Abstract
Although the two pathological hallmarks of Alzheimer's disease (AD), senile plaques composed of amyloid-β (Aβ) peptides and neurofibrillary tangles (NFTs) consisting of hyperphosphorylated tau, have been studied extensively in postmortem AD and relevant animal and cellular models, the pathogenesis of AD remains unknown, particularly in the early stages of the disease where therapies presumably would be most effective. We and others have demonstrated that Aβ plaques and NFTs are present in varying degrees before the onset and throughout the progression of dementia. In this regard, aged people with no cognitive impairment (NCI), mild cognitive impairment (MCI, a presumed prodromal AD transitional state), and AD all present at autopsy with varying levels of pathological hallmarks. Cognitive decline, a requisite for the clinical diagnosis of dementia associated with AD, generally correlates better with NFTs than Aβ plaques. However, correlations are even higher between cognitive decline and synaptic loss. In this review, we illustrate relevant clinical pathological research in preclinical AD and throughout the progression of dementia in several areas including Aβ and tau pathobiology, single population expression profiling of vulnerable hippocampal and basal forebrain neurons, neuron plasticity, neuroimaging, cerebrospinal fluid (CSF) biomarker studies and their correlation with antemortem cognitive endpoints. In each of these areas, we provide evidence for the importance of studying the pathological hallmarks of AD not in isolation, but rather in conjunction with other molecular, cellular, and imaging markers to provide a more systematic and comprehensive assessment of the multiple changes that occur during the transition from NCI to MCI to frank AD.
Introduction
At the turn of the twentieth century, several prominent neurobiologists described the presence of extracellular lesions in the brain of people with dementia. Blocq and Marinesco (1892) reported abnormal argyrophilic extracellular plaques in post mortem brain tissue from two aged persons with dementia [1, 2], a finding confirmed by Emil Reich and Oskar Fisher (Fig. 1A) [3]. The German psychiatrist, Alois Alzheimer (Fig. 1B), was treating a 51 year old woman named Auguste Deter (Fig. 1C), who presented with signs of paranoia and memory loss, who subsequently died five years later. At autopsy, Alzheimer observed brain atrophy, neuronal loss, and dense argyrophilic fibrillary bundles in form of tangles (Fig. 1D, E) and plaques (Fig.1F). In 1910, Dr. Emil Kraepelin, suggested that this triad of signs and symptoms be termed “Alzheimer's disease”. Today, senile plaques and neurofibrillary tangles (NFTs) are considered the defining pathological lesions of Alzheimer's disease (AD).
Figure. 1.
Photomicrographs of (A) Oskar Fisher, (B) Alois Alzheimer, (C) Auguste Deter. (D) Neurofibrillary tangle visualized by the Bielschowsky silver method from the brain of Auguste Deter, (E) NFT containing the tau epitope Alz-50 and (F) Fluorescent image of a cored amyloid beta immunostained plaque (red) surrounded by a GFAP positive glia cell (green) in AD frontal cortex.
Amyloid plaques accumulate in the extracellular matrix (Fig. 1F) and consist of insoluble fibrils of amyloid-beta peptides (Aβ), which are cleaved from the larger transmembrane Aβ precursor protein (APP) by successive cleavage through the beta-site APP cleaving enzyme 1 (BACE1) and the intramembrane γ secretase complex [4] [5]. NFTs are argyrophilic aggregates of hyperphosphorylated tau protein [6] [7]. Both protein aggregates display a β-pleated sheet conformation and are thought to interfere with cytoskeletal integrity, disrupt axonal transport, synapse and neuronal function. In a small portion of cases (<1%), the disease has an autosomal dominant pattern of inheritance termed familial AD (FAD), caused by mutations in one of three identified genes: APP, presenilin 1 (PS1), or presenilin 2 (PS2) [8]. Genotyping of tissue from the original histological slides from Deter's brain autopsy revealed she likely had PS1-linked FAD [9] [10]. In contrast to the original “amyloid cascade” hypothesis that Aβ plaques drive the neuropathological cascade leading to dementia [11], Dr. Alzheimer wrote “...the plaques are not the cause of senile dementia, but only an accompanying feature of senile involution of the central nervous system” [12] [13]. In 1999, Mesulam [14] wrote, “It seems as if the Aβ plaques appear at the wrong time and in the wrong places with respect to the clinical dementia and there is little evidence that they cause the NFT”. The revised amyloid cascade hypothesis posits that soluble Aβ oligomers initiate the pathological cascade of AD leading to synaptic dysfunction, neuronal cell death, and dementia[15]. These findings support the contention that amyloid deposition is not the singular pathogenetic cause of sporadic AD.
AD is now thought to have an extensive preclinical stage, which is initiated 15-20 years prior to emergence of clinical signs (Fig. 2) [16, 17]. The clinical concept of mild cognitive impairment (MCI) arose from memory clinics. In the 1990's, some cases were characterized by an amnestic disorder and classified as amnestic MCI (aMCI) [18]. Although memory disorder clinics reported that aMCI was a more common form, MCI comprises a heterogeneous population: those with memory deficits only are referred to as single domain aMCI, while those with a deficit in memory and another cognitive domain are termed multi-domain MCI (mdMCI) [19]. Individuals with aMCI are at a higher risk of developing AD [19]. MCI was initially characterized as an intermediate phase when NFT and Aβ lesions are increased relative to people with no cognitive impairment (NCI) [20] [21]. However, Accumulating evidence revealed that many clinically diagnosed NCI or MCI cases exhibit significant amyloid plaque (Fig. 3) as well as NFT pathology equal to or greater than what is often seen neuropathologically in mild to moderate AD, challenging the singular notion that these lesions alone drive dementia onset [21] [22] [23]. The demonstration that a cohort of NCI subjects who came to autopsy with high amyloid and NFT pathology support the developing concept of pathological aging or preclinical AD phase [24] [25].
Figure. 2.
Diagram illustrating the extensive preclinical stage of Alzheimer's disease.
Figure. 3.
Photomicrographs showing the distribution of β-amyloid staining within the entorhinal cortex in MCI. A. Note that the crescent shaped band of β-amyloid staining extends to the border of the perirhinal cortex. B. Higher magnification of the region outlined in panel A by curved arrows. C. Higher magnification photomicrograph of densely stained β-amyloid plaques (black arrow in B). D. Cluster of neurofibrillary tangles within layer 2 of the EC (open arrow in B). Scale bar represents: A=800μm, B=200μm, C and D=40μm. (E) Histogram showing the adjusted b-amyloid loads for each NCI, MCI, and AD case examined. The means and standard errors are indicated above each diagnostic group. Adopted with permission from [47]
Despite the extensive emphasis on amyloid and tau as the major factors leading to dementia onset and serving as the predominant focus of drug discovery and clinical trials, it is becoming evident that these lesions are not acting in isolation during the preclinical phases of AD [22] [26]. The fact that virtually every anti-amyloid clinical trial to date has not met its primary end-points of improving cognition [27] indicates that a monotherapy strategy may not be sufficient to treat this pervasive progressive multisystem neurodegenerative disorder [28]. On the other hand, it has been argued that AD is a disease of long projections neurons innervating the hippocampus and neocortex [26] [29], that degenerate early in the disease progression ) [26] [29], which may disrupt brain connectivity and neuroplasticity. Although not receiving as much attention as amyloid and tau pathobiology, herein we review the molecular and cellular neuropathobiology of preclinical and prodromal AD. This information is essential for the development of appropriate therapeutic targets aimed at preventing or slowing cognitive decline prior to frank AD.
Amyloid and NFT pathology
Aβ peptides, which forms the core of parenchymal Aβ plaques are generated from APP, a holoprotein that is cleaved by successive β- and γ-secretases to form toxic Aβ [8].The distribution of Aβ plaques changes with time and reflects the spread of Aβ deposition in the diseased brain [30]. Diffuse and “fleecy” plaques appear first throughout the neocortex and extend hierarchically into other brain regions [31]. In the next stages, Aβ plaques occur in allocortical areas, the basal ganglia, thalamus, hypothalamus, midbrain, medulla, pons and cerebellum. Later stages feature neuritic and dense cored Aβ plaques [32]. The other major AD pathological features are NFTs composed of aggregates of hyperphosphorylated forms of the tau protein (Fig. 1D, E) [33, 34]. NFT formation and maturation progresses according to a linear sequence of molecular and conformational alterations of the tau molecule [35]. Braak and Braak [32] described the topographic spread of NFTs from the mesial temporal lobe (MTL) to the neocortex, according to six stages depending upon location of the NFT-bearing neurons and the severity of lesions (transentorhinal stages I-II: clinically silent cases; limbic stages III-IV: incipient AD; neocortical stages V-VI: fully developed AD). Clinical subtypes of MCI as well as NCI cases display Braak staging scores ranging from stages 0-V [36]. These observations indicate that there may be no clear demarcation of neuropathology between some NCI, MCI and AD brains using the Thal staging criteria for amyloid [37] or the Braak staging criteria for NFTs [38].
NFT and cognition in preclinical AD
Although tau and Aβ pathology increase with age [39], their association with antimortem cognitive function remains relatively unexplored. NCI cases displaying AD neuropathology at autopsy despite a lack of clinically significant antemortem cognitive impairment may represent a pre-clinical disease stage [40]. Studies suggest a negative association between the presence of both Aβ plaques and NFTs with ante-mortem cognitive performance [30] suggesting that older individuals in the pre-clinical stages of AD according to their brain pathology at time of death continue to perform within normal limits on cognitive tests. Whether these individuals represent preclinical MCI remains an intriguing area of research.
While several groups have investigated the neuropathology of MCI and possible/probable AD, very few have concentrated on the relationship of Braak staging to clinical dysfunction in people with NCI [41]. An investigation of a small number of NCI subjects from the Rush Religious Orders Study (RROS) cohort revealed relatively similar percentages of low (40%) and high (60%) Braak scores compared to NCI, MCI and AD subjects examined [42] demonstrated a minor association between episodic memory and neuropathology using NIA-Reagan Institute neuropathological criteria for AD, which combines the assessment of neuritic plaques using CERAD scores and NFT pathology using Braak scores [43]. Individuals with low and high Braak scores and moderate to frequent CERAD scores displayed greater hippocampal and total brain volume loss suggesting brain volume as a variable that effects cognitive decline in the face of AD neuropathology [44]. Recently, we evaluated the association of low and high Braak NFT scores upon several cognitive domains {episodic, executive, semantic and a global cognitive score (GCS)} and plaque pathology in a cohort of elderly persons who were diagnosed as NCI at last clinical evaluation before death [22]. In this cross sectional investigation, the majority of the NCI subjects were Braak stage III (intermediate) and none were Braak stage VI (severe) (Fig. 4). Interestingly, females were significantly more likely to have higher Braak scores (III-V) despite being NCI, which is highly relevant due to the known gender differences associated with AD [45] [46]
Figure 4.
Linear graphs showing relationships between cognitive domains (A), Braak scores, (B) APOE ε4 status and (C) gender for the cohort of cases who died with clinical diagnosis of no cognitive impairment but post mortem neuropathological evaluation revealed amyloid and plaque pathology similar to those with AD dementia. Note the significant differences between male and female for semantic memory (p=0.006) and visuospatial (p=0.04) scores. Circles and triangles represent mean z-scores, error bars represent standard deviation. Adapted from with permission from [187]
Although the low Braak group of NCI performed better on most cognitive domains, only performance on semantic memory was significantly higher but this difference was not maintained when the data was adjusted for age, gender and years of education suggesting that this relationship was not directly related to Braak stage [36]. Hence, NFTs within the entorhinalperirhinal cortex (stages I–II), hippocampus (stages III–IV) and neocortex (stage V) may not be a necessary precondition for cognitive impairment in the elderly. We also evaluated whether Braak stage was associated with longitudinal change in the same cognitive domains [36]. Surprisingly, age but not Braak stage played a more prominent role in cognitive decline in NCI over time suggesting that the extent of NFT pathology does not directly influence cognitive decline in the elderly. Interestingly, NCI females, which displayed significantly greater NFT pathology (discussed above), performed worse than males on semantic memory and visuospatial domain tests, suggesting that NFT pathology influences cognitive decline in females to a greater degree than males. In addition, we found significant correlations between hippocampal CA1 neuritic plaques (NPs) and episodic memory or GCS [47] suggesting that this type of a lesion combining Aβ (plaque) and tau (dystrophic neurites) pathologies plays a more critical role in cognitive decline during this preclinical stage of dementia.
Amyloid deposition and cognitive decline
A review of the literature suggests that cognition is affected by NPs and not diffuse plaques (DPs) [48] [49]. Greater hippocampal burden of NPs correlated with worse episodic memory and global cognitive performance and greater hippocampal/entorhinal cortex Aβ load was associated with greater NFT pathology in cases with an antemortem clinical diagnosis of NCI and a continuum of Braak stages [22]. In this same cohort, we found that NPs but not DPs were associated with significantly lower cognitive test performance across several cognitive domains [50], supporting previous reports [30, 51]. We also observed that NPs correlated with Braak stage stronger than DPs [30] [48]. Although we found that DPs were more prevalent than NPs in early Braak stages, NPs tended to increase and were nearly equal to DPs in higher Braak stages in NCI. Moreover, we found that in pathology burdened NCI, higher Braak stages were associated with greater amyloid plaque load in the entorhinal cortex but not in the hippocampus [47]. Group analysis found no significant effects of either hippocampal or entorhinal cortex amyloid load or APOE ε4 status on Braak score. These findings demonstrate that people with NCI exhibit a wide range of Braak scores and plaque pathology similar to prodromal and frank AD.
