A negatively regulates the actin cytoskeleton through threonine phosphorylation of N-WASP. J. Cell Sci. 2012, 125, 67–80. [Google Scholar] [CrossRef] [Green Version]
  337. Lepagnol-Bestel, A.M.; Zvara, A.; Maussion, G.; Quignon, F.; Ngimbous, B.; Ramoz, N.; Imbeaud, S.; Loe-Mie, Y.; Benihoud, K.; Agier, N.; et al. DYRK1A interacts with the REST/NRSF-SWI/SNF chromatin remodelling complex to deregulate gene clusters involved in the neuronal phenotypic traits of Down syndrome. Hum. Mol. Genet. 2009, 18, 1405–1414. [Google Scholar] [CrossRef] [Green Version]
  338. Martinez de Lagran, M.; Benavides-Piccione, R.; Ballesteros-Yanez, I.; Calvo, M.; Morales, M.; Fillat, C.; Defelipe, J.; Ramakers, G.J.A.; Dierssen, M. Dyrk1A influences neuronal morphogenesis through regulation of cytoskeletal dynamics in mammalian cortical neurons. Cereb. Cortex 2012, 22, 2867–2877. [Google Scholar] [CrossRef] [Green Version]
  339. Kim, Y.; Park, J.; Song, W.J.; Chang, S. Overexpression of Dyrk1A causes the defects in synaptic vesicle endocytosis. Neurosignals 2010, 18, 164–172. [Google Scholar] [CrossRef]
  340. Kwon, H.B.; Kozorovitskiy, Y.; Oh, W.J.; Peixoto, R.T.; Akhtar, N.; Saulnier, J.L.; Gu, C.; Sabatini, B.L. Neuroligin-1-dependent competition regulates cortical synaptogenesis and synapse number. Nat. Neurosci. 2012, 15, 1667–1674. [Google Scholar] [CrossRef] [Green Version]
  341. Toiber, D.; Azkona, G.; Ben-Ari, S.; Toran, N.; Soreq, H.; Dierssen, M. Engineering DYRK1A overdosage yields Down syndrome-characteristic cortical splicing aberrations. Neurobiol. Dis. 2010, 40, 348–359. [Google Scholar] [CrossRef]
  342. Fan, F.; Funk, L.; Lou, X. Dynamin 1- and 3-Mediated Endocytosis Is Essential for the Development of a Large Central Synapse In Vivo. J. Neurosci. 2016, 36, 6097–6115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Garcia-Cerro, S.; Martinez, P.; Vidal, V.; Corrales, A.; Florez, J.; Vidal, R.; Rueda, N.; Arbonés, M.L.; Martínez-Cué, C. Overexpression of Dyrk1A is implicated in several cognitive, electrophysiological and neuromorphological alterations found in a mouse model of Down syndrome. PLoS ONE 2014, 9, e106572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Wang, W.; Rai, A.; Hur, E.M.; Smilansky, Z.; Chang, K.T.; Min, K.T. DSCR1 is required for both axonal growth cone extension and steering. J. Cell Biol. 2016, 213, 451–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Ma, H.; Xiong, H.; Liu, T.; Zhang, L.; Godzik, A.; Zhuohua, Z. Aggregate formation and synaptic abnormality induced by DSCR1. J. Neurochem. 2004, 88, 1485–1496. [Google Scholar] [CrossRef] [PubMed]
  346. Martin, K.R.; Corlett, A.; Dubach, D.; Mustafa, T.; Coleman, H.A.; Parkington, H.C.; Mersen, T.D.; Bourne, J.A.; Porta, S.; Arbonés, M.L.; et al. Over-expression of RCAN1 causes Down syndrome-like hippocampal deficits that alter learning and memory. Hum. Mol. Genet. 2012, 21, 3025–3041. [Google Scholar] [CrossRef]
  347. Sugiura, R.; Sio, S.O.; Shuntoh, H.; Kuno, T. Molecular genetic analysis of the calcineurin signaling pathways. Cell. Mol. Life Sci. 2001, 58, 278–288. [Google Scholar] [CrossRef]
  348. Marks, B.; McMahon, H.T. Calcium triggers calcineurin-dependent synaptic vesicle recycling in mammalian nerve terminals. Curr. Biol. 1998, 8, 740–749. [Google Scholar] [CrossRef] [Green Version]