Oxidative stress and synaptic homeostasis in the early progression of AD
Recently, we evaluated the relationship between oxidative stress and synaptic homeostasis in the early progression of AD [52] using NCI cases that may represent preclinical aMCI or preclinical AD (PCAD) from the University of Kentucky Alzheimer's Disease Brain Bank. Here, we compared age- and postmortem interval-matched hippocampus from NCI individuals who came to autopsy with high levels of AD pathology (HPNCI), low or no AD pathology (LPNCI) and an aMCI group for changes in both presynaptic (e.g., Synapsin-1, Synaptophysin) and postsynaptic (e.g., Drebrin, PSD-95 and SAP-97) proteins and markers of oxidative stress (e.g., protein carbonyls, 4-hydroxynonenal and 3-nitrotyrosine) (Fig. 5). Surprisingly, both the HPNCI and aMCI cohorts showed a significant decline in levels of the different synaptic proteins compared to LPNCI but aMCI and HPNCI groups did not differ significantly from each other (Fig. 5A, B). Measurements of oxidative stress also showed significant group differences with the greatest elevations observed in aMCI and HPNCI (Fig. 5C) and a significant association between changes in oxidative stress and synaptic proteins. The decline in both presynaptic- and postsynaptic proteins indicates a loss of synaptic homeostasis in the hippocampus early in the AD process. The increase in oxidative stress in aMCI was expected since numerous previous studies have emphasized such a scenario in the progression of the disease [53-56] Unexpected was that oxidative stress increased in the HPNCI cohort, further supporting the idea that this group may represent PCAD.
Figure 5.
Scatter plots showing changes in different synaptic proteins within the hippocampus for each subject from the different cohorts: Low pathology no cognitive impairment (LPNCI), high pathology no cognitive impairment (HPNCI), amnestic mild cognitive impairment (aMCI). (A) Antibodies directed against presynaptic proteins synapsin-1 and synaptophysin are shown. (B) Antibodies directed against postsynaptic proteins Drebrin, SAP-97, and PSD-95. Horizontal lines indicate group medians. *p<0.05, *p<0.01, ***p<0.005, #p<0.001 compared to LPNCI. (C) Representative slot-blot for protein carbonyl, 4-hydroxynonenal, and 3-nitrotyrosine fractions from individuals classified as no cognitive impairment and low AD-like pathology (LPNCI), no cognitive impairment and high AD-like pathology (HPNCI), and amnestic mild cognitive impairment (aMCI). (D) Scatterplots showing the levels of soluble Aß1-42. No significant difference were observed between the two groups with individuals labeled as NCI (p > 0.1). However, both the low pathology and high pathology NCI groups were significantly lower than the aMCI) cohort. *p<0.05. Circles=LPNCI, Squares=HPNCI, Triangles=aMCI. Adapted from with permission from [188]
The factors underlying the decline in synaptic proteins in HPNCI cases are still to be determined. It may be related to changes in oxidative stress, since we found that levels of both pre- and postsynaptic proteins decreased as oxidative stress increased. Another possibility is that synaptic protein loss is associated with an increase in soluble Aβ1-42 (sAβ1-42), which has been shown to increase during the progression of AD and is closely associated with changes in cognition [57]. Examination of the role of sAβ1-42 in the synaptic decline of the HPNCI group synaptic decline revealed that while the levels in aMCI were significantly higher than both NCI groups, there was no difference between the LPNCI and HPNCI, suggesting that the decline in synaptic proteins was unrelated to sAβ1-42 levels (Fig. 5D). In addition, we found a significant association between CERAD scores and the levels of sAβ1-42 in aMCI suggesting that while changes in sAβ1-42 are probably downstream of synaptic changes, there are additional factors responsible for the transition from PCAD to aMCI.
During the last few years, oligomeric Aβ species have been suggested to be one of the triggering mechanism for the synaptic impairment and tau phosphorylation in AD [8, 58] [59]. Cortical levels of Aβ oligomers accumulate in MCI and mild to moderate AD compared to age matched non-demented controls [60], correlate with severity of cognitive impairment, Braak staging, and lower levels of presynaptic and postsynaptic proteins [61]. Oligomeric Aβ within synaptic terminals is associated with the accumulation of hyperphosphorylated tau in AD [59]. Interestingly, dimeric Aβ induces tau phosphorylation and dystrophic axonal profiles [62], suggesting a prion-like seeding or propagation mechanism underlying AD pathogenesis [63] or that selectively vulnerable subcortical long projection neurons may be sensitive to retrograde toxicity of Aβ oligomers. Therefore, therapeutic agents targeting synaptic Aβ oligomers early in the disease process may slow disease onset and trajectory.
Mechanisms delaying cognitive decline in non-cognitively impaired elderly
It is still unclear why some individuals with high AD pathology continue to perform well on cognitive tests. It is possible that NFTs and plaques are simply pathological bystanders and do not necessarily contribute to the initiation of cognitive dysfunction. Had these individuals lived longer they may transition to aMCI. Alternatively, one might consider the concepts of brain resilience and/or cognitive reserve as an explanation for this paradox [64] [65] [66] [67]. Brain reserve postulates a greater number of either neurons or synapses at the initiation of disease allowing the continued performance of cognitive tasks [65, 66]. On the other hand, cognitive reserve may involve the recruitment of brain regions either not or less severely affected by the disease process, which aid in task performance and even development of new brain connections [66]. Although the mechanism(s) initiating these constructs remain unknown, one mechanism may be related to brain neuroplasticity.
Brain Plasticity in MCI
The MTL memory circuit is highly neuroplastic [68] [69] and behaviorally resistant [22] to AD pathology. For example, similar entorhinal cortex amyloid loads were found between NCI, MCI and AD cases [47]. Studies using hippocampal tissue revealed a preservation of synapse number [70], synaptic protein [71], and neurotrophic factor receptors [72] in non-demented aged subjects with a wide range of Braak stages as well as an increase in dendritic spine size in prodromal AD [73]. Clinical pathological investigations also indicate preserved number and larger neuronal size in the hippocampus in “asymptomatic AD” compared with normal, MCI and clinical AD [74] indicating early reorganizational responses. A report revealed a significant increase in dendritic length (18%) and complexity (23%) in hippocampal CA1 pyramidal in MCI compared to NCI individuals (Fig. 6A-C, F) and amyloid plaques distorted the dendritic architecture of CA1 neurons (Fig. 6E) [73]. Conversely, there was a significant reduction in branch length (−39%) and arbor complexity (−25%) during progression from MCI to AD (Fig. 5). Interestingly, increased synaptic contact size has been observed early in AD [75]. These types of early structural reorganization may represent a viable window for potential therapeutic strategies aimed at restoring or maintaining hippocampal function during the transition from NCI to MCI to AD.
Figure. 6.
Golgi impregnated hippocampal CA1 pyramidal neurons analyzed for dendritic branching of the basilar tree from cases with a clinical diagnosis of (A) no cognitive impairment, (B) mild cognitive impairment and (C) Alzheimer's disease. (D) Note that amyloid deposit (*) distorts the dendritic architecture of a CA1 dendrite. (E) Scholl analysis of the amount, distribution and complexity of the arbor showed a significant increase in these parameters (length, + 18%; complexity, +23%) in MCI CA1 neurons compared to NCI. Conversely, there was a significant reduction in branch length (−39%) and arbor complexity (−25%) in the progression from MCI to AD. Adopted from with permission from [22]
In addition to structural alterations, there are biochemical neuroplastic responses, as well. For example, activity levels of choline acetyltransferease (ChAT), the rate-limiting synthetic enzyme for acetylcholine, are increased significantly in the hippocampus and superior frontal cortex in MCI compared to NCI or mild-moderate AD (Fig. 7A) [68] [69]. Across the three clinical diagnostic groups examined, increased hippocampal ChAT activity correlated with progression of hippocampal and entorhinal cortex NP pathology and limbic Braak stages [69], suggesting that this elevation is indeed a compensatory response to entorhinal-hippocampal disconnection (Fig. 7B, C). Further examination of the up-regulation of frontal cortical ChAT activity in MCI revealed that it was not paralleled by an increase in cholinergic fiber density in the same cortical region [76] suggesting that structural reorganization was not the mechanism activating this cholinergic plasticity response. It is more likely the result of a biochemical process that increases ChAT production within the cholinergic cortical projection neurons located within the nucleus basalis of Meynert [77], which is anterogradely transported to its cortical projection sites. Both the hippocampal and frontal cortex ChAT responses are transient [68], suggesting that this neuroplasticity is lost during the continued onslaught of the disease.
Figure 7.
(A) Choline acetyltransferase (ChAT) activity increased in the hippocampus in MCI and returned to control levels in mild AD. (B and C) Schematic drawings of coronal section of the hippocampus illustrating the loss of innervation arising from the glutamatergic layer II entorhinal cortex neurons (red) triggering a cholinergic plasticity response (blue) in the MCI hippocampus. Abbreviations: AD: Alzheimer's disease; CA1, CA2, CA3, CA4- Cornu Ammonis hippocampal subfields; CS- collateral sulcus, DG-dentate gyrus, Ent-entorhinal cortex; f-fornix, gl-granular cell layer, ml- molecular layer; NCI- no cognitive impairment, MCI-mild cognitive impairment, pp-perforant pathway, TEC- transentorhinal cortex, Sub-subiculum.
Another example of early neural reorganization is associated with entorhinal layer II glutamatergic neurons that innervate the hippocampus and degenerate early in the disease process [78]. It has been shown that alterations in glutamate receptor subunits precedes NFT formation in these two regions [79]. An investigation using specific immunological markers of glutamatergic neurons found a pathology-dependent pattern of glutamatergic synaptic remodeling within frontal cortex across the progressive phases of AD with MCI displaying an elevation in glutamatergic presynaptic bouton density, which depletes and is lost with disease progression [29]. Increased glutamatergic presynaptic bouton density correlated with improved cognition in AD, but not MCI, in which the increase in glutamatergic presynaptic bouton density was paradoxically related with decreased cognitive ability [29]. Perhaps this late chemical upregulation indicates a type of compensatory response intended to offset the effects of preexisting synaptotoxicity, or is an uncoordinated aberrant synaptic plasticity lacking functionality late in the disease. While neither interpretation has been confirmed experimentally, in light of the cholinergic hippocampal and frontal cortex reorganization findings and their association with better cognitive performance, the former seems more likely. Memantine, an FDA-approved partial antagonist of N-methyl-D-aspartate (NMDA) glutamate receptors, which acts to normalize glutamatergic neurotransmission has only modest symptomatic efficacy [80] [81]. New and more potent memantine-based drugs are needed to hopefully provide greater efficacy either alone or in combination with other compounds. In this regard, Namzaric, combines memantine hydrochloride extended-release (Namenda) and donepezil hydrochloride (Aricept) an anticholinersterase which inhibits degradation of acetylcholine at the synapse. The extent to which other ascending neurotransmitter systems are capable of neuroplasticity during the progression of dementia remains an active area of investigation and a potential new set of drug targets.
Apolipoprotein E (ApoE) allele status, cognition and pathology
The genetic risk factor for AD, ApoE ε4, may also play a prominent role in the development of cognitive decline and pathology in AD [82]. Cognitively normal ε4 carriers are more likely to have significant AD-related pathology relative to non-carriers, but rates of cognitive decline are not significantly different between genotypes [83]. ApoE gene status, which mediates AD-related neuropathology, affects the onset and trajectory of cognitive decline [84] [85]. ApoE ε4 carriers display significantly greater amyloid load determined by Pittsburgh compound-B (PiB) imaging and show significantly lower cognitive test scores compared to non-carriers in non-demented elderly subjects [85] suggesting that APOE ε4 modifies the harmful effect of Aβ on cognition [86]. We found that soluble Aβ1-42 and fibrillary β-amyloid protein levels within the precuneus measured by PiB binding were significantly higher in AD than MCI and NCI, but were not associated with APOE ε4 status [87] [88]. Neuropathological reports show that AD pathology is limited within the precuneus suggesting a disconnection between amyloid toxicity and histopathology in this brain region [89] [88]. Perhaps this component of the default memory network is resilient to amyloid pathology [88].
Molecular signature of prodromal AD
A major area of interest in AD, is defining the molecular and cellular substrates of dementia onset. To accomplish this it is imperative to sample gene and noncoding RNA (ncRNA) expression from homogeneous brain cell types and correlate these changes with antemortem cognitive measures. Our group developed a strategy for the evaluation of individual genes that are differentially regulated as well as characterizing relevant classes of transcripts by gene ontology (GO) grouping during dementia progression using custom-designed microarray analysis following laser capture microdissection of immunoshistochemically labelled neurons [90] [91]. For example, we observe a downregulation of transcripts during the progression of dementia in GO categories including glutamatergic neurotransmission, GABAergic neurotransmission, and synaptic-related markers [73, 90] [92] consistent with current hypotheses that synaptic degeneration, as well as deficits in excitatory and inhibitory neurotransmission underlie AD pathogenesis, which acting independent, or synergistically with amyloid deposits and NFTs [73, 92].
In contrast to observations of a gene downregulation in the majority of GO categories analyzed to date, significant upregulation of select endosomal-lysosomal pathway genes were found in CA1 pyramidal and cholinergic basal forebrain (CBF) neurons (Fig. 8). Quantitative analysis in CA1 pyramidal neurons indicated upregulation of early endosome effectors rab4 (AD>MCI & NCI) and rab5 (AD>MCI>NCI), late endosome constituent rab7 (AD & MCI>NCI), and the trafficking molecule rab24 (AD>MCI & NCI) [93]. Based on the comparison of expression level differences between MCI and AD, alterations in rab5 and rab7 were considered early, whereas rab4 and rab24 upregulation was considered a later alteration [93]. A strong negative correlation was found between cognitive scores and rab4, rab5, rab7, and rab24 CA1 neuron expression levels [93]. In addition, there was an upregulation of the acid hydrolase cathepsin D (Ctsd; AD & MCI>NCI), the early endosome antigen 1 (Eea1; AD & MCI>NCI), and lysosomal-associated membrane protein 1 (Lamp1; AD>MCI & NCI) [93]. In contrast, no differential regulation of rab GTPases that are not regulators of endosomes was observed across clinical groups [93]. Upregulated expression of rab4, rab5, rab7, and rab27 in CBF neurons correlated with antemortem measures of cognitive decline in MCI and AD [94]. Results were validated by sub-regional qPCR and immunoblot analysis [93] [94].
Figure 8.
(A) Heat map and histograms (B) showing changes in endosomal-lysosomal and nerve growth factor receptor gene levels during the progression of AD.