  349. Cousin, M.A.; Tan, T.C.; Robinson, P.J. Protein phosphorylation is required for endocytosis in nerve terminals: Potential role for the dephosphins dynamin I and synaptojanin, but not AP180 or amphiphysin. J. Neurochem. 2001, 76, 105–116. [Google Scholar] [CrossRef] [Green Version]
  350. Li, S.; Sukeena, J.M.; Simmons, A.B.; Hansen, E.J.; Nuhn, R.E.; Samuels, I.S.; Fuerst., P.G. DSCAM promotes refinement in the mouse retina through cell death and restriction of exploring dendrites. J. Neurosci. 2015, 35, 5640–5654. [Google Scholar] [CrossRef]
  351. Alves-Sampaio, A.; Troca-Marín, J.A.; Montesinos, M.L. NMDA mediated regulation of DSCAM dendritic local translation is lost in a mouse model of Down’s syndrome. J. Neurosci. 2009, 30, 13537–13548. [Google Scholar] [CrossRef]
  352. Sache, S.M.; Lievens, S.; Ribeiro, L.; Dascenco, D.; Massachaele, D.; Horré, K.; Misbaer, A.; Vanderroost, N.; De Smet, A.S.; Salta, E.; et al. Nuclear import of the DSCAM-cytoplasmatic domain drives signaling capable of inhibiting synapse formation. EMBO J. 2019, 38, e99669. [Google Scholar]
  353. O’Bryan, J.P. Intersecting pathways in cell biology. Sci. Signal. 2010, 3, re10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  354. Guipponi, M.; Scott, H.S.; Hattori, M.; Ishii, K.; Sakaki, Y.; Antonarakis, S.E. Genomic structure, sequence, and refined mapping of the human intersectin gene (itsn), which encompasses 250 kb on chromosome 21q22.1→q22.2. Cytogenet. Cell. Genet. 1998, 83, 218–220. [Google Scholar] [CrossRef]
  355. Pucharcos, C.; Fuentes, J.J.; Casas, C.; de la Luna, S.; Alcantara, S.; Arbones, M.L.; Soriano, E.; Estivill, X.; Pritchard., M. Alu-splice cloning of human intersectin (itsn), a putative multivalent binding protein expressed in proliferating and differentiating neurons and overexpressed in Down syndrome. Eur. J. Hum. Genet. 1999, 7, 704–712. [Google Scholar] [CrossRef]
  356. Wilmot, B.; McWeeney, S.K.; Nixon, R.R.; Montine, T.J.; Laut, J.; Harrington, C.A.; Kaye, J.A.; Kramer, P.L. Translational gene mapping of cognitive decline. Neurobiol. Aging 2008, 29, 524–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  357. Nixon, R.A. Endosome function and dysfunction in Alzheimer’s disease and other neurodegenerative diseases. Neurobiol. Aging 2005, 26, 373–382. [Google Scholar] [CrossRef]
  358. Nishimura, T.; Yamaguch, T.; Tokunaga, A.; Hara, A.; Hamaguchi, T.; Kato, K.; Iwamatsu, A.; Okano, H.; Kaibuchi, K. Role of numb in dendritic spine development with a Cdc42 GEF intersectin and EphB2. Mol. Biol. Cell 2009, 17, 1273–1285. [Google Scholar] [CrossRef]
  359. Gunner, G.; Cheadle, L.; Johnson, K.M.; Ayata, P.; Badimon, A.; Mondo, E.; Nagy, M.A.; Liu, L.; Beiller, S.M.; Kim, K.-W.; et al. Sensory lesioning induces microglial synapse elimination via ADAM10 and fractalkine signaling. Nat. Neurosci. 2019, 22, 1075–1088. [Google Scholar] [CrossRef]
  360. Jiang, T.; Zhang, Y.D.; Gao, Q.; Zhou, J.S.; Zhu, X.C.; Lu, H.; Shi, J.-Q.; Tan, L.; Chen, Q.; Yu, J.-T. TREM1 facilitates microglial phagocytosis of amyloid beta. Acta Neuropathol. 2016, 132, 667–683. [Google Scholar] [CrossRef]
  361. Edwards, F.A. A unifying hypothesis for Alzheimer’s disease: From plaques to neurodegeneration. Trends Neurosci. 2019, 42, 310–322. [Google Scholar] [CrossRef]