Recently, we found upregulation of select endosomal-lysosomal genes, consistent with our previous single label analyses of vulnerable CBF neurons [94] at different stages of NFT evolution [23]. Pretangle-bearing CBF neurons (marked with the tau antibody pS422) [95] displayed a significant upregulation rab5, rab7, Ctsd, and Lamp1 expression. Later stage NFT, visualized by dual-labeling immunohistochemistry using the pretangle, pS422 marker, and TauC3, a later stage tau neoepitope [96], was associated with rab4 upregulation [23]. No expression level changes were found for amyloid-beta precursor protein (APP), amyloid-beta precursor-like protein 1 (Aplp1), amyloid-beta precursor-like protein 2 (Aplp2), Bace1, presenilin 1 (Psen1), presenilin 2 (Psen2), low-density lipoprotein-related protein 1 (Lrp1), high-density lipoprotein binding protein (Hdlbp), alpha-2-macroglobulin (A2m), and beta-2-microglobulin (B2m), among other AD-related genes [23] [92] [94]. The lack of expression changes related to amyloid and presenilin genes in these highly vulnerable neurons suggests that these mRNAs are not playing a major role in cellular deregulation early in disease process. Since deficits in the endocytic system is one of the earliest disturbances in AD and precede amyloid and NFT deposition as well as clinical symptoms of AD [97], dysfunction in this gene family may underlie, in part, the early selective vulnerability of hippocampal CA1 pyramidal and CBF neurons. Future studies using RNA sequencing (RNA-seq) at the single population level during the progression of dementia will likely enable the assessment of endosomal-lysosomal and other pathways for coordinate analyses through GO queries, as well as the ncRNAs that potentially regulate their expression.
The presence of small non-coding microRNAs (miRNAs) that negatively regulate mRNA stability [98] is an under researched area in AD [99, 100]. Various miRNAs regulate a multitude of brain functions (e.g., synaptic plasticity and energy metabolism) [101-104] suggesting that perturbations in miRNA function is involved in the pathogenesis of complex neurodegenerative disorders including AD [99, 100]. AD brains display altered expression of miRNAs that regulate BACE1 enzyme [105, 106] and are linked to tau phosphorylation [107-109] and pro-inflammatory actions [107, 110-112]. The role that the dysregulation of brain miRNA networks play early in AD are unclear. Our group using microarray and quantitative PCR analyses compared levels of miRNAs isolated from frontal cortex (FC) and inferior temporal cortex (ITC) tissue obtained from cases who died with a clinical diagnosis of NCI, aMCI, or mild AD [113]. Two families of miRNAs, miR-212/132 and miR-23a/b were down regulated in the FC in aMCI and AD compared to NCI, but were preserved in the ITC. Down-regulation of either group of miRNAs was predicted to increase the deacetylase Sirtuin 1 (Sirt1), which is involved in the mediation of protective neuronal cell stress responses [114]. Sirt1 mRNA levels were greater in the aMCI FC but stable in ITC, suggesting a connection between miR-212/132 and miR-23a/b down-regulation and decreased transcriptional repression of Sirt1 expression. Experimental down-regulation of miR-212 and miR-23a in vitro up-regulated Sirt1 resulting in neuroprotection against Aβ toxicity. These data suggest that up-regulation of Sirt1 is a novel neuroprotective pathway activated early in the onset of AD.
Biomarkers during AD progression
Neuropathological examinations of older people who died with a clinical diagnosis of NCI or MCI often reveal similar pathological signatures to those with frank AD [40, 89, 115-118]. Hence, identification of individuals in the preclinical and prodromal stages of AD is essential for the timely administration of disease modifying therapies. Several initiatives have defined a panel of diagnostic and prognostic imaging as well as cerebrospinal fluid (CSF) and blood biomarkers that identify people at the earliest stages of the disease, with most fluid biomarker studies focusing detecting changes in CSF Aβ peptide and tau protein levels at varying AD stages [119]. The National Institute on Aging-Alzheimer's Association (NIA-AA) developed criteria for using biomarkers to determine the likelihood of AD pathology and for classifying patients accordingly [119, 120] which included CSF Aβ42, positron emission tomography (PET) amyloid, CSF total tau, pT181 phospho-tau, MTL atrophy on Magnetic Resonance Imaging (MRI), tempoparietal/precuneus hypometabolism or hypoperfusion on PET or single-photon emission computed tomography (SPECT) [119]. Findings indicate that loss of hippocampal volume and the ratio of CSF Aβ42 to total tau or phospho-tau are predictive of longitudinal changes in cognitive measures [121-124]. Arterial spin labeling MRI is used to examine the influence of changes in resting cerebral blood flow as well as blood oxygenation level dependent signal response in relation to PET-derived regional amyloid load [125, 126] or to memory encoding in the MTL [127]. It has become increasingly clear that imaging radioligands, alone or in combination with other AD biomarkers will be critical for the earliest detection of AD pathology and timely initiation of therapy.
(3H)PiB PET imaging in NCI, MCI and AD
PiB 6-OH-BTA-1; [128]2-(4’-methylaminophenyl)-6-hydroxybenzothiazole)] binds with a high affinity to β-sheet structured aggregates of fibrillar Aβ [129] and was the first PET imaging agent to differentiate AD from NCI (Fig. 9) [130]. It binds most prominently in NPs and vascular Aβ associated with cerebral amyloid angiopathy (CAA) [131], but does not bind to classic intracellular NFTs or other neuropathological elements [132]. In vitro [C-11]PiB binding across multiple brain regions distinguishes between clinical categories and correlates with fibrillar Aβ by enzyme-linked immunosorbent assay (ELISA) [133, 134], and correlates with antemortem [C-11]PiB PET retention [135]. Interestingly, 10%-20% of AD patients are PiB(−), in agreement with autopsy reports [136] [40], and 20-30% of NCI are also PiB(+)[137] , which increases up to 65% in those >80 years [40, 138], suggesting a clinical misdiagnosis and/or a distinct dementia syndrome. Interestingly, APOEε4 is associated with higher PiB PET retention in elderly NCI [139], while in MCI it confers an increased likelihood of converting to AD [140]. The presence of Aβ deposits in PiB negative (−) subjects questions the sensitivity of this tracer [135].
Figure 9.
Representative images of [C-11]PiB PET in subjects with NCI, MCI, or AD. Some NCI and MCI subjects have positive [C-11]PiB PET scans (NCI+, MCI+) indicating an ongoing process of brain amyloidosis. Image courtesy of the University of Pittsburgh Amyloid Imaging Group.
The relatively short radioactive half-life of carbon-11 (~20 minutes), limits the distribution of PiB to PET imaging centers and supports the requirement of longer lived radioligands such as F-18 flutemetamol a 3’-fluoro analog of PiB (3’-F-PiB) [141], F-18 florbetapir [142] and F-18 florbetaben [143]. PET ligands for amyloid detection will be useful tools for the differential clinical diagnosis in large populations of individuals with suspected cognitive decline. To better define clinical states the field also needs tools to image tau profiles.
Tau PET imaging
Although research efforts have been successful in developing PET tracers that specifically bind to fibrillar Aβ pathology, a positive amyloid PET scan alone may not be sufficient for a positive diagnosis for AD. Since tau pathology in form of NFT burden is a better correlate of AD dementia, a PET tracer that selectively targets tau aggregates maybe a more relevant biomarker for AD conversion and for other tauopathies.[144] In normal brain there is an equal ratio of 3-repeat (3R) and 4-repeat (4R) tau isoforms, but in pathological states, tauopathies can express different isoform ratios with diverse morphologies, making it more difficult to develop a disease-specific tau PET tracer with similar affinity for every phenotype.[144, 145] In addition, tau undergoes several posttranslational modifications, resulting in conformational changes and excessive phosphorylation in the aggregates, potentially resulting in different tau ligand binding affinities. Since tau aggregates co-occur with Aß deposits in AD, tau ligands need to be highly selective. During the past several years, several candidate tau PET imaging agents have been developed and used to image tau in different clinical phenotypes of AD (Fig. 10). [144, 146] Several groups reported tau-selective PET radioligands, including [F-18] labeled THK compounds [147] [148] [149-151] [152], PBB compounds [153] and [F-18] labeled T807 and T808 compounds [154] [155]. The latter compounds are the most widely used PET tracers for tau; they discriminated AD from control and non-AD dementias and demonstrated topographical patterns similar to neuropathological stages, suggesting their utility for assessing tau-related neuronal injury in vivo [150, 156, 157]. For example, in a study using 18F AV-1451 [155], patterns of cortical binding visualized with surface-projected SUVR thresholds were shown to be anatomically consistent with the ordinal stages of Braak pathology (0, I/II, III/IV, and V/VI) [38]. Similar to Braak stages 0, I, or II, many control cases displayed low overall or only mesial temporal lobe binding (Fig 10A–C), whereas greater neocortical binding was always associated with higher levels of temporal lobe binding in impaired cases (Fig 10D–G). The highest neocortical binding levels, consistent with Braak stages V-VI, were related with selective sparing of primary cortices (Fig 10F–G). These anatomic findings were confirmed quantitatively by voxel-wise and region of interest measures. 18FAV-1451 PET was found to be abnormally high in cortical, entorhinal, and parahippocampal regions (but not in the hippocampus) in MCI and AD compared to NCI, and greater radioligand retention in the inferior temporal gyrus correlated with impaired cognition [155]. Postmortem studies indicate that AV-1451 binds better to NFTs than Aβ plaques, but also identified off-target binding in some areas [158]. Other, less selective ligands also demonstrated good utility when compared to imaging of brain atrophy and glucose metabolism. For example, (F-18)FDDNP (2-(1-{6-[159]fluoroethyl) (methyl)amino]-2-naphthyl}ethylidene)malononitrile), a lipophilic tracer, which binds to both Aβ plaques and NFTs [160] revealed increases in regional brain uptake which correlated with greater brain atrophy on MRI and reduced FDG-PET [159]. Although (F-18)FDDNP PET and (3H)PiB PET have similar retention patterns in neocortical regions, (F-18)FDDNP retention was greater in the mesial temporal lobe [161]. Higher global cortical uptake of (F-18) FDDNP PET and (3H)PiB PET was seen in MCI and AD compared to NCI, however, (3H)PiB proved better in detecting differences among the clinical groups [162]. (F-18)AV-1451 also has a promising pattern of retention in relation to clinical severity of AD [163], with greater PET retention in MCI and AD patients compared to NCI [154].
Figure 10.
Cortical patterns of F-18T807 binding. Coronal 18F T807 positron emission tomographic (PET) images (top row) and whole-brain surface renderings of standardized uptake value ratio (SUVR; cerebellar reference; second row) from 3 clinically normal (CN) and 4 impaired (2 mild cognitive impairment [MCI] and 2 mild Alzheimer dementia [43] dementia) participants. Top: (A) A 71-year-old CN subject with low amyloid β (Aβ) by Pittsburgh compound B (PiB) PET (mean cortical distribution volume ratio [DVR]=1.0) had low, nonspecific 18F T807 binding in cortex, consistent with a Braak stage less than III/IV. (B) A 74-year-old CN subject with high Aβ (DVR=1.2) with 18F T807 binding in inferior temporal cortex, left>right, consistent with Braak stage III/IV. (C) A 79-year-old CN subject with high Aβ (DVR=1.8) had binding in inferior temporal neocortex, consistent with Braak stage of III/IV. B and C show focally intense subcortical uptake that is likely due to off-target binding. (D–G) Cognitively impaired participants all with high Aβ and with successively greater levels of cortical 18F T807 binding successively involving temporal, parietal, frontal, and occipital cortices. Bottom: 18F T807 SUVR calculated at vertices indicating the extent of cortical binding, with left hemisphere views (lateral, inferior, superior, medial) at left. The 52-year-old AD dementia patient (G) showed confluent 18F T807 binding that is nearly pancortical, sparing only portions of primary cortex and consistent with Braak stage V/VI. Dx=classification; MMSE=Mini-Mental State Examination; PET Braak=estimate of Braak stages based on the anatomic pattern of T807 binding assessed visually and quantitatively in regions and full volume data. Legend and figure are reproduced with permission from Annals of Neurology Volume 79, Issue 1, pages 110-119, 15 DEC 2015 DOI: 10.1002/ana.24546 http://onlinelibrary.wiley.com/doi/10.1002/ana.24546/full#ana24546-fig-0001.
Taken together amyloid and tau PET imaging represent major advances in AD field, however there are still a number of limitations and unresolved questions. Sensitivity of PET imaging with [C-11]PiB and related [F-18] amyloid ligands is not well characterized for relatively low but histologically detectable Aβ deposits [164]. Analyses of large numbers of PET positive and PET negative cases, with short imaging-to-autopsy interval, are required to resolve these issues and establish threshold levels of Aβ and tau pathologies necessary for in vivo PET detection. A major challenge for neuroimaging in AD is how to determine the onset of amyloid and tau accumulation in pathology burdened NCI and MCI, and its association with cognitive measures and other biomarkers. Accordingly, the focus of PET imaging studies is currently shifting from AD towards NCI with the earliest fibrillar Aβ and tau deposits, to help determine the clinical significance of pre-symptomatic pathology and identify subjects at risk for cognitive decline. More studies are needed to directly compare the relative merit of amyloid and tau PET, MRI, FDG, clinical measures and CSF biomarkers.
CSF Biomarkers
Imaging-based technologies are relatively expensive and not readily transferred to community-based clinical settings, so fluid biomarkers are still desirable should the level of sensitivity and specificity for predicting conversion to dementia remain high. Panels of plasma inflammatory marker and related proteins [165], either alone or in combination with CSF biomarkers, have been proposed. CSF concentrations of Aβ1-42 and tau are indirect measures of AD pathology in the brain, with relatively high accuracy for identifying incipient AD [122] and predicting the development and rate of cognitive decline [123] [166]. In AD, CSF Aβ1-42 is reduced and phosphorylated tau is increased, which correlates with post-mortem amyloid and NFT pathology [167]. A strong concordance between PiB PET and CSF Aβ1-42 exists in NCI [168], and mixed cohorts of NCI and AD [169] [170], with no correlation between PiB PET and CSF tau [171]. Longitudinal studies suggested that amyloid PET may be more sensitive than CSF Aβ1-42 in identifying MCI cases that will convert to AD [172] [173]. A plasma phospholipid panel was recently identified that predicted conversion to MCI or AD within a 2-3 year timeframe with over 90% accuracy [174]. However, the inherent variability of AD neuropathology in the aged NCI, MCI and AD brain is also reflected by variation in these markers, particularly CSF and blood amyloid and tau [175], resulting in equivocal specificity and sensitivity for identifying at-risk individuals. Figure 11 shows a summary of data related to CSF biomarker diagnostic performance across a large number of primary studies published after the introduction of criteria recommended by the NIA-Alzheimer's Association Workgroups [176] showing the sensitivity, specificity, and likelihood ratios of CSF core biomarkers [177], and head-to-head CSF biomarker performance based on average AD to control ratios [178]. Together in combination with clinical methods, cognitive tests and imaging, CSF biomarkers can contribute to the assessment of dementia and whether AD pathology is present in cognitively normal individuals [179]. Currently, there is a need to augment current CSF biomarker panels with novel proteins reflecting AD molecular pathogenesis to improve diagnostic accuracy in longitudinal studies assuming that AD is a multifactorial disease [180].