  362. Magdesian, M.H.; Carvalho, M.M.; Mendes, F.A.; Saraiva, L.M.; Juliano, M.A.; Juliano, L.; Garcia-Abreu, J.; Ferreira, S.T. Amyloid-beta binds to the extracellular cysteine-richdomain of Frizzled and inhibits Wnt/beta-catenin signaling. J. Biol. Chem. 2008, 283, 9359–9368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  363. Araujo, B.H.S.; Kaid, C.; De Souza, J.S.; Gomes da Silva, S.; Goulart, E.; Caires, L.C.J.; Musso, C.M.; Torres, L.B.; Ferrasa, A.; Herai, R.; et al. Down syndrome iPSC-derived astrocytes impair neuronal synaptogenesis and the mTOR pathway in vitro. Mol. Neurobiol. 2018, 55, 5962–5975. [Google Scholar] [CrossRef]
  364. Weston, M.C.; Chen, H.; Swann, J.W. Multiple roles for mammalian target of rapamycin signaling in both glutamatergic and GABAergic synaptic transmission. J. Neurosci. 2012, 32, 11441–11452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  365. Zhang, L.; Zhou, F.; ten Dijke, P. Signaling interplay between transforming growth factor-beta receptor and PI3K/AKT pathways in cancer. Trends Biochem. Sci. 2013, 38, 612–620. [Google Scholar] [CrossRef] [PubMed]
  366. Huang, H.; Liu, H.; Yan, R.; Hu, M. PI3K/Akt and ERK/MAPK Signaling Promote Different Aspects of Neuron Survival and Axonal Regrowth Following Rat Facial Nerve Axotomy. Neurochem. Res. 2017, 42, 3515–3524. [Google Scholar] [CrossRef] [PubMed]
  367. Chidambaram, H.; Chinnathambi, S. G-protein coupled receptors and tau-different roles in Alzheimer’s disease. Neuroscience 2020, 438, 198–214. [Google Scholar] [CrossRef] [PubMed]
  368. Duka, V.; Lee, J.-H.; Credle, J.; Wills, J.; Oaks, A.; Smolinsky, C.; Shah, K.; Mash, D.C.; Masliah, E.; Sidhu, A. Identification of the sites of tau hyperphosphorylation and activation of tau kinases in synucleinopathies and Alzheimer’s diseases. PLoS ONE 2013, 8, e75025. [Google Scholar] [CrossRef]
  369. Zhu, C.; Xu, B.; Sun, X.; Zhu, Q.; Sui, Y. Targeting CCR3 to reduce amyloid-b production, Tau hyperphosphorylation, and synaptic loss in a mouse model of Alzheimer’s disease. Mol. Neurobiol. 2017, 54, 7964–7978. [Google Scholar] [CrossRef]
  370. Thathiah, A.; De Strooper, B. The role of G protein-coupled receptors in the pathology of Alzheimer’s disease. Nat. Rev. Neurosci. 2011, 12, 73. [Google Scholar] [CrossRef]
  371. Niswender, C.M.; Conn, P.J. Metabotropic glutamate receptors: Physiology, pharmacology, and disease. Annu. Rev. Pharm. Toxicol. 2010, 50, 295–322. [Google Scholar] [CrossRef] [Green Version]
  372. Lee, Y.-S.; Silva, A.J. The molecular and cellular biology of enhanced cognition. Nat. Rev. Neurosci. 2009, 10, 126. [Google Scholar] [CrossRef] [PubMed]
  373. Rueda, N.; Flórez, J.; Dierssen, M.; Martínez-Cué, C. Translational validity and implications of pharmacotherapies in preclinical models of Down syndrome. Prog. Brain Res. 2020, 251, 245–268. [Google Scholar] [PubMed]
  374. Panza, F.; Lozupone, M.; Seripa, D.; Imbimbo, B.P. Amyloid-β immunotherapy for alzheimer disease: Is it now a long shot? Ann. Neurol. 2019, 85, 303–315. [Google Scholar] [CrossRef] [PubMed]
  375. Zmijewski, P.A.; Gao, L.Y.; Saxena, A.R.; Chavannes, N.K.; Hushmendy, S.F.; Bhoiwala, D.L.; Crawford, D.R. Fish oil improves gene targets of Down syndrome in C57BL and BALB/c mice. Nutr. Res. 2015, 35, 440–448. [Google Scholar] [CrossRef]
  376. Martínez-Martínez, S.; Redondo, J.M. Inhibitors of the calcineurin/NFAT pathway. Curr. Med. Chem. 2004, 11, 997–1007. [Google Scholar] [CrossRef]