Figure 11.
Summary of CSF biomarker diagnostic performance. A) Sensitivity, specificity, and likelihood ratios of CSF core biomarkers based on primary studies published after the introduction of criteria recommended by the NIA-Alzheimer's Association Workgroups* [176]. B) Head-to-head CSF biomarker performance based on average AD to control ratios. Biomarkers are differentiated based on significant differences with good effect sizes (purple), significant differences with moderate effect sizes (purple), or non-significant or significant differences with minor effect sizes.** *Reprinted from Frontiers in Aging Neuroscience, Ferreira et al., Meta-review of CSF core biomarkers in Alzheimer's disease: the state-of-the-art after the new revised diagnostic criteria, pp. 1-24, 2014 (Open Access). [177]. *Reprinted from The Lancet Neurology, Olsson et al., CSF and blood biomarkers for the diagnosis of Alzheimer's disease: a systematic review and meta-analysis, doi:10.1016/S1474-4422(16)00070-3 2016, with permission from Elsevier. [189]
We discovered that protein levels of the precursor of nerve growth factor (NGF), termed proNGF, were selectively increased in postmortem neocortex [181, 182] and hippocampus [72, 183] of subjects who died with a clinical diagnosis of MCI or mild AD compared to NCI, respectively, which correlated with poorer performance on antemortem cognitive tests [181, 183]. Therefore, we tested whether CSF proNGF levels are an early marker of cognitive impairment using ventricular CSF (vCSF) obtained postmortem from the RROS cohort who came to autopsy with a clinical diagnosis of NCI, aMCI, or mild/moderate AD, and lumbar CSF collected from Washington University Knight AD Research Center subjects clinically diagnosed CDR 0 (no dementia), CDR 0.5 (MCI or very mild AD), or CDR 1 (mild AD) [184]. Quantitative western blotting of vCSF revealed a significant 55% increase in CSF proNGF levels obtained from aMCI compared to NCI and a 70% increase in AD compared to NCI, which showed a significant inverse association between increasing vCSF proNGF levels, cognitive deterioration and GCS [184]. Similarly, lumbar CSF samples revealed a significant 30% increase in proNGF levels in the CDR 0.5 and CDR 1 compared to CDR 0 cases. However, there were no differences in levels of Aβ1-42, total tau, phospho-tau, or phospho-tau/ Aβ1-42 among the groups, whereas the ratio of total tau/Aβ1-42 levels was 50% higher in CDR 1 subjects compared to CDR 0. To determine whether the inclusion of proNGF would improve the reliability of these biomarkers, we calculated proNGF/Aβ1-42, proNGF/total tau, and proNGF/phospho-tau ratios. Notably, proNGF/Aβ1-42 levels were 50% higher in CDR 0.5 and CDR 1 compared to CDR 0, whereas proNGF/total tau and proNGF/phospho-tau were not different between the groups. Our data suggest adding proNGF to a growing list of candidate biomarkers for AD. Increased CSF proNGF levels likely mirror the up regulation of this mainly proapoptotic protein seen in the neocortex and hippocampus in MCI and AD subjects, which we have suggested indicates a shift from cell survival to death [26]. Hence, we posit that increased CSF proNGF is an early pathobiological marker for disease onset. In sum, combining single or multiple tracer imaging using PET radioligands with CSF biomarkers will allow for earlier detection of AD, monitoring of pathology progression and effects of therapies, and selection of appropriate subjects for clinical trials of future AD therapies.
Concluding comments
Based on accumulating molecular, cellular, biochemical, neuroplasticity, neuroimaging, and clinical pathological findings, perhaps the field of AD research has reached a point where there is a need for a paradigm shift away from a strict interpretation of the amyloid hypothesis as it relates to the biological cascade of events that culminate in dementia. In his classic book “The Structure of Scientific Revolutions” the philosopher, Thomas Kuhn, proposed that intellectual progress is not gradual but is marked by sudden paradigm shifts [185]. The argument is made that there is a period when the field follows a paradigm that has great promise such as the amyloid cascade hypothesis. However, during the last few years this hypothesis has been challenged by clinical pathological investigations demonstrating a large population of older individuals that display extensive pathological hallmarks of this disease, amyloid and tau lesions but do not develop dementia even in the eighth and ninth decades of life [22] suggesting that these pathologies alone may not be necessary or sufficient to initiate cognitive decline [186]. Whether these lesions are primary drivers of pathology, or pathological bystanders remains to be determined. The reviewed molecular, cellular, neuroimaging, and clinical pathological findings as well as the poor efficacy of amyloid vaccine clinical trials to date suggests that the disease process is more complicated than a singular hypothesis. We posit that it is time to move away from concentrating on a unitary bio-mechanistic trigger and subsequent mono-therapeutic approach to a more inclusive molecular mechanistic concept that involves not only amyloid and tau but the disconnection of vulnerable neuronal circuits related to attention, memory, and executive functions as well as the brain's neuroplasticity early in the disease process. This paradigm shift may lead to more diverse and multidrug treatment approaches for this most intransient neurological disease of aging.
Highlights.
Review of Pathophysiology of prodromal Alzheimer's Disease (AD)
Novel Biomarkers for AD: Imaging and CSF
Brain plasticity in prodromal AD
Review of literature of the neurobiology of Prodromal AD suggests a rethinking of amyloid hypothesis
Acknowledgements
This study was supported by NIH grants PO1AG14449, RO1AG043375, P30AG010161, PO1AG107617, R21AG026032, R21AG042146, and Barrow Neurological Institute Barrow and Beyond.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Blocq P, Marinesco G. Sur les lésions et la pathogénie de l'épilepsie dite essentielle. Sem Med. 12:445–446. [Google Scholar]
- 2.Zigman WB, Schupf N, Devenny DA, Miezejeski C, Ryan R, Urv TK, Schubert R, Silverman W. Incidence and prevalence of dementia in elderly adults with mental retardation without down syndrome. American journal of mental retardation : AJMR. 2004;109(2):126–41. doi: 10.1352/0895-8017(2004)109<126:IAPODI>2.0.CO;2. [DOI] [PubMed] [Google Scholar]
- 3.Oliver C, Holland AJ. Down's syndrome and Alzheimer's disease: a review. Psychol Med. 1986;16(2):307–22. doi: 10.1017/s0033291700009120. [DOI] [PubMed] [Google Scholar]
- 4.Thinakaran G, Koo EH. Amyloid precursor protein trafficking, processing, and function. J Biol Chem. 2008;283(44):29615–9. doi: 10.1074/jbc.R800019200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Shoji M, Golde TE, Ghiso J, Cheung TT, Estus S, Shaffer LM, Cai XD, McKay DM, Tintner R, Frangione B, et al. Production of the Alzheimer amyloid beta protein by normal proteolytic processing. Science. 1992;258(5079):126–9. doi: 10.1126/science.1439760. [DOI] [PubMed] [Google Scholar]
- 6.Trojanowski JQ, Schmidt ML, Shin RW, Bramblett GT, Rao D, Lee VM. Altered tau and neurofilament proteins in neuro-degenerative diseases: diagnostic implications for Alzheimer's disease and Lewy body dementias. Brain Pathol. 1993;3(1):45–54. doi: 10.1111/j.1750-3639.1993.tb00725.x. [DOI] [PubMed] [Google Scholar]
- 7.Yoshiyama Y, Lee VM, Trojanowski JQ. Therapeutic strategies for tau mediated neurodegeneration. J Neurol Neurosurg Psychiatry. 2013;84(7):784–95. doi: 10.1136/jnnp-2012-303144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hardy J. The amyloid hypothesis for Alzheimer's disease: a critical reappraisal. J Neurochem. 2009;110(4):1129–34. doi: 10.1111/j.1471-4159.2009.06181.x. [DOI] [PubMed] [Google Scholar]
- 9.Muller U, Winter P, Graeber MB. A presenilin 1 mutation in the first case of Alzheimer's disease. Lancet Neurol. 2013;12(2):129–30. doi: 10.1016/S1474-4422(12)70307-1. [DOI] [PubMed] [Google Scholar]
- 10.Rupp C, Beyreuther K, Maurer K, Kins S. A presenilin 1 mutation in the first case of Alzheimer's disease: revisited. Alzheimers Dement. 2014;10(6):869–72. doi: 10.1016/j.jalz.2014.06.005. [DOI] [PubMed] [Google Scholar]
- 11.Hardy JA, Higgins GA. Alzheimer's disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184–5. doi: 10.1126/science.1566067. [DOI] [PubMed] [Google Scholar]
- 12.Graeber MB, Kosel S, Egensperger R, Banati RB, Muller U, Bise K, Hoff P, Moller HJ, Fujisawa K, Mehraein P. Rediscovery of the case described by Alois Alzheimer in 1911: historical, histological and molecular genetic analysis. Neurogenetics. 1997;1(1):73–80. doi: 10.1007/s100480050011. [DOI] [PubMed] [Google Scholar]
- 13.Moller HJ, Graeber MB. The case described by Alois Alzheimer in 1911. Historical and conceptual perspectives based on the clinical record and neurohistological sections. Eur Arch Psychiatry Clin Neurosci. 1998;248(3):111–22. doi: 10.1007/s004060050027. [DOI] [PubMed] [Google Scholar]
- 14.Mesulam MM. Neuroplasticity failure in Alzheimer's disease: bridging the gap between plaques and tangles. Neuron. 1999;24(3):521–9. doi: 10.1016/s0896-6273(00)81109-5. [DOI] [PubMed] [Google Scholar]
- 15.Hardy J. Testing times for the “amyloid cascade hypothesis”. Neurobiol Aging. 2002;23(6):1073–4. doi: 10.1016/s0197-4580(02)00042-8. [DOI] [PubMed] [Google Scholar]
- 16.Sperling RA, Johnson KA, Doraiswamy PM, Reiman EM, Fleisher AS, Sabbagh MN, Sadowsky CH, Carpenter A, Davis MD, Lu M, Flitter M, Joshi AD, Clark CM, Grundman M, Mintun MA, Skovronsky DM, Pontecorvo MJ, Group AAS. Amyloid deposition detected with florbetapir F 18 ((18)F-AV-45) is related to lower episodic memory performance in clinically normal older individuals. Neurobiol Aging. 2013;34(3):822–31. doi: 10.1016/j.neurobiolaging.2012.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sperling RA, Karlawish J, Johnson KA. Preclinical Alzheimer disease-the challenges ahead. Nature reviews. Neurology. 2013;9(1):54–8. doi: 10.1038/nrneurol.2012.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E. Mild cognitive impairment: clinical characterization and outcome. Arch Neurol. 1999;56(3):303–8. doi: 10.1001/archneur.56.3.303. [DOI] [PubMed] [Google Scholar]
- 19.Petersen R. Conceptual Overview. In: Petersen R, editor. Mild cognitive impairment Aging to Alzheimer's disease. Oxford University Press; New York: 2003. [Google Scholar]
- 20.Gamblin TC, Chen F, Zambrano A, Abraha A, Lagalwar S, Guillozet AL, Lu M, Fu Y, Garcia-Sierra F, LaPointe N, Miller R, Berry RW, Binder LI, Cryns VL. Caspase cleavage of tau: linking amyloid and neurofibrillary tangles in Alzheimer's disease. Proc Natl Acad Sci U S A. 2003;100(17):10032–7. doi: 10.1073/pnas.1630428100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Markesbery WR. Neuropathologic alterations in mild cognitive impairment: a review. J Alzheimers Dis. 2010;19(1):221–8. doi: 10.3233/JAD-2010-1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mufson E, Malek-Ahmadi M, Perez SE, Chen a.K. Braak staging, plaque pathology and APOE status in elderly persons without cognitive impairment. Neurobiol Aging. 2016;37:147–153. doi: 10.1016/j.neurobiolaging.2015.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tiernan CT, Ginsberg SD, Guillozet-Bongaarts AL, Ward SM, He B, Kanaan NM, Mufson EJ, Binder LI, Counts SE. Protein homeostasis gene dysregulation in pretangle bearing nucleus basalis neurons during the progression of Alzheimer's disease. Neurobiol Aging. 2016 doi: 10.1016/j.neurobiolaging.2016.02.031. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Jicha GA, Abner EL, Schmitt FA, Kryscio RJ, Riley KP, Cooper GE, Stiles N, Mendiondo MS, Smith CD, Van Eldik LJ, Nelson PT. Preclinical AD Workgroup staging: pathological correlates and potential challenges. Neurobiol Aging. 2012;33(3):622, e1–622, e16. doi: 10.1016/j.neurobiolaging.2011.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.DeKosky ST, Scheff SW, Styren SD. Structural correlates of cognition in dementia: quantification and assessment of synapse change. Neurodegeneration : a journal for neurodegenerative disorders, neuroprotection, and neuroregeneration. 1996;5(4):417–21. doi: 10.1006/neur.1996.0056. [DOI] [PubMed] [Google Scholar]
- 26.Mufson EJ, Binder L, Counts SE, DeKosky ST, de Toledo-Morrell L, Ginsberg SD, Ikonomovic MD, Perez SE, Scheff SW. Mild cognitive impairment: pathology and mechanisms. Acta Neuropathol. 2012;123(1):13–30. doi: 10.1007/s00401-011-0884-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hampel H, Schneider LS, Giacobini E, Kivipelto M, Sindi S, Dubois B, Broich K, Nistico R, Aisen PS, Lista S. Advances in the therapy of Alzheimer's disease: targeting amyloid beta and tau and perspectives for the future. Expert review of neurotherapeutics. 2015;15(1):83–105. doi: 10.1586/14737175.2015.995637. [DOI] [PubMed] [Google Scholar]
- 28.Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO molecular medicine. 2016 doi: 10.15252/emmm.201606210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bell KF, Ducatenzeiler A, Ribeiro-da-Silva A, Duff K, Bennett DA, Cuello AC. The amyloid pathology progresses in a neurotransmitter-specific manner. Neurobiol Aging. 2006;27(11):1644–57. doi: 10.1016/j.neurobiolaging.2005.09.034. [DOI] [PubMed] [Google Scholar]
- 30.Dowling NM, Tomaszewski Farias S, Reed BR, Sonnen JA, Strauss ME, Schneider JA, Bennett DA, Mungas D. Neuropathological associates of multiple cognitive functions in two community-based cohorts of older adults. Journal of the International Neuropsychological Society : JINS. 2011;17(4):602–14. doi: 10.1017/S1355617710001426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Thal DR, Sassin I, Schultz C, Haass C, Braak E, Braak H. Fleecy amyloid deposits in the internal layers of the human entorhinal cortex are comprised of N-terminal truncated fragments of Abeta. J Neuropathol Exp Neurol. 1999;58(2):210–6. doi: 10.1097/00005072-199902000-00010. [DOI] [PubMed] [Google Scholar]
- 32.Braak H, Braak E. Demonstration of amyloid deposits and neurofibrillary changes in whole brain sections. Brain Pathol. 1991;1(3):213–6. doi: 10.1111/j.1750-3639.1991.tb00661.x. [DOI] [PubMed] [Google Scholar]
- 33.Binder LI, Frankfurter A, Rebhun LI. The distribution of tau in the mammalian central nervous system. J Cell Biol. 1985;101(4):1371–8. doi: 10.1083/jcb.101.4.1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Braak H, Braak E. Neurofibrillary changes confined to the entorhinal region and an abundance of cortical amyloid in cases of presenile and senile dementia. Acta Neuropathol. 1990;80(5):479–86. doi: 10.1007/BF00294607. [DOI] [PubMed] [Google Scholar]
- 35.Garcia-Sierra F, Ghoshal N, Quinn B, Berry RW, Binder LI. Conformational changes and truncation of tau protein during tangle evolution in Alzheimer's disease. J Alzheimers Dis. 2003;5(2):65–77. doi: 10.3233/jad-2003-5201. [DOI] [PubMed] [Google Scholar]
- 36.Mufson EJ, Malek-Ahmadi M, Perez SE, Chen K. Braak staging, plaque pathology, and APOE status in elderly persons without cognitive impairment. Neurobiol Aging. 2016;37:147–53. doi: 10.1016/j.neurobiolaging.2015.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Thal DR, Rub U, Orantes M, Braak H. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58(12):1791–800. doi: 10.1212/wnl.58.12.1791. [DOI] [PubMed] [Google Scholar]
- 38.Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–59. doi: 10.1007/BF00308809. [DOI] [PubMed] [Google Scholar]
- 39.Kawas C, Gray S, Brookmeyer R, Fozard J, Zonderman A. Age-specific incidence rates of Alzheimer's disease: the Baltimore Longitudinal Study of Aging. Neurology. 2000;54(11):2072–7. doi: 10.1212/wnl.54.11.2072. [DOI] [PubMed] [Google Scholar]
- 40.Price JL, Morris JC. Tangles and plaques in nondemented aging and “preclinical” Alzheimer's disease. Ann Neurol. 1999;45(3):358–68. doi: 10.1002/1531-8249(199903)45:3<358::aid-ana12>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
- 41.Gold G, Bouras C, Kovari E, Canuto A, Glaria BG, Malky A, Hof PR, Michel JP, Giannakopoulos P. Clinical validity of Braak neuropathological staging in the oldest-old. Acta Neuropathol. 2000;99(5):579–82. doi: 10.1007/s004010051163. discussion 583-4. [DOI] [PubMed] [Google Scholar]
- 42.Bennett DA, Schneider JA, Arvanitakis Z, Kelly JF, Aggarwal NT, Shah RC, Wilson RS. Neuropathology of older persons without cognitive impairment from two community-based studies. Neurology. 2006;66(12):1837–44. doi: 10.1212/01.wnl.0000219668.47116.e6. [DOI] [PubMed] [Google Scholar]
- 43.Consensus recommendations for the postmortem diagnosis of Alzheimer's disease. The National Institute on Aging, and Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer's Disease. Neurobiol Aging. 1997;18(4 Suppl):S1–2. [PubMed] [Google Scholar]
- 44.Erten-Lyons D, Woltjer RL, Dodge H, Nixon R, Vorobik R, Calvert JF, Leahy M, Montine T, Kaye J. Factors associated with resistance to dementia despite high Alzheimer disease pathology. Neurology. 2009;72(4):354–60. doi: 10.1212/01.wnl.0000341273.18141.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Counts SE, Che S, Ginsberg SD, Mufson EJ. Gender differences in neurotrophin and glutamate receptor expression in cholinergic nucleus basalis neurons during the progression of Alzheimer's disease. J Chem Neuroanat. 2011;42(2):111–7. doi: 10.1016/j.jchemneu.2011.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Mazure CM, Swendsen J. Sex differences in Alzheimer's disease and other dementias. Lancet Neurol. 2016;15(5):451–2. doi: 10.1016/S1474-4422(16)00067-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Mufson EJ, Chen EY, Cochran EJ, Beckett LA, Bennett DA, Kordower JH. Entorhinal cortex beta-amyloid load in individuals with mild cognitive impairment. Exp Neurol. 1999;158(2):469–90. doi: 10.1006/exnr.1999.7086. [DOI] [PubMed] [Google Scholar]
- 48.Bancher C, Jellinger K, Lassmann H, Fischer P, Leblhuber F. Correlations between mental state and quantitative neuropathology in the Vienna Longitudinal Study on Dementia. Eur Arch Psychiatry Clin Neurosci. 1996;246(3):137–46. doi: 10.1007/BF02189115. [DOI] [PubMed] [Google Scholar]
- 49.Nelson PT, Jicha GA, Schmitt FA, Liu H, Davis DG, Mendiondo MS, Abner EL, Markesbery WR. Clinicopathologic correlations in a large Alzheimer disease center autopsy cohort: neuritic plaques and neurofibrillary tangles “do count” when staging disease severity. J Neuropathol Exp Neurol. 2007;66(12):1136–46. doi: 10.1097/nen.0b013e31815c5efb. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Malek-Ahmadi M, Perez SE, Chen K, Mufson EJ. Neuritic and Diffuse Plaque Associations with Memory in Non-Cognitively Impaired Elders submitted. doi: 10.3233/JAD-160365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wilson RS, Leurgans SE, Boyle PA, Bennett DA. Cognitive decline in prodromal Alzheimer disease and mild cognitive impairment. Arch Neurol. 2011;68(3):351–6. doi: 10.1001/archneurol.2011.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Scheff SW, Ansari MA, Mufson EJ. Relationship between oxidative stress and hippocampal synaptic proteins: evidence for prodromal Alzheimer's disease. Neurobiology of aging. 2016 doi: 10.1016/j.neurobiolaging.2016.02.030. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ansari MA, Scheff SW. Oxidative stress in the progression of Alzheimer disease in the frontal cortex. J Neuropathol Exp Neurol. 2010;69(2):155–67. doi: 10.1097/NEN.0b013e3181cb5af4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Keller JN, Schmitt FA, Scheff SW, Ding Q, Chen Q, Butterfield DA, Markesbery WR. Evidence of increased oxidative damage in subjects with mild cognitive impairment. Neurology. 2005;64(7):1152–6. doi: 10.1212/01.WNL.0000156156.13641.BA. [DOI] [PubMed] [Google Scholar]
- 55.Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, Atwood CS, Petersen RB, Smith MA. Oxidative damage is the earliest event in Alzheimer disease. Journal of neuropathology and experimental neurology. 2001;60(8):759–67. doi: 10.1093/jnen/60.8.759. [DOI] [PubMed] [Google Scholar]
- 56.Pratico D, Sung S. Lipid peroxidation and oxidative imbalance: early functional events in Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2004;6(2):171–5. doi: 10.3233/jad-2004-6209. [DOI] [PubMed] [Google Scholar]
- 57.Walsh DM, Selkoe DJ. A beta oligomers - a decade of discovery. J Neurochem. 2007;101(5):1172–84. doi: 10.1111/j.1471-4159.2006.04426.x. [DOI] [PubMed] [Google Scholar]
- 58.Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, Gallagher M, Ashe KH. A specific amyloid-beta protein assembly in the brain impairs memory. Nature. 2006;440(7082):352–7. doi: 10.1038/nature04533. [DOI] [PubMed] [Google Scholar]
- 59.Bilousova T, Miller CA, Poon WW, Vinters HV, Corrada M, Kawas C, Hayden EY, Teplow DB, Glabe C, Albay R, 3rd, Cole GM, Teng E, Gylys KH. Synaptic Amyloid-beta Oligomers Precede p-Tau and Differentiate High Pathology Control Cases. Am J Pathol. 2016;186(1):185–98. doi: 10.1016/j.ajpath.2015.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Morris JC. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology. 1993;43(11):2412–4. doi: 10.1212/wnl.43.11.2412-a. [DOI] [PubMed] [Google Scholar]
- 61.Pham E, Crews L, Ubhi K, Hansen L, Adame A, Cartier A, Salmon D, Galasko D, Michael S, Savas JN, Yates JR, Glabe C, Masliah E. Progressive accumulation of amyloid-beta oligomers in Alzheimer's disease and in amyloid precursor protein transgenic mice is accompanied by selective alterations in synaptic scaffold proteins. The FEBS journal. 2010;277(14):3051–67. doi: 10.1111/j.1742-4658.2010.07719.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Jin M, Shepardson N, Yang T, Chen G, Walsh D, Selkoe DJ. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A. 2011;108(14):5819–24. doi: 10.1073/pnas.1017033108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Walker LC, Jucker M. Neurodegenerative diseases: expanding the prion concept. Annual review of neuroscience. 2015;38:87–103. doi: 10.1146/annurev-neuro-071714-033828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Perez-Nievas BG, Stein TD, Tai HC, Dols-Icardo O, Scotton TC, Barroeta-Espar I, Fernandez-Carballo L, de Munain EL, Perez J, Marquie M, Serrano-Pozo A, Frosch MP, Lowe V, Parisi JE, Petersen RC, Ikonomovic MD, Lopez OL, Klunk W, Hyman BT, Gomez-Isla T. Dissecting phenotypic traits linked to human resilience to Alzheimer's pathology. Brain. 2013;136(Pt 8):2510–26. doi: 10.1093/brain/awt171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Negash S, Wilson RS, Leurgans SE, Wolk DA, Schneider JA, Buchman AS, Bennett DA, Arnold SE. Resilient brain aging: characterization of discordance between Alzheimer's disease pathology and cognition. Curr Alzheimer Res. 2013;10(8):844–51. doi: 10.2174/15672050113109990157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Stern Y. Cognitive reserve. Neuropsychologia. 2009;47(10):2015–28. doi: 10.1016/j.neuropsychologia.2009.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Arnold SE, Louneva N, Cao K, Wang LS, Han LY, Wolk DA, Negash S, Leurgans SE, Schneider JA, Buchman AS, Wilson RS, Bennett DA. Cellular, synaptic, and biochemical features of resilient cognition in Alzheimer's disease. Neurobiol Aging. 2013;34(1):157–68. doi: 10.1016/j.neurobiolaging.2012.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.DeKosky ST, Ikonomovic MD, Styren SD, Beckett L, Wisniewski S, Bennett DA, Cochran EJ, Kordower JH, Mufson EJ. Upregulation of choline acetyltransferase activity in hippocampus and frontal cortex of elderly subjects with mild cognitive impairment. Ann Neurol. 2002;51(2):145–55. doi: 10.1002/ana.10069. [DOI] [PubMed] [Google Scholar]
- 69.Ikonomovic MD, Mufson EJ, Wuu J, Cochran EJ, Bennett DA, DeKosky ST. Cholinergic plasticity in hippocampus of individuals with mild cognitive impairment: correlation with Alzheimer's neuropathology. J Alzheimers Dis. 2003;5(1):39–48. doi: 10.3233/jad-2003-5106. [DOI] [PubMed] [Google Scholar]
- 70.Scheff SW, Price DA, Schmitt FA, DeKosky ST, Mufson EJ. Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology. 2007;68(18):1501–8. doi: 10.1212/01.wnl.0000260698.46517.8f. [DOI] [PubMed] [Google Scholar]