  377. Wang, C.; Yu, J.T.; Miao, D.; Wu, Z.C.; Tan, M.S.; Tan, L. Targeting the mTOR signaling network for Alzheimer’s disease therapy. Mol. Neurobiol. 2014, 49, 120–135. [Google Scholar] [CrossRef]
  378. Amidfar, M.; de Oliveira, J.; Kucharska, E.; Budni, J.; Kim, Y.K. The role of CREB and BDNF in neurobiology and treatment of Alzheimer’s disease. Life Sci. 2020, 257, 118020. [Google Scholar] [CrossRef]
  379. Zhao, S.; Zhang, L.; Yang, C.; Li, Z.; Rong, S. Procyanidins and Alzheimer’s Disease. Mol. Neurobiol. 2019, 56, 5556–5567. [Google Scholar] [CrossRef]
  380. Congdon, E.E.; Sigurdsson, E.M. Tau-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 399–415. [Google Scholar] [CrossRef]
  381. Allen, S.J.; Watson, J.J.; Shoemark, D.K.; Barua, N.U.; Patel, N.K. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharm. Ther. 2013, 138, 155–175. [Google Scholar] [CrossRef]
  382. Chu, L.W. Alzheimer’s disease: Early diagnosis and treatment. Hong Kong Med. J. 2012, 18, 228–237. [Google Scholar] [PubMed]
  383. Macdonald, R.; Barnes, K.; Hastings, C.; Mortiboys, H. Mitochondrial abnormalities in Parkinson’s disease and Alzheimer’s disease: Can mitochondria be targeted therapeutically? Biochem. Soc. Trans. 2018, 46, 891–909. [Google Scholar] [CrossRef] [PubMed]
  384. Boccardi, V.; Murasecco, I.; Mecocci, P. Diabetes drugs in the fight against Alzheimer’s disease. Ageing Res. Rev. 2019, 54, 100936. [Google Scholar] [CrossRef] [PubMed]
  385. Frautschy, S.A.; Cole, G.M. Why pleiotropic interventions are needed for Alzheimer’s disease. Mol. Neurobiol. 2010, 41, 392–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  386. Wang, S.; Colonna, M. Microglia in Alzheimer’s disease: A target for immunotherapy. J. Leukoc. Biol. 2019, 106, 219–227. [Google Scholar] [CrossRef] [PubMed]
  387. Cummings, J.L.; Tong, G.; Ballard, C. Treatment Combinations for Alzheimer’s Disease: Current and Future Pharmacotherapy Options. J. Alzheimers Dis. 2019, 67, 779–794. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Graphical display of the main pathways (circled) implicated in each pathological characteristic of AD (squared) as well as their interconnections. Line and arrow colors depict the influence of the different signalling pathways and/or pathological characteristics circled or squared using the same color on other pathways or systems. Black arrows represent the feedback loops between the main pathological characteristics of AD. ↑: up-regulated, ↓down-regulated.
Figure 1. Graphical display of the main pathways (circled) implicated in each pathological characteristic of AD (squared) as well as their interconnections. Line and arrow colors depict the influence of the different signalling pathways and/or pathological characteristics circled or squared using the same color on other pathways or systems. Black arrows represent the feedback loops between the main pathological characteristics of AD. ↑: up-regulated, ↓down-regulated.
Ijms 21 06906 g001
Table 1. Signalling pathways implicated in the main neuropathological characteristics of Alzheimer’s disease in individuals with and without Down syndrome.
Table 1. Signalling pathways implicated in the main neuropathological characteristics of Alzheimer’s disease in individuals with and without Down syndrome.