- 71.Counts SE, He B, Nadeem M, Wuu J, Scheff SW, Mufson EJ. Hippocampal drebrin loss in mild cognitive impairment. Neuro-degenerative diseases. 2012;10(1-4):216–9. doi: 10.1159/000333122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Mufson EJ, He B, Nadeem M, Perez SE, Counts SE, Leurgans S, Fritz J, Lah J, Ginsberg SD, Wuu J, Scheff SW. Hippocampal ProNGF Signaling Pathways and beta-Amyloid Levels in Mild Cognitive Impairment and Alzheimer Disease. J Neuropathol Exp Neurol. 2012;71(11):1018–1029. doi: 10.1097/NEN.0b013e318272caab. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mufson EJ, Mahady L, Waters D, Counts SE, Perez SE, DeKosky ST, Ginsberg SD, Ikonomovic MD, Scheff SW, Binder LI. Hippocampal plasticity during the progression of Alzheimer's disease. Neuroscience. 2015;309:51–67. doi: 10.1016/j.neuroscience.2015.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Riudavets MA, Iacono D, Resnick SM, O'Brien R, Zonderman AB, Martin LJ, Rudow G, Pletnikova O, Troncoso JC. Resistance to Alzheimer's pathology is associated with nuclear hypertrophy in neurons. Neurobiol Aging. 2007;28(10):1484–92. doi: 10.1016/j.neurobiolaging.2007.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.DeKosky ST, Scheff SW. Synapse loss in frontal cortex biopsies in Alzheimer's disease: correlation with cognitive severity. Ann Neurol. 1990;27(5):457–64. doi: 10.1002/ana.410270502. [DOI] [PubMed] [Google Scholar]
- 76.Ikonomovic MD, Abrahamson EE, Isanski BA, Wuu J, Mufson EJ, DeKosky ST. Superior frontal cortex cholinergic axon density in mild cognitive impairment and early Alzheimer disease. Arch Neurol. 2007;64(9):1312–7. doi: 10.1001/archneur.64.9.1312. [DOI] [PubMed] [Google Scholar]
- 77.Mesulam MM, Mufson EJ, Wainer BH, Levey AI. Central cholinergic pathways in the rat: an overview based on an alternative nomenclature (Ch1-Ch6) Neuroscience. 1983;10(4):1185–201. doi: 10.1016/0306-4522(83)90108-2. [DOI] [PubMed] [Google Scholar]
- 78.Gomez-Isla T, Price JL, McKeel DW, Jr., Morris JC, Growdon JH, Hyman BT. Profound loss of layer II entorhinal cortex neurons occurs in very mild Alzheimer's disease. J Neurosci. 1996;16(14):4491–500. doi: 10.1523/JNEUROSCI.16-14-04491.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ikonomovic MD, Mizukami K, Davies P, Hamilton R, Sheffield R, Armstrong DM. The loss of GluR2(3) immunoreactivity precedes neurofibrillary tangle formation in the entorhinal cortex and hippocampus of Alzheimer brains. J Neuropathol Exp Neurol. 1997;56(9):1018–27. doi: 10.1097/00005072-199709000-00007. [DOI] [PubMed] [Google Scholar]
- 80.Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius HJ. A 24-week open-label extension study of memantine in moderate to severe Alzheimer disease. Arch Neurol. 2006;63(1):49–54. doi: 10.1001/archneur.63.1.49. [DOI] [PubMed] [Google Scholar]
- 81.Danysz W, Parsons CG. Alzheimer's disease, beta-amyloid, glutamate, NMDA receptors and memantine--searching for the connections. Br J Pharmacol. 2012;167(2):324–52. doi: 10.1111/j.1476-5381.2012.02057.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Roses AD, Saunders AM. APOE is a major susceptibility gene for Alzheimer's disease. Current opinion in biotechnology. 1994;5(6):663–7. doi: 10.1016/0958-1669(94)90091-4. [DOI] [PubMed] [Google Scholar]
- 83.Vos SJ, van Rossum IA, Verhey F, Knol DL, Soininen H, Wahlund LO, Hampel H, Tsolaki M, Minthon L, Frisoni GB, Froelich L, Nobili F, van der Flier W, Blennow K, Wolz R, Scheltens P, Visser PJ. Prediction of Alzheimer disease in subjects with amnestic and nonamnestic MCI. Neurology. 2013;80(12):1124–32. doi: 10.1212/WNL.0b013e318288690c. [DOI] [PubMed] [Google Scholar]
- 84.Caselli RJ, Dueck AC, Osborne D, Sabbagh MN, Connor DJ, Ahern GL, Baxter LC, Rapcsak SZ, Shi J, Woodruff BK, Locke DE, Snyder CH, Alexander GE, Rademakers R, Reiman EM. Longitudinal modeling of age-related memory decline and the APOE epsilon4 effect. N Engl J Med. 2009;361(3):255–63. doi: 10.1056/NEJMoa0809437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Reiman EM, Chen K, Liu X, Bandy D, Yu M, Lee W, Ayutyanont N, Keppler J, Reeder SA, Langbaum JB, Alexander GE, Klunk WE, Mathis CA, Price JC, Aizenstein HJ, DeKosky ST, Caselli RJ. Fibrillar amyloid-beta burden in cognitively normal people at 3 levels of genetic risk for Alzheimer's disease. Proc Natl Acad Sci U S A. 2009;106(16):6820–5. doi: 10.1073/pnas.0900345106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kantarci K, Yang C, Schneider JA, Senjem ML, Reyes DA, Lowe VJ, Barnes LL, Aggarwal NT, Bennett DA, Smith GE, Petersen RC, Jack CR, Jr., Boeve BF. Antemortem amyloid imaging and beta-amyloid pathology in a case with dementia with Lewy bodies. Neurobiol Aging. 2012;33(5):878–85. doi: 10.1016/j.neurobiolaging.2010.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ikonomovic MD, Klunk WE, Abrahamson EE, Wuu J, Mathis CA, Scheff SW, Mufson EJ, DeKosky ST. Precuneus amyloid burden is associated with reduced cholinergic activity in Alzheimer disease. Neurology. 2011;77(1):39–47. doi: 10.1212/WNL.0b013e3182231419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Perez SE, He B, Nadeem M, Wuu J, Scheff SW, Abrahamson EE, Ikonomovic MD, Mufson EJ. Resilience of precuneus neurotrophic signaling pathways despite amyloid pathology in prodromal Alzheimer's disease. Biol Psychiatry. 2015;77(8):693–703. doi: 10.1016/j.biopsych.2013.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Nelson PT, Braak H, Markesbery WR. Neuropathology and cognitive impairment in Alzheimer disease: a complex but coherent relationship. J Neuropathol Exp Neurol. 2009;68(1):1–14. doi: 10.1097/NEN.0b013e3181919a48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ginsberg SD, Alldred MJ, Che S. Gene expression levels assessed by CA1 pyramidal neuron and regional hippocampal dissections in Alzheimer's disease. Neurobiol Dis. 2012;45:99–107. doi: 10.1016/j.nbd.2011.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Alldred MJ, Lee SH, Petkova E, Ginsberg SD. Expression profile analysis of hippocampal CA1 pyramidal neurons in aged Ts65Dn mice, a model of Down syndrome (DS) and Alzheimer's disease (AD) Brain structure & function. 2015;220:2983–2996. doi: 10.1007/s00429-014-0839-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Counts SE, Alldred MJ, Che S, Ginsberg SD, Mufson EJ. Synaptic gene dysregulation within hippocampal CA1 pyramidal neurons in mild cognitive impairment. Neuropharmacology. 2014;79:172–9. doi: 10.1016/j.neuropharm.2013.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ginsberg SD, Mufson EJ, Counts SE, Wuu J, Alldred MJ, Nixon RA, Che S. Regional selectivity of rab5 and rab7 protein upregulation in mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 2010;22(2):631–9. doi: 10.3233/JAD-2010-101080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ginsberg SD, Mufson EJ, Alldred MJ, Counts SE, Wuu J, Nixon RA, Che S. Upregulation of select rab GTPases in cholinergic basal forebrain neurons in mild cognitive impairment and Alzheimer's disease. J Chem Neuroanat. 2011;42(2):102–10. doi: 10.1016/j.jchemneu.2011.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Vana L, Kanaan NM, Ugwu IC, Wuu J, Mufson EJ, Binder LI. Progression of tau pathology in cholinergic Basal forebrain neurons in mild cognitive impairment and Alzheimer's disease. Am J Pathol. 2011;179(5):2533–50. doi: 10.1016/j.ajpath.2011.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Guillozet-Bongaarts AL, Cahill ME, Cryns VL, Reynolds MR, Berry RW, Binder LI. Pseudophosphorylation of tau at serine 422 inhibits caspase cleavage: in vitro evidence and implications for tangle formation in vivo. J Neurochem. 2006;97(4):1005–14. doi: 10.1111/j.1471-4159.2006.03784.x. [DOI] [PubMed] [Google Scholar]
- 97.Cataldo AM, Petanceska S, Terio NB, Peterhoff CM, Durham R, Mercken M, Mehta PD, Buxbaum J, Haroutunian V, Nixon RA. Abeta localization in abnormal endosomes: association with earliest Abeta elevations in AD and Down syndrome. Neurobiol Aging. 2004;25(10):1263–72. doi: 10.1016/j.neurobiolaging.2004.02.027. [DOI] [PubMed] [Google Scholar]
- 98.Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294(5543):853–8. doi: 10.1126/science.1064921. [DOI] [PubMed] [Google Scholar]
- 99.Nelson PT, Wang WX, Rajeev BW. MicroRNAs (miRNAs) in neurodegenerative diseases. Brain Pathol. 2008;18(1):130–8. doi: 10.1111/j.1750-3639.2007.00120.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hebert SS, De Strooper B. Alterations of the microRNA network cause neurodegenerative disease. Trends Neurosci. 2009;32(4):199–206. doi: 10.1016/j.tins.2008.12.003. [DOI] [PubMed] [Google Scholar]
- 101.Aschrafi A, Schwechter AD, Mameza MG, Natera-Naranjo O, Gioio AE, Kaplan BB. MicroRNA-338 regulates local cytochrome c oxidase IV mRNA levels and oxidative phosphorylation in the axons of sympathetic neurons. J Neurosci. 2008;28(47):12581–90. doi: 10.1523/JNEUROSCI.3338-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Fineberg SK, Kosik KS, Davidson BL. MicroRNAs potentiate neural development. Neuron. 2009;64(3):303–9. doi: 10.1016/j.neuron.2009.10.020. [DOI] [PubMed] [Google Scholar]
- 103.Rajasethupathy P, Fiumara F, Sheridan R, Betel D, Puthanveettil SV, Russo JJ, Sander C, Tuschl T, Kandel E. Characterization of small RNAs in Aplysia reveals a role for miR-124 in constraining synaptic plasticity through CREB. Neuron. 2009;63(6):803–17. doi: 10.1016/j.neuron.2009.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Schratt GM, Tuebing F, Nigh EA, Kane CG, Sabatini ME, Kiebler M, Greenberg ME. A brain-specific microRNA regulates dendritic spine development. Nature. 2006;439(7074):283–9. doi: 10.1038/nature04367. [DOI] [PubMed] [Google Scholar]
- 105.Wang WX, Rajeev BW, Stromberg AJ, Ren N, Tang G, Huang Q, Rigoutsos I, Nelson PT. The expression of microRNA miR-107 decreases early in Alzheimer's disease and may accelerate disease progression through regulation of beta-site amyloid precursor protein-cleaving enzyme 1. J Neurosci. 2008;28(5):1213–23. doi: 10.1523/JNEUROSCI.5065-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Hebert SS, Horre K, Nicolai L, Papadopoulou AS, Mandemakers W, Silahtaroglu AN, Kauppinen S, Delacourte A, De Strooper B. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer's disease correlates with increased BACE1/beta-secretase expression. Proc Natl Acad Sci U S A. 2008;105(17):6415–20. doi: 10.1073/pnas.0710263105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Absalon S, Kochanek DM, Raghavan V, Krichevsky AM. MiR-26b, upregulated in Alzheimer's disease, activates cell cycle entry, tau-phosphorylation, and apoptosis in postmitotic neurons. J Neurosci. 2013;33(37):14645–59. doi: 10.1523/JNEUROSCI.1327-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hebert SS, Papadopoulou AS, Smith P, Galas MC, Planel E, Silahtaroglu AN, Sergeant N, Buee L, De Strooper B. Genetic ablation of Dicer in adult forebrain neurons results in abnormal tau hyperphosphorylation and neurodegeneration. Human molecular genetics. 2010;19(20):3959–69. doi: 10.1093/hmg/ddq311. [DOI] [PubMed] [Google Scholar]
- 109.Banzhaf-Strathmann J, Benito E, May S, Arzberger T, Tahirovic S, Kretzschmar H, Fischer A, Edbauer D. MicroRNA-125b induces tau hyperphosphorylation and cognitive deficits in Alzheimer's disease. The EMBO journal. 2014;33(15):1667–80. doi: 10.15252/embj.201387576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Cui JG, Li YY, Zhao Y, Bhattacharjee S, Lukiw WJ. Differential regulation of interleukin-1 receptor-associated kinase-1 (IRAK-1) and IRAK-2 by microRNA-146a and NF-kappaB in stressed human astroglial cells and in Alzheimer disease. J Biol Chem. 2010;285(50):38951–60. doi: 10.1074/jbc.M110.178848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li YY, Alexandrov PN, Pogue AI, Zhao Y, Bhattacharjee S, Lukiw WJ. miRNA-155 upregulation and complement factor H deficits in Down's syndrome. Neuroreport. 2012;23(3):168–73. doi: 10.1097/WNR.0b013e32834f4eb4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lukiw WJ, Cui JG, Yuan LY, Bhattacharjee PS, Corkern M, Clement C, Kammerman EM, Ball MJ, Zhao Y, Sullivan PM, Hill JM. Acyclovir or Abeta42 peptides attenuate HSV-1-induced miRNA-146a levels in human primary brain cells. Neuroreport. 2010;21(14):922–7. doi: 10.1097/WNR.0b013e32833da51a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Weinberg RB, Mufson EJ, Counts SE. Evidence for a neuroprotective microRNA pathway in amnestic mild cognitive impairment. Frontiers in neuroscience. 2016 doi: 10.3389/fnins.2015.00430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bonda DJ, Lee HG, Camins A, Pallas M, Casadesus G, Smith MA, Zhu X. The sirtuin pathway in ageing and Alzheimer disease: mechanistic and therapeutic considerations. Lancet Neurol. 10(3):275–9. doi: 10.1016/S1474-4422(11)70013-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Morris JC, Storandt M, Miller JP, McKeel DW, Price JL, Rubin EH, Berg L. Mild cognitive impairment represents early-stage Alzheimer disease. Arch Neurol. 2001;58(3):397–405. doi: 10.1001/archneur.58.3.397. [DOI] [PubMed] [Google Scholar]
- 116.Bennett DA, Schneider JA, Bienias JL, Evans DA, Wilson RS. Mild cognitive impairment is related to Alzheimer disease pathology and cerebral infarctions. Neurology. 2005;64(5):834–41. doi: 10.1212/01.WNL.0000152982.47274.9E. [DOI] [PubMed] [Google Scholar]
- 117.Markesbery WR, Schmitt FA, Kryscio RJ, Davis DG, Smith CD, Wekstein DR. Neuropathologic substrate of mild cognitive impairment. Arch Neurol. 2006;63(1):38–46. doi: 10.1001/archneur.63.1.38. [DOI] [PubMed] [Google Scholar]
- 118.Mufson EJ, Chen EY, Cochran EJ, Beckett LA, Bennett DA, Kordower JH. Entorhinal cortex beta-amyloid load in individuals with mild cognitive impairment. Exp Neurol. 1999;158(2):469–90. doi: 10.1006/exnr.1999.7086. [DOI] [PubMed] [Google Scholar]
- 119.Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Cedarbaum J, Green RC, Harvey D, Jack CR, Jagust W, Luthman J, Morris JC, Petersen RC, Saykin AJ, Shaw L, Shen L, Schwarz A, Toga AW, Trojanowski JQ, I. Alzheimer's Disease Neuroimaging 2014 Update of the Alzheimer's Disease Neuroimaging Initiative: A review of papers published since its inception. Alzheimers Dement. 2015;11(6):e1–120. doi: 10.1016/j.jalz.2014.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Galluzzi S, Marizzoni M, Babiloni C, Albani D, Antelmi L, Bagnoli C, Bartres-Faz D, Cordone S, Didic M, Farotti L, Fiedler U, Forloni G, Girtler N, Hensch T, Jovicich J, Leeuwis A, Marra C, Molinuevo JL, Nobili F, Pariente J, Parnetti L, Payoux P, Del Percio C, Ranjeva JP, Rolandi E, Rossini PM, Schonknecht P, Soricelli A, Tsolaki M, Visser PJ, Wiltfang J, Richardson JC, Bordet R, Blin O, Frisoni GB, PharmaCog C. Clinical and biomarker profiling of prodromal Alzheimer's disease in workpackage 5 of the Innovative Medicines Initiative PharmaCog project: a ‘Eu`ropean ADNI study’. Journal of internal medicine. 2016 doi: 10.1111/joim.12482. [DOI] [PubMed] [Google Scholar]
- 121.Fagan AM, Roe CM, Xiong C, Mintun MA, Morris JC, Holtzman DM. Cerebrospinal fluid tau/beta-amyloid(42) ratio as a prediction of cognitive decline in nondemented older adults. Arch Neurol. 2007;64(3):343–9. doi: 10.1001/archneur.64.3.noc60123. [DOI] [PubMed] [Google Scholar]
- 122.Mattsson N, Zetterberg H, Hansson O, Andreasen N, Parnetti L, Jonsson M, Herukka SK, van der Flier WM, Blankenstein MA, Ewers M, Rich K, Kaiser E, Verbeek M, Tsolaki M, Mulugeta E, Rosen E, Aarsland D, Visser PJ, Schroder J, Marcusson J, de Leon M, Hampel H, Scheltens P, Pirttila T, Wallin A, Jonhagen ME, Minthon L, Winblad B, Blennow K. CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairment. Jama. 2009;302(4):385–93. doi: 10.1001/jama.2009.1064. [DOI] [PubMed] [Google Scholar]
- 123.Snider BJ, Fagan AM, Roe C, Shah AR, Grant EA, Xiong C, Morris JC, Holtzman DM. Cerebrospinal fluid biomarkers and rate of cognitive decline in very mild dementia of the Alzheimer type. Arch Neurol. 2009;66(5):638–45. doi: 10.1001/archneurol.2009.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Trojanowski JQ, Vandeerstichele H, Korecka M, Clark CM, Aisen PS, Petersen RC, Blennow K, Soares H, Simon A, Lewczuk P, Dean R, Siemers E, Potter WZ, Weiner MW, Jack CR, Jr., Jagust W, Toga AW, Lee VM, Shaw LM. Update on the biomarker core of the Alzheimer's Disease Neuroimaging Initiative subjects. Alzheimers Dement. 2010;6(3):230–8. doi: 10.1016/j.jalz.2010.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Mattsson N, Tosun D, Insel PS, Simonson A, Jack CR, Jr., Beckett LA, Donohue M, Jagust W, Schuff N, Weiner MW, Alzheimer's Disease Neuroimaging I. Association of brain amyloid-beta with cerebral perfusion and structure in Alzheimer's disease and mild cognitive impairment. Brain. 2014;137(Pt 5):1550–61. doi: 10.1093/brain/awu043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Tosun D, Joshi S, Weiner MW, I. the Alzheimer's Disease Neuroimaging Multimodal MRI-based Imputation of the Abeta+ in Early Mild Cognitive Impairment. Annals of clinical and translational neurology. 2014;1(3):160–170. doi: 10.1002/acn3.40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Bangen KJ, Restom K, Liu TT, Wierenga CE, Jak AJ, Salmon DP, Bondi MW. Assessment of Alzheimer's disease risk with functional magnetic resonance imaging: an arterial spin labeling study. J Alzheimers Dis. 2012;31(Suppl 3):S59–74. doi: 10.3233/JAD-2012-120292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Law MP, Osman S, Davenport RJ, Cunningham VJ, Pike VW, Camici PG. Biodistribution and metabolism of [N-methyl-11C]m-hydroxyephedrine in the rat. Nuclear medicine and biology. 1997;24(5):417–24. doi: 10.1016/s0969-8051(97)00007-3. [DOI] [PubMed] [Google Scholar]
- 129.Levine H., 3rd Soluble multimeric Alzheimer beta(1-40) pre-amyloid complexes in dilute solution. Neurobiol Aging. 1995;16(5):755–64. doi: 10.1016/0197-4580(95)00052-g. [DOI] [PubMed] [Google Scholar]
- 130.Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP, Bergstrom M, Savitcheva I, Huang GF, Estrada S, Ausen B, Debnath ML, Barletta J, Price JC, Sandell J, Lopresti BJ, Wall A, Koivisto P, Antoni G, Mathis CA, Langstrom B. Imaging brain amyloid in Alzheimer's disease with Pittsburgh Compound-B. Ann Neurol. 2004;55(3):306–19. doi: 10.1002/ana.20009. [DOI] [PubMed] [Google Scholar]
- 131.Bacskai BJ, Frosch MP, Freeman SH, Raymond SB, Augustinack JC, Johnson KA, Irizarry MC, Klunk WE, Mathis CA, Dekosky ST, Greenberg SM, Hyman BT, Growdon JH. Molecular imaging with Pittsburgh Compound B confirmed at autopsy: a case report. Arch Neurol. 2007;64(3):431–4. doi: 10.1001/archneur.64.3.431. [DOI] [PubMed] [Google Scholar]
- 132.Ikonomovic MD, Klunk WE, Abrahamson EE, Mathis CA, Price JC, Tsopelas ND, Lopresti BJ, Ziolko S, Bi W, Paljug WR, Debnath ML, Hope CE, Isanski BA, Hamilton RL, DeKosky ST. Post-mortem correlates of in vivo PiB-PET amyloid imaging in a typical case of Alzheimer's disease. Brain. 2008;131(Pt 6):1630–45. doi: 10.1093/brain/awn016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Niedowicz DM, Beckett TL, Matveev S, Weidner AM, Baig I, Kryscio RJ, Mendiondo MS, LeVine H, 3rd, Keller JN, Murphy MP. Pittsburgh compound B and the postmortem diagnosis of Alzheimer disease. Ann Neurol. 2012;72(4):564–70. doi: 10.1002/ana.23633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Beckett TL, Webb RL, Niedowicz DM, Holler CJ, Matveev S, Baig I, LeVine H, 3rd, Keller JN, Murphy MP. Postmortem Pittsburgh Compound B (PiB) binding increases with Alzheimer's disease progression. J Alzheimers Dis. 2012;32(1):127–38. doi: 10.3233/JAD-2012-120655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Ikonomovic MD, Abrahamson EE, Price JC, Hamilton RL, Mathis CA, Paljug WR, Debnath ML, Cohen AD, Mizukami K, DeKosky ST, Lopez OL, Klunk WE. Early AD pathology in a [C-11]PiB-negative case: a PiB-amyloid imaging, biochemical, and immunohistochemical study. Acta Neuropathol. 2012;123(3):433–47. doi: 10.1007/s00401-012-0943-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Haroutunian V, Perl DP, Purohit DP, Marin D, Khan K, Lantz M, Davis KL, Mohs RC. Regional distribution of neuritic plaques in the nondemented elderly and subjects with very mild Alzheimer disease. Arch Neurol. 1998;55(9):1185–91. doi: 10.1001/archneur.55.9.1185. [DOI] [PubMed] [Google Scholar]
- 137.Aizenstein HJ, Nebes RD, Saxton JA, Price JC, Mathis CA, Tsopelas ND, Ziolko SK, James JA, Snitz BE, Houck PR, Bi W, Cohen AD, Lopresti BJ, DeKosky ST, Halligan EM, Klunk WE. Frequent amyloid deposition without significant cognitive impairment among the elderly. Arch Neurol. 2008;65(11):1509–17. doi: 10.1001/archneur.65.11.1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Mathis CA, Kuller LH, Klunk WE, Snitz BE, Price JC, Weissfeld LA, Rosario BL, Lopresti BJ, Saxton JA, Aizenstein HJ, McDade EM, Kamboh MI, DeKosky ST, Lopez OL. In vivo assessment of amyloid-beta deposition in nondemented very elderly subjects. Ann Neurol. 2013;73(6):751–61. doi: 10.1002/ana.23797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Morris JC, Roe CM, Xiong C, Fagan AM, Goate AM, Holtzman DM, Mintun MA. APOE predicts amyloid-beta but not tau Alzheimer pathology in cognitively normal aging. Ann Neurol. 2010;67(1):122–31. doi: 10.1002/ana.21843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Rowe CC, Ng S, Ackermann U, Gong SJ, Pike K, Savage G, Cowie TF, Dickinson KL, Maruff P, Darby D, Smith C, Woodward M, Merory J, Tochon-Danguy H, O'Keefe G, Klunk WE, Mathis CA, Price JC, Masters CL, Villemagne VL. Imaging beta-amyloid burden in aging and dementia. Neurology. 2007;68(20):1718–25. doi: 10.1212/01.wnl.0000261919.22630.ea. [DOI] [PubMed] [Google Scholar]
- 141.Curtis C, Gamez JE, Singh U, Sadowsky CH, Villena T, Sabbagh MN, Beach TG, Duara R, Fleisher AS, Frey KA, Walker Z, Hunjan A, Holmes C, Escovar YM, Vera CX, Agronin ME, Ross J, Bozoki A, Akinola M, Shi J, Vandenberghe R, Ikonomovic MD, Sherwin PF, Grachev ID, Farrar G, Smith AP, Buckley CJ, McLain R, Salloway S. Phase 3 trial of flutemetamol labeled with radioactive fluorine 18 imaging and neuritic plaque density. JAMA neurology. 2015;72(3):287–94. doi: 10.1001/jamaneurol.2014.4144. [DOI] [PubMed] [Google Scholar]
- 142.Fleisher AS, Chen K, Liu X, Roontiva A, Thiyyagura P, Ayutyanont N, Joshi AD, Clark CM, Mintun MA, Pontecorvo MJ, Doraiswamy PM, Johnson KA, Skovronsky DM, Reiman EM. Using positron emission tomography and florbetapir F18 to image cortical amyloid in patients with mild cognitive impairment or dementia due to Alzheimer disease. Arch Neurol. 2011;68(11):1404–11. doi: 10.1001/archneurol.2011.150. [DOI] [PubMed] [Google Scholar]
- 143.Richards D, Sabbagh MN. Florbetaben for PET Imaging of Beta-Amyloid Plaques in the Brain. Neurol Ther. 2014;3(2):79–88. doi: 10.1007/s40120-014-0022-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Declercq L, Celen S, Lecina J, Ahamed M, Tousseyn T, Moechars D, Alcazar J, Ariza M, Fierens K, Bottelbergs A, Marien J, Vandenberghe R, Andres IJ, Van Laere K, Verbruggen A, Bormans G. Comparison of New Tau PET-Tracer Candidates With [18F]T808 and [18F]T807. Molecular imaging. 2016;15 doi: 10.1177/1536012115624920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Villemagne VL, Okamura N. Tau imaging in the study of ageing, Alzheimer's disease, and other neurodegenerative conditions. Curr Opin Neurobiol. 2016;36:43–51. doi: 10.1016/j.conb.2015.09.002. [DOI] [PubMed] [Google Scholar]
- 146.Abatzoglou JT, Rupp DE, Mote PW. Questionable evidence of natural warming of the northwestern United States. Proc Natl Acad Sci U S A. 2014;111(52):5605–6. doi: 10.1073/pnas.1421311112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Harada R, Okamura N, Furumoto S, Tago T, Maruyama M, Higuchi M, Yoshikawa T, Arai H, Iwata R, Kudo Y, Yanai K. Comparison of the binding characteristics of [18F]THK-523 and other amyloid imaging tracers to Alzheimer's disease pathology. European journal of nuclear medicine and molecular imaging. 2013;40(1):125–32. doi: 10.1007/s00259-012-2261-2. [DOI] [PubMed] [Google Scholar]
- 148.Villemagne VL, Furumoto S, Fodero-Tavoletti MT, Mulligan RS, Hodges J, Harada R, Yates P, Piguet O, Pejoska S, Dore V, Yanai K, Masters CL, Kudo Y, Rowe CC, Okamura N. In vivo evaluation of a novel tau imaging tracer for Alzheimer's disease. European journal of nuclear medicine and molecular imaging. 2014;41(5):816–26. doi: 10.1007/s00259-013-2681-7. [DOI] [PubMed] [Google Scholar]
- 149.Lemoine L, Saint-Aubert L, Marutle A, Antoni G, Eriksson JP, Ghetti B, Okamura N, Nennesmo I, Gillberg PG, Nordberg A. Visualization of regional tau deposits using (3)H-THK5117 in Alzheimer brain tissue. Acta Neuropathol Commun. 2015;3:40. doi: 10.1186/s40478-015-0220-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Chiotis K, Saint-Aubert L, Savitcheva I, Jelic V, Andersen P, Jonasson M, Eriksson J, Lubberink M, Almkvist O, Wall A, Antoni G, Nordberg A. Imaging in-vivo tau pathology in Alzheimer's disease with THK5317 PET in a multimodal paradigm. European journal of nuclear medicine and molecular imaging. 2016 doi: 10.1007/s00259-016-3363-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Okamura N, Harada R, Furumoto S, Arai H, Yanai K, Kudo Y. Tau PET imaging in Alzheimer's disease. Current neurology and neuroscience reports. 2014;14(11):500. doi: 10.1007/s11910-014-0500-6. [DOI] [PubMed] [Google Scholar]
- 152.Okamura N, Furumoto S, Harada R, Tago T, Yoshikawa T, Fodero-Tavoletti M, Mulligan RS, Villemagne VL, Akatsu H, Yamamoto T, Arai H, Iwata R, Yanai K, Kudo Y. Novel 18F-labeled arylquinoline derivatives for noninvasive imaging of tau pathology in Alzheimer disease. J Nucl Med. 2013;54(8):1420–7. doi: 10.2967/jnumed.112.117341. [DOI] [PubMed] [Google Scholar]
- 153.Maruyama M, Shimada H, Suhara T, Shinotoh H, Ji B, Maeda J, Zhang MR, Trojanowski JQ, Lee VM, Ono M, Masamoto K, Takano H, Sahara N, Iwata N, Okamura N, Furumoto S, Kudo Y, Chang Q, Saido TC, Takashima A, Lewis J, Jang MK, Aoki I, Ito H, Higuchi M. Imaging of tau pathology in a tauopathy mouse model and in Alzheimer patients compared to normal controls. Neuron. 2013;79(6):1094–108. doi: 10.1016/j.neuron.2013.07.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Chien DT, Bahri S, Szardenings AK, Walsh JC, Mu F, Su MY, Shankle WR, Elizarov A, Kolb HC. Early clinical PET imaging results with the novel PHF-tau radioligand [F-18]-T807. J Alzheimers Dis. 2013;34(2):457–68. doi: 10.3233/JAD-122059. [DOI] [PubMed] [Google Scholar]
- 155.Johnson KA, Schultz A, Betensky RA, Becker JA, Sepulcre J, Rentz D, Mormino E, Chhatwal J, Amariglio R, Papp K, Marshall G, Albers M, Mauro S, Pepin L, Alverio J, Judge K, Philiossaint M, Shoup T, Yokell D, Dickerson B, Gomez-Isla T, Hyman B, Vasdev N, Sperling R. Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann Neurol. 2016;79(1):110–9. doi: 10.1002/ana.24546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Ossenkoppele R, Schonhaut DR, Scholl M, Lockhart SN, Ayakta N, Baker SL, O'Neil JP, Janabi M, Lazaris A, Cantwell A, Vogel J, Santos M, Miller ZA, Bettcher BM, Vossel KA, Kramer JH, Gorno-Tempini ML, Miller BL, Jagust WJ, Rabinovici GD. Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer's disease. Brain. 2016 doi: 10.1093/brain/aww027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Schwarz AJ, Yu P, Miller BB, Shcherbinin S, Dickson J, Navitsky M, Joshi AD, Devous MD, Sr., Mintun MS. Regional profiles of the candidate tau PET ligand 18F-AV-1451 recapitulate key features of Braak histopathological stages. Brain. 2016 doi: 10.1093/brain/aww023. [DOI] [PubMed] [Google Scholar]
- 158.Marquie M, Normandin MD, Vanderburg CR, Costantino IM, Bien EA, Rycyna LG, Klunk WE, Mathis CA, Ikonomovic MD, Debnath ML, Vasdev N, Dickerson BC, Gomperts SN, Growdon JH, Johnson KA, Frosch MP, Hyman BT, Gomez-Isla T. Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann Neurol. 2015;78(5):787–800. doi: 10.1002/ana.24517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Small GW, Kepe V, Ercoli LM, Siddarth P, Bookheimer SY, Miller KJ, Lavretsky H, Burggren AC, Cole GM, Vinters HV, Thompson PM, Huang SC, Satyamurthy N, Phelps ME, Barrio JR. PET of brain amyloid and tau in mild cognitive impairment. N Engl J Med. 2006;355(25):2652–63. doi: 10.1056/NEJMoa054625. [DOI] [PubMed] [Google Scholar]
- 160.Agdeppa ED, Kepe V, Liu J, Flores-Torres S, Satyamurthy N, Petric A, Cole GM, Small GW, Huang SC, Barrio JR. Binding characteristics of radiofluorinated 6-dialkylamino-2-naphthylethylidene derivatives as positron emission tomography imaging probes for beta-amyloid plaques in Alzheimer's disease. J Neurosci. 2001;21(24):RC189. doi: 10.1523/JNEUROSCI.21-24-j0004.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Shin J, Lee SY, Kim SH, Kim YB, Cho SJ. Multitracer PET imaging of amyloid plaques and neurofibrillary tangles in Alzheimer's disease. Neuroimage. 2008;43(2):236–44. doi: 10.1016/j.neuroimage.2008.07.022. [DOI] [PubMed] [Google Scholar]
- 162.Tolboom N, van der Flier WM, Yaqub M, Koene T, Boellaard R, Windhorst AD, Scheltens P, Lammertsma AA, van Berckel BN. Differential association of [11C]PIB and [18F]FDDNP binding with cognitive impairment. Neurology. 2009;73(24):2079–85. doi: 10.1212/WNL.0b013e3181c679cc. [DOI] [PubMed] [Google Scholar]
- 163.Xia CF, Arteaga J, Chen G, Gangadharmath U, Gomez LF, Kasi D, Lam C, Liang Q, Liu C, Mocharla VP, Mu F, Sinha A, Su H, Szardenings AK, Walsh JC, Wang E, Yu C, Zhang W, Zhao T, Kolb HC. [(18)F]T807, a novel tau positron emission tomography imaging agent for Alzheimer's disease. Alzheimers Dement. 2013;9(6):666–76. doi: 10.1016/j.jalz.2012.11.008. [DOI] [PubMed] [Google Scholar]
- 164.Ikonomovic MD, Abrahamson EE, Price JC, Hamilton RL, Mathis CA, Paljug WR, Debnath ML, Cohen AD, Mizukami K, DeKosky ST, Lopez OL, Klunk WE. Early AD pathology in a [C-11]PiB-negative case: a PiB-amyloid imaging, biochemical, and immunohistochemical study. Acta Neuropathol. 2012;123(3):433–47. doi: 10.1007/s00401-012-0943-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Leung R, Proitsi P, Simmons A, Lunnon K, Guntert A, Kronenberg D, Pritchard M, Tsolaki M, Mecocci P, Kloszewska I, Vellas B, Soininen H, Wahlund LO, Lovestone S. Inflammatory proteins in plasma are associated with severity of Alzheimer's disease. PLoS One. 2013;8(6):e64971. doi: 10.1371/journal.pone.0064971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Buchhave P, Minthon L, Zetterberg H, Wallin AK, Blennow K, Hansson O. Cerebrospinal fluid levels of beta-amyloid 1-42, but not of tau, are fully changed already 5 to 10 years before the onset of Alzheimer dementia. Arch Gen Psychiatry. 2012;69(1):98–106. doi: 10.1001/archgenpsychiatry.2011.155. [DOI] [PubMed] [Google Scholar]
- 167.Strozyk D, Blennow K, White LR, Launer LJ. CSF Abeta 42 levels correlate with amyloidneuropathology in a population-based autopsy study. Neurology. 2003;60(4):652–6. doi: 10.1212/01.wnl.0000046581.81650.d0. [DOI] [PubMed] [Google Scholar]
- 168.Fagan AM, Mintun MA, Shah AR, Aldea P, Roe CM, Mach RH, Marcus D, Morris JC, Holtzman DM. Cerebrospinal fluid tau and ptau(181) increase with cortical amyloid deposition in cognitively normal individuals: implications for future clinical trials of Alzheimer's disease. EMBO molecular medicine. 2009;1(8-9):371–80. doi: 10.1002/emmm.200900048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Hinrichs AL, Mintun MA, Head D, Fagan AM, Holtzman DM, Morris JC, Goate AM. Cortical binding of pittsburgh compound B, an endophenotype for genetic studies of Alzheimer's disease. Biol Psychiatry. 2010;67(6):581–3. doi: 10.1016/j.biopsych.2009.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Grimmer T, Riemenschneider M, Forstl H, Henriksen G, Klunk WE, Mathis CA, Shiga T, Wester HJ, Kurz A, Drzezga A. Beta amyloid in Alzheimer's disease: increased deposition in brain is reflected in reduced concentration in cerebrospinal fluid. Biol Psychiatry. 2009;65(11):927–34. doi: 10.1016/j.biopsych.2009.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Fagan AM, Mintun MA, Mach RH, Lee SY, Dence CS, Shah AR, LaRossa GN, Spinner ML, Klunk WE, Mathis CA, DeKosky ST, Morris JC, Holtzman DM. Inverse relation between in vivo amyloid imaging load and cerebrospinal fluid Abeta42 in humans. Ann Neurol. 2006;59(3):512–9. doi: 10.1002/ana.20730. [DOI] [PubMed] [Google Scholar]
- 172.Forsberg A, Engler H, Almkvist O, Blomquist G, Hagman G, Wall A, Ringheim A, Langstrom B, Nordberg A. PET imaging of amyloid deposition in patients with mild cognitive impairment. Neurobiol Aging. 2008;29(10):1456–65. doi: 10.1016/j.neurobiolaging.2007.03.029. [DOI] [PubMed] [Google Scholar]
- 173.Koivunen J, Verkkoniemi A, Aalto S, Paetau A, Ahonen JP, Viitanen M, Nagren K, Rokka J, Haaparanta M, Kalimo H, Rinne JO. PET amyloid ligand [11C]PIB uptake shows predominantly striatal increase in variant Alzheimer's disease. Brain. 2008;131(Pt 7):1845–53. doi: 10.1093/brain/awn107. [DOI] [PubMed] [Google Scholar]
- 174.Mapstone M, Cheema AK, Fiandaca MS, Zhong X, Mhyre TR, MacArthur LH, Hall WJ, Fisher SG, Peterson DR, Haley JM, Nazar MD, Rich SA, Berlau DJ, Peltz CB, Tan MT, Kawas CH, Federoff HJ. Plasma phospholipids identify antecedent memory impairment in older adults. Nat Med. 2014;20(4):415–8. doi: 10.1038/nm.3466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Humpel C. Identifying and validating biomarkers for Alzheimer's disease. Trends in biotechnology. 2011;29(1):26–32. doi: 10.1016/j.tibtech.2010.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Jr., Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, Mohs RC, Morris JC, Rossor MN, Scheltens P, Carrillo MC, Thies B, Weintraub S, Phelps CH. The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 2011;7(3):263–9. doi: 10.1016/j.jalz.2011.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Ferreira D, Perestelo-Perez L, Westman E, Wahlund LO, Sarria A, Serrano-Aguilar P. Meta-Review of CSF Core Biomarkers in Alzheimer's Disease: The State-of-the-Art after the New Revised Diagnostic Criteria. Frontiers in aging neuroscience. 2014;6:47. doi: 10.3389/fnagi.2014.00047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Firbank MJ, Lloyd J, Williams D, Barber R, Colloby SJ, Barnett N, Olsen K, Davison C, Donaldson C, Herholz K, O'Brien JT. An evidence-based algorithm for the utility of FDG-PET for diagnosing Alzheimer's disease according to presence of medial temporal lobe atrophy. The British journal of psychiatry : the journal of mental science. 2016;208(5):491–6. doi: 10.1192/bjp.bp.114.160804. [DOI] [PubMed] [Google Scholar]
- 179.Holtzman DM. CSF biomarkers for Alzheimer's disease: current utility and potential future use. Neurobiol Aging. 2011;32(Suppl 1):S4–9. doi: 10.1016/j.neurobiolaging.2011.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Storandt M, Head D, Fagan AM, Holtzman DM, Morris JC. Toward a multifactorial model of Alzheimer disease. Neurobiol Aging. 2012;33(10):2262–71. doi: 10.1016/j.neurobiolaging.2011.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Peng S, Wuu J, Mufson EJ, Fahnestock M. Increased proNGF Levels in Subjects with Mild Cognitive Impairment and Mild Alzheimer's Disease. J Neuropathol Exp Neurol. 2004;63(6):641–649. doi: 10.1093/jnen/63.6.641. [DOI] [PubMed] [Google Scholar]
- 182.Peng S, Wuu J, Mufson EJ, Fahnestock M. Increased proNGF levels in subjects with mild cognitive impairment and mild Alzheimer disease. J Neuropathol Exp Neurol. 2004;63(6):641–9. doi: 10.1093/jnen/63.6.641. [DOI] [PubMed] [Google Scholar]
- 183.Mufson EJ, He B, Nadeem M, Perez SE, Counts SE, Leurgans S, Fritz J, Lah J, Ginsberg SD, Wuu J, Scheff SW. Hippocampal proNGF signaling pathways and beta-amyloid levels in mild cognitive impairment and Alzheimer disease. J Neuropathol Exp Neurol. 2012;71(11):1018–29. doi: 10.1097/NEN.0b013e318272caab. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Counts SE, He B, Prout JG, Michalski B, Farotti L, Fahnestock M, Mufson EJ. Cerebrospinal fluid proNGF: A putative biomarker for early Alzheimer's disease. Curr Alzheimer Res. 2016 doi: 10.2174/1567205013666160129095649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Kuhn TS. The structure of scientific revolutions. University of Chicago Press; Chicago: 1962. [Google Scholar]
- 186.Herrup K. The case for rejecting the amyloid cascade hypothesis. Nat Neurosci. 2015;18(6):794–9. doi: 10.1038/nn.4017. [DOI] [PubMed] [Google Scholar]
- 187.Mufson EJ, Malek-Ahmadi M, Perez SE, Chen a.K. Braak staging, plaque pathology and APOE status in elderly persons without cognitive impairment. Neurobiol Aging. 2016 doi: 10.1016/j.neurobiolaging.2015.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.[New perspectives for Alzheimer patients: acetylcholine inhibition with Rivastigmine. Congress of the European Neurologic Society. Nice, 6 July 199] Deutsche medizinische Wochenschrift. 1998;123(48 Suppl):1–4. [PubMed] [Google Scholar]
- 189.Olsson B, Lautner R, Andreasson U, Ohrfelt A, Portelius E, Bjerke M, Holtta M, Rosen C, Olsson C, Strobel G, Wu E, Dakin K, Petzold M, Blennow K, Zetterberg H. CSF and blood biomarkers for the diagnosis of Alzheimer's disease: a systematic review and meta-analysis. Lancet Neurol. 2016 doi: 10.1016/S1474-4422(16)00070-3. [DOI] [PubMed] [Google Scholar]