Neuropathological CharacteristicSignalling PathwayUp- or DownregulationPathophysiological Role in AD
Amyloid plaquesAPP [31]↑ in DS and ADGeneration of Aβ oligomers
DYR1A [37,38,39,40,47]↑ in DS and ADAβ degradation, APP phosphorylation
RCAN1/NFAT [42,43,45,46,47,48]↑ RCAN/↓ NFAT in DS and ADMediation of Aβ-induced neuronal death, disruption of Ca2+ homeostasis
PIK3/Akm/mTOR [51,52,55,56,57,64,65,66,67,68,74,75]↑ in DS and ADContribution to Aβ generation and aggregation, inhibition of autophagy, reduction of Aβ clearance
CREB [80,81,82,83,84,85,86]↓ in DS and ADInduction of synaptic loss by Aβ
BDNF/NGF [88,92,93,98,99,100]↓ in DS and ADAccumulation of APP C-terminal fragments and aggregation of Aβ
Neurofibrillary tanglesDYRK1A [117,118,119,120,121,122,123,124,125]↑ in DS and ADModifications in tau splicing and enhancement of tau phosphorylation
RCAN1/NFAT [126,127,128,129,130,131]↑ RCAN/↓ NFAT in DS and ADPrevention of tau degradation and enhancement of tau phosphorylation
CDK5 [133,134,135,136]↑ in DSEnhancement of tau phosphorylation
PP2A [137,138,139,140,141]↓ in DSEnhancement of tau phosphorylation
mTOR/SIRT1 [56,143,144,145]↑ mTOR/↓ SIRT in DS and ADEnhancement of tau phosphorylation, promotion of tau accumulation
Cholinergic system [156,157,158,159,160,161,162,163,164,165,166,167,168,169]↓ in DS and ADTau pathology in cholinergic neurons that aggravates neurodegeneration
Cholinergic neurodegenerationNGF/proNGF/TrkA/p75NTR [170,171,172,173]↓ in DS and ADReduction in survival of cholinergic neurons
Aβ [172,173,174]↑ in DS and ADFacilitation of cholinergic neurodegeneration
Oxidative stressSOD1 [182]↑ in DSInduction of Redox imbalance
RCAN1/NFAT [46,183,184,185,186,187]↑ RCAN/↓ NFAT in DS and ADAlterations in mitochondrial function and increase in ROS production
APP/Aβ [188,191]↑ in DS and ADEnhancement of lipid, DNA, and RNA oxidation
Glutamatergic system [189,190]↑ in ADPromotion of OS-induced excitotoxicity
Cholinergic system [191]↓ in DS and ADAβ-induced enhancement of OS in cholinergic neurons
mTOR [192,193,194,196,198]↑ in DS and ADOS disruption of mTOR function and mTOR enhancement of oxidative damage
Mitochondrial dysfunctionEnhanced oxidative stress [191,200,201]↑ in DS and ADEnhancement of ROS-mediated disruption of mitochondrial integrity and function
OXPHOS [203,204,205,206,209,210,211,212,213,214,215,216,217,218,219,220,221,222]↓ in DS and ADEnhancement of Aβ production, alterations in cell membranes and synapses, reduction in mitochondrial inner membrane potential, reduction in energy production, and lower mitochondrial function
Raptor/mTOR [197,223,224]↑ in DS and ADAlterations in mitochondrial activity and metabolism
Energy metabolismInsulin signaling [241,242,243]↓ in DS and ADAlterations in energy metabolism, impairment of neuronal activity, plasticity and survival, and facilitation of Aβ aggregation
Glucose transport and metabolism [245,246,247,248,249,250]↓ in DS and ADReduction in energy for synaptic transmission and neurotransmitter biosynthesis, alterations in autophagy
PI3-K/Akt/mTOR [65,67,253–256↑ in DS and ADDysregulation of energy balance, induction of insulin resistance, altered autophagy
Cellular senescenceRelease of proinflammatory cytokines [269,270]↑ in DS and ADInduction of cellular senescence and enhancement by senescence
Oxidative stress and mitochondrial dysfunction [15,262]↑ in DS and ADInduction of cellular senescence and enhancement by senescence
Proteostasis (Aβ and tau) [267,268]↑ in DS and ADInduction of cellular senescence and enhancement by senescence, induction of cellular senescence and enhancement by senescence
USP16-Wnt [271,272,273]↑ UPS16 in DS/ ↓ Wnt in DS and ADInduction of senescence through DNA damage, downregulation of the Wnt pathway reducing stem cell renewal
Immune response/inflammationp38MAPK [281,282,283,284,285]↑ in DS and ADIncrease in release of cytokines
Aβ/APP [287,290]↑ in DS and ADIncrease in release of cytokines which further aggravates Aβ pathology
HPA [292,293]↑ in ADCytokines produce excessive activation of the HPA, which aggravates the energy deficits and enhances OS
Wnt [304]↓ in DS and ADAltered microglia activation, enhancement of neuroinflammation, tau hyperphosphorylation, and synaptic loss
Cell proliferation/differentiation and migrationDYRK1A [305,306,307]↑ in DS and AD Induction of cell cycle exit, premature differentiation or precursors resulting in a reduced number of adult neurons
DYRK1A/DREAM [308,309]-Inhibition of cell proliferation due to cell cycle arrest
DYRK1A/Cyclin D1 [310,311]Inhibition of proliferation and promotion of premature differentiation, prevention the entry into the S phase of the cycle
DYRK1A/Notch [312,313,314]Inhibition of notch signaling that controls neurogenesis, induction of a shift from neurogenic to glionenic fate of progenitors
DYRK1A/NFAT [316]Delay of neurogenesis by the synergic effect with RCAN1
mTOR [317,318]↑ in DS and ADApoptotic death of NPCs
BDNF [92,322]↓ in DS and ADImpairment of cell proliferation and differentiation
Shh [323,324,325]↓ in DSImpairment of proliferation of NPCs
APP [327]↑ in DS and ADAlterations in cell cycle regulation, neural precursor maturation
Neurotransmitter releaseDYRK1A [328,330]↑ in DS and ADReductions in neurotransmitter synthesis and release
RCAN1 [331,332,333]↑ in DS and ADReductions in neurotransmitter synthesis and release
SynapsesDYRK1A [334,335,336,337,338,339,340,341,342,343]↑ in DS and ADImpairment in dendritic growth and complexity; dendritic spine formation; reduction of synaptic components necessary for synapse formation, maintenance, and functioning
RCAN1/NFAT [344,345,346,349]↑ RCAN/↓ NFAT in DS and ADModification of the localization of synaptic proteins, decreased phosphorylation of proteins necessary for synaptic plasticity
DSCAM [351]↑ in DS and ADInhibition of dendritic branching and synapse formation
ITSN [353,355,356,357,358]↑ in DS and ADEnlargement of the early endosomal compartment, altered endocytic trafficking, leading to a reduced number and recycling of synaptic vesicles
Wnt [359,362]↓ in DS and ADAlterations in synapse number and function
PI3K/AKT/mTOR [365,366]↑ in DS and ADLoss of synapses partly mediated by enhanced cytokine release, impairment of synaptic development
ReceptorsMuscarinic ACh receptors [158]↓ in DS and ADImpairment of cholinergic transmission: the loss of these receptors is mediated by tau phosphorylation
CXR2 and CC3 chemokine receptors [369,370]↑ in ADEnhancement of tau phosphorylation ad cytokine release
mGluR2 receptors [371]↑ in ADEnhancement of tau phosphorylation
Other GPCRs [367]-Alteration of neurotransmission by different mechanisms including tau phosphorylation, increased cytokine release, and aggravation of amyloid pathology
Aβ: β-amyloid; ACh: acetylcholine; AD: Alzheimer’s disease; APP: Amyloid Precursor Protein; BDNF: Brain-Derived Neurotrophic Factor; CREB: cAMP Response Element-Binding protein; CDK5: Cyclin-Dependent Kinase 5; DS: Down syndrome: DSCAM: Down syndrome Cell Adhesion Molecule; DYRK1A: Dual Specificity Tyrosine-Regulated Protein Kinase 1; GPCR: G-protein coupled receptors; HPA: Hypothalamic-Pituitary-Adrenal axis; ITSN: Intersectin; mGluR2: metabotropic glutamate receptor 2; mTOR: Mammalian Target of Rapamycin; NFAT: Nuclear Factor of Activated T cells; NGF: Nerve Growth Factor, OXPHOS: Oxidative Phosphorylation; p38MAPK: p38 Mitogen-Activated Protein Kinase; PIK3: Phosphoinositide 3 Kinase; PP2A: Phosphatase 2A; RCAN1: Regulator of Calcineurin 1; Shh: Sonic Hedgehog; SIRT1: Sirtuin 1; SOD1: Superoxide Dismutase; USP16: Ubiquitin-Specific Peptidase 16; Wnt: Wingless and Int-1; ↑: up-regulated, ↓down-regulated.

Share and Cite

MDPI and ACS Style

Martínez-Cué, C.; Rueda, N. Signalling Pathways Implicated in Alzheimer′s Disease Neurodegeneration in Individuals with and without Down Syndrome. Int. J. Mol. Sci. 2020, 21, 6906. https://doi.org/10.3390/ijms21186906

AMA Style

Martínez-Cué C, Rueda N. Signalling Pathways Implicated in Alzheimer′s Disease Neurodegeneration in Individuals with and without Down Syndrome. International Journal of Molecular Sciences. 2020; 21(18):6906. https://doi.org/10.3390/ijms21186906

Chicago/Turabian Style

Martínez-Cué, Carmen, and Noemí Rueda. 2020. "Signalling Pathways Implicated in Alzheimer′s Disease Neurodegeneration in Individuals with and without Down Syndrome" International Journal of Molecular Sciences 21, no. 18: 6906. https://doi.org/10.3390/ijms21186906

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop