Open access

Down Syndrome Model of Alzheimer’s Disease: Beyond Trisomy 21Nondisjunction

Written By

Antoneta Granic and Huntington Potter

Submitted: 08 April 2011 Published: 29 August 2011

DOI: 10.5772/24948

From the Edited Volume

Genetics and Etiology of Down Syndrome

Edited by Subrata Dey

Chapter metrics overview

3,202 Chapter Downloads

View Full Metrics

1. Introduction

Alzheimer’s disease (AD), the most common type of dementia in old age, is a complex, multifactorial neurodegenerative disorder currently affecting 35,6 million people in either familial (genetic) or sporadic form, whose prevalence is expected to quadruple worldwide by the year 2050 (Alzheimer’s Disease International, 2010; Ferri et al., 2005). While genetic or early-onset AD accounts for only 5% of all cases (Cummings, 2004), 95% of sporadic or late-onset AD is attributed to the interaction between advancing age, environmental factors (Grant, et al., 2002; Tanzi & Bertram, 2001), and to the few risk-enhancing genetic polymorphisms discovered so far (Bertram & Tanzi, 2004). Autosomal dominant mutations in the amyloid precursor protein (APP), presenilin 1 (PS1), and presenilin 2 (PS2) are linked to early-onset (familial) AD (FAD, Hardy, 2009). A combination of bio-psycho-social factors, especially if experienced at midlife, such as abnormal glucose and cholesterol metabolism, cerebral hypoperfusion, hypertension, physical and mental inactivity, diet, head injury, depression and small social networks are thought to contribute to sporadic AD initiation and development (reviewed in Flicker, 2010; Bendlin et al., 2010; Hughes & Ganguli, 2009). There is a temporal gap between clinical and pathological manifestation of Alzheimer’s dementia; AD has a long prodromal phase and many clinically silent older adults are experiencing substantial pathological burden (Elias et al., 2000; Small et al., 2000). Common clinical manifestation of AD include memory impairment and dysfunctions in several cognitive domains such as language, problem solving, executive function, visuospatial skills, and others along with changes in personality and behavior (Maurer & Hoyer, 2006).

1.1. Neuropathology of Alzheimer’s disease

The brain pathology of AD is characterized by extracellular deposits of the amyloid-beta peptide (Aβ), a main constituent of senile plaques, and intracellular accumulation of the hyperphosphorylated protein tau (p-tau) (Selkoe, 1999; 2001). The central hypothesis of AD, the amyloid cascade hypothesis, posits that an imbalance between Aβ production, aggregation and clearance from the brain is the initiating event in the disease process leading to synaptic loss, neuronal degeneration, inflammation, senile plaque formation and dementia (Blennow, de Leon, & Zetterberg, 2006; Hardy, 2006; Hardy & Selkoe, 2002; Lee et al., 2004; Potter et al., 2001).

The main constituent of the extracellular senile plaques is a highly neurotoxic Aβ42 isoform of Aβ protein, produced by consecutive action of two cleaving proteases, β- (BACE-1) and γ-secretase, from a larger transmembrane protein, the Amyloid Precursor Protein (APP) (Hardy, 2009; Haass & Selkoe, 1993; Wolfe, 2003) within cholesterol rich membrane domains (Wahrle et al., 2002). A number of biochemical and genetic studies have indicated that oligomerization or polymerization of Aβ peptide driven by inflammation-induced proteins is a crucial step in AD pathogenesis (Potter et al., 2001; Hardy, 2006). Aβ42 self-aggregates and forms insoluble plaque deposits that include dystrophic neurites, activated microglia and reactive astrocytes (Itagati et al., 1989) which express a number of proinflammatory proteins (Akiyama et al., 2000; Potter et al., 2001). Furthermore, Aβ induces increased phosphorylation of the intracellular protein tau, resulting in destabilization of microtubules (MT) (Small & Duff, 2008). The evidence from transgenic mice and human studies indicates that not only extracellular, but also intracellular Aβ accumulates in diseased neurons and contributes to dementia progression by affecting the function of mitochondria, calcium ion channels and synapses (La Ferla, Green, and Oddo, 2007; Li et al., 2007). It is still unclear which toxic species of the Aβ peptide are most damaging to the neurons or whether the process of aggregation itself is detrimental to neuronal membranes.

Neurofibrillary tangles (NFTs) are the second major pathological hallmark in AD, which result from abnormally aggregated tau protein filling the intracellular space of the neurons. In AD brain, tau gets hyperphosphorylated, detaches from microtubules and forms paired helical filaments, which cause disruption in neuronal signaling, synaptic failure, impaired nutrient trafficking and neuronal death (Alonso et al., 2006; Grundke-Iqbal et al., 1986). Furthermore, oxidative damage as a consequence of free radical and reactive oxygen species (ROS) attacks has been observed in post-mortem AD brains in the form of oxidized lipids and proteins, mutated DNA, and mitochondrial damage (Floyd & Hensley, 2002). Formation of amyloid plaques and neurofibrillary tangles is not only associated with neuronal and synaptic loss, and depletion of neurotransmitters (Scheff, et al., 2006; Selkoe, 1999; 2004), but with abnormal axonal transport of key molecules and organelles important for neural cells survival and communication early in the disease process (Stokin et al., 2005; Stokin & Goldstein, 2006). Neurons, with their long axons, branched dendrites and large cell surface are especially vulnerable to energy and/or oxygen deprivation, impaired movement of molecules, and neurotoxins observed in AD pathology (Mattson & Magnus, 2006). The development of early diagnosis and successful treatments of AD will be greatly aided by a complete understanding of the pathological pathway that leads to formation of misfolded proteins, inflammation and neurodegeneration.

1.2. The Down syndrome model of Alzheimer’s disease

The discovery that Down syndrome (DS) patients who live beyond the age of 30 or 40 develop neuropathology indistinguishable from the one observed in classic AD (Glenner & Wang, 1984; Olson & Shaw, 1969; Wisniewski et al., 1988) provided an important insight into AD pathogenesis, shifting the focus on nondisjunction of human chromosome 21 (HSA21) where the APP gene resides (Goldgaber et al. 1987; Neve et al., 1988; Petterson et al., 1988; Tanzi et al., 1987 ), and on the consequence of the gene overexpression in DS (Epstein, 1990). The fact that an extra copy of APP and subsequent 50% increase in gene dosage due to trisomy 21 in every cell of the body in DS individuals is sufficient to cause AD later in life instigated research on common biological links between AD and DS (Potter, 1991, 2008; Geller & Potter, 1999). Trisomy 21 results in altered APP processing and in an increased ratio of more amyloidogenic Aβ42 over Aβ40 (Teller et al., 1996), similar to the process observed in animal models of AD and in patients harboring mutations in FAD genes, PS1, PS2 and APP (Haas & De Stooper, 1999; Hardy & Selkoe, 2002; Suzuki et al., 1994; Wolfe, 2003).

Several biochemical and genetic studies have shown that both sporadic and familial AD patients, including those carrying FAD mutations, are abnormal in one or more aspects of the cell cycle (Arendt et al., 1996; Geller & Potter, 1999; Potter, 1991; Varvel et al., 2008; Yang et al., 2001; 2006; Yang & Herrup, 2007; reviewed in Nagy, 2005; Obrenovic et al., 2003; Potter, 2004, 2008). The universal presence of AD pathology in DS individuals and the occurrence of an aberrant cell cycle in the brains of FAD mouse models and AD patients, led us to hypothesize that a slow accumulation of aneuploid, including trisomy 21 cells through defective mitosis and chromosome mis-segregation in central and peripheral tissues over the course of life of an individual could cause or at least help promote late-onset Alzheimer’s (Potter, 1991; 2008). The extra copy of chromsome 21 that in full human trisomy of DS leads to neurodegeneration and dementia, could account for both genetic and sporadic AD, depending upon whether the chromosomal instability and mosaic aneuploidy was induced by a genetic (familial) mutation or by environmental insults. We further postulated that the microtubule disfunction likely responsible for the mitotic defects and genomic instability in AD could be linked to altered APP production and increased Aβ levels, probably affecting other aspects of neuronal physiology and function (Borysov et al., 2011; Granic et al., 2010; Potter, 2008).

The Down syndrome model and chromosome mis-segregation/microtubule disfunction hypothesis of AD made several predictions (Geller & Potter, 1999; Potter, 2008):

  1. Alzheimer’s patients should harbor a small number of aneuploid, including trisomy 21 cells in their somatic tissues. Altered genomic stability and development of trisomy 21 mosaicism would contribute to dementia onset and neurodegeneration but at slower pace than in DS due to the modulating effect of mostly disomic cells in the body.

  2. Mutations that cause familial AD should occur in genes that encode proteins directly or indirectly involved in the cell cycle and chromosome segregation.

  3. There should be alternations in microtubules, mitotic spindle apparatus and mitosis-related proteins in AD cells that could lead to aneuploidy, including trisomy 21 mosaicism.

In the past twenty years, compelling epidemiological and molecular evidence from our and other laboratories has been accumulated to test all three predictions. Together, the evidence suggests a link between pathological changes observed in the brains of DS and AD individuals and chromosomal instability and mosaic aneuploidy, including nondisjunction of HSA21 which likely contributes to dementia initiation and/or progression, with important implications for AD diagnosis and therapy.

Advertisement

2. The epidemiology of trisomy 21 mosaicism in Alzheimer’s disease

Early epidemiological evidence indicating that chromosome mis-segregation and trisomy 21 mosaicism might be implicated in AD pathogenesis came for the studies showing a significantly higher number of Down syndrome offspring born in some families with FAD mutations (Heston et al., 1981; Heyman et al., 1983). The studies that failed to confirm this association reported to have too small sample sizes to observe statistically significant results (Amaducci et al., 1986; Chandra et al., 1987) suggesting that larger scale studies are needed to establish a connection between a higher frequency of DS children in families with genetic forms of AD. An important result that provided support for the trisomy 21 model of AD came from a retrospective study of young mothers (aged <35) showing a five-fold greater risk of developing AD later in life compared to either older DS mothers or the general population (Schupf et al., 1994; 2001). Schupf and her colleagues interpreted this phenomenon as a novel form of ‘accelerated aging’. In the light of our trisomy 21 model of AD, we postulated that the young DS mothers were most likely mosaic for chromosome 21 and had a predisposition for genomic instability, which resulted in DS progeny and their own increased risk of AD later in life. Indeed, a recent study by Migliore et al. (2006; 2009) confirmed the susceptibility to aneuploidy and trisomy 21 nondisjunction in young mothers of DS children. Case studies of patients with trisomy 21 mosaicism and no intellectual impairments of the DS type who developed AD by the age of 40 demonstrated that a small percentage of chromosomal instability is sufficient to result in early-onset AD (Hardy, et al., 1989; Ringman et al. 2008; Schapiro et al., 1989). Similarly, a slow accumulation of a low number of trisomy 21 cells over the life span may lead and/or contribute to the pathogenesis of both genetic and sporadic form of AD.

In order to directly test if Alzheimer’s patients harboured mis-segregated including trisomy 21 cells, we and others have used fluorescence in situ hybridization (FISH), a cytogenetic technique that allows one to detect the number of copies of a particular chromosome in both metaphase and interphase cells (Ried, 1998) with great sensitivity and specificity. This method is especially suitable for poorly and non-dividing cells, or for the cells with a very low level of aneuploidy (e.g., lymphocytes). In our early study of chromosome mis-segregation in AD, we found more than twice the frequency of trisomy 21 in skin fibroblasts of AD patients (p=0.007) compared to age-matched controls, which was not related to the age of affected individuals (Figure 1, Geller & Potter, 1999). A small parallel study of chromosome 18 nondisjunction showed a similar increase in aneuploidy, indicating that the mitotic defect likely affected all chromosomes. The AD fibroblasts in our study included those with sporadic (late-onset) AD and those carrying a familial AD mutation in PS1, PS2, or APP which are now known to cause early AD onset and altered APP processing (e.g., Li et al., 1995; Schellenberg et al., 1993; Rogaev et al. 1995).

Figure 1.

AD fibroblasts, including those with PS1 or PS2 mutation (FAD) and those of sporadic origin (SAD) showed increased trisomy 21, *p < 0.01; **p < 0.001. (Used with permission from Geller & Potter, 1999, Neurobiology of Disease).

Trisomy 21 mosaicism and chromosome mis-segregation have also been observed among peripheral blood lymphocytes (Migliore 1997; 1999; Trippi et al., 2001), buccal cells (Thomas & Fenech, 2008), and neurons from sporadic AD patients (Arendt et al., 2010; Iourov et al., 2009; Mosch et al., 2007; Yang et al., 2001). Specifically, AD brains harbor up to 35% of hyperploid, including trisomy 21 and other tetrasomic neurons which are detectable at mild stages of the disease before any evident neuronal loss (Arendt et al., 2010; Iourov et al. 2009). The presence of tetrasomic neurons as explained by some reseachers, may indicate re-entry into an incomplete cell cycle and selective vulnerability to cell death of post-mitotic, mosaic neurons as an important pathogenic event in AD (Arendt et al., 2010; Obrenovich et al., 2003; Varvel et al., 2008; Vincent et al., 1996; Yang & Herrup, 2007). In our studies of AD mosaicism (Boeras et al., 2008; Geller & Potter, 1999; Granic et al., 2010), we failed to observe statistically significant induction of tetrasomic cells in sporadic and familial models of AD, but rather pronounced accumulation of trisomy cells in both peripheral and brain tissues. A larger scale cytogenetic autopsy and biopsy study, preferably at the single cell level is needed to further elucitate the type and extent of pathologic genetic instability in AD and other neurodegenerative diseases.

In summary, several laboratories confirmed that trisomy 21 mosaicism is commonly associated with AD. Moreover, there is an indication of a dose response effect in which full trisomy 21 in DS individuals elicits AD-like pathology by age 20 and by middle age in familial AD and even later in sporadic Alzheimer’s patients, who also belonged to the trisomy 21 mosaic group found to have an increased frequency of DS children before the age of 35. The recent discovery of families that develop early-onset inherited AD only because the APP gene on one chromosome 21 is duplicated (Mc Naughton et al., 2010; Rovelet-Lecrux et al., 2006; Sleegers et al., 2006) indicates that the extra copy of the APP gene and consequent overproduction of Aβ peptide is the likely cause of AD in both Down syndrome and trisomy 21 mosaic individuals.

2.1. Mitotic defects in Alzheimer’s disease

Along with the studies described above, separate lines of investigations provided independent evidence that other mitotic defects and mitosis-specific proteins may be present in the cells of AD patients (reviewed in Potter, 2004). For instance, the mitotic spindles in dividing AD cells exhibit abnormalities and susceptibility to premature centromere division (PCD) and micronucleation upon chemically induced (e.g., colchicines) microtubule damage (e.g., Fitzgerald et al., 1986; Potter et al., 1995; Trippi et al., 2001). The event of PCD, in which individual sister chromatids are separated by a clear gap and not connected at the centromeres, has been observed in patients prone to genomic instability and chromosome mis-segregation, and recently confirmed in neurons of individuals with sporadic AD (Spremo-Potparević et al., 2008).

Further evidence linking cell cycle defects with AD pathogenesis came for the finding that both APP and the microtubule-stabilizing tau protein get increasingly phosphorylated during mitosis (Suzuki et al., 1994; Pope et al., 1994; Padmanabhan et al., 2011). Also, phospho-tau and other mitosis-specific phospho-proteins are overexpressed in AD but not in normal brains (e.g., Arendt et al., 1996; Vincent et al., 1996; Nagy et al., 1997), indicating a cycling stage of AD neurons. The idea of unscheduled cycle in fully differentiated neurons challenged the dogma of their post-mitotic nature and the inability to replicate (Rakic, 1985). To date, the evidence drawn from animal models of AD and autopsy studies of human brains indicates that reactivation of the cell cycle and DNA duplication (e.g., Yang et al., 2001) in the vulnerable population of AD neurons may present a fundamental initiator of AD pathogenesis (Nagy, 2005; Vincent et al., 1996; Varvel et al., 2008; Yang & Herrup, 2007; Yang et al., 2006) present before deposition of fibrilar Aβ and neuronal death, and could also lead to chromosome mis-segregation and aneuploidy.

Advertisement

3. Presenilin and APP mutations induced aneuploidy

The finding that fibroblast cultures from patients carrying autosomal dominant mutations in the PS1, PS2 or APP gene harbour chromosomal instability and trisomy 21 mosaicism provided the first indication that FAD genes are likely to be involved in mitosis and chromosome mis-segregation, or to be associated with structures and proteins of the cell cycle (Geller & Potter, 1999).

Indeed, we and others have confirmed a major location of PS1 and PS2 proteins in dividing cells in the centromeres, the nuclear envelope, and the kinetochores during interphase (Li et al., 1997; Honda et al., 2000). Further support for a mitotic function of presenilins comes from studies showing inhibition of the cell cycle (Janicki & Montero, 1999) and increased sensitivity to apoptosis (Vito et al., 1996; Wolozin et al., 1996) in the cells carrying a mutated PS gene. Furthermore, polymorphisms in the PS1 gene have been associated with an increased risk of AD (Wragg et al., 1996; Higuchi et al., 1996; Scott et al., 1996) and with an increase of DS offspring via a meiosis II defect (Petersen et al., 2000; Lucarelli et al., 2004) as a more direct confirmation of PS1 involvement in the cell cycle.

Further research should be directed at discovering the mechanism by which the mutant presenilins influence chromosome segregation. One possibility supported by the data collected so far points to inability of altered presenilin proteins to properly link the chromosomes to the nuclear envelope and to release them at the appropriate time during mitosis, which may lead to chromosome mis-segregation and other cell cycle abnormalities. Another possibility discussed further below links PS mutations to altered processing of APP and increased production of neurotoxic Aβ42 as a likely effector molecule responsible for cell cycle defects, including mitotic spindle abnormalities and chromosome mis-segregation (Boeras et al., 2008; Borysov et al., 2011, Granic et al., 2010). Similarly, APP is also found to localize to the centrosomes and nuclear membrane in dividing cells (Nizzari et al., 2007; Zitnik et al., 2006), and to get increasingly phosphorylated during the cell cycle (Padmanabhan et al., 2011).

In a series of in vivo and in vitro experiments we investigated the role of mutated PS1 and APP genes and their proteolytic product, the Aβ peptide in chromosome mis-segregation and trisomy 21 mosaicism. All assays, tissues and cells from transgenic mice carrying AD mutations and the cells transfected with FAD genes or treated with Aβ peptide yielded comparable results: overexpression of FAD genes in vivo and in vitro and exposure to Aβ peptide induced chromosome instability and trisomy 21 mosaicism through several defects in mitotic spindle apparatii and dysfunction of microtubule assembly (Boeras et al., 2008; Borysov et al., 2011; Granic et al., 2010; Potter et al., 2008).

3.1. Presenilin and APP mutations induced aneuploidy in transgenic mice

We asked whether chromosome mis-segregation and trisomy 21 mosaicism observed in human fibroblasts with PS1 or APP mutations could be mimicked in peripheral and brain tissues of FAD-transgenic mice. For example, whole brains from PS1 (M146l and M146V) and APP (V717) mutant mice and non-transgenic littermates were processed to yield primary cultures. The isolated neurons were hybridized with a mouse chromosome 16 BAC probe (Kulnane et al., 2002), followed by immunocytochemistry to stain for neurons. Most cells were disomic with two copies of chromosome 16; while the neurons from PS1 mutant and PS1 knock in mice exhibited up to 4% of trisomy 16 (data not shown, Boeras et al, 2008), the APP-transgenic mice had about 6.5% of trisomies (Figure 2, Granic et al., 2010).

Figure 2.

Quantitative FISH analysis revealed significantly higher levels of trisomy 16 in APP mice (A & B) compared to controls but no tetrasomy (D) and no induction of aneuploidy in non-neuronal cells (C). (Used with permission from Granic et al., 2010, Molecular Biology of the Cell).

3.2. Presenilin and APP mutations induced aneuploidy in transfected cells

To determine whether the aneuploidy observed in FAD-transgenic and knock-in mice was caused directly by mutated genes and not by some other factors, parallel cultures of the hTERT-HME1, an immortalized primary mammary epithelial cell line with a stable karyotype (Clontech) were transiently transfected with WT-PS1, mutant PS1 (M146L), mutant APP (K595N/M596L and V642I or V717) and control empty vector (pcDNA3 and paG3). FISH was used to assess the levels of aneuploidy for chromosome 21 and 12. Overexpression of FAD-genes induced between 2-3% of trisomy 21 and/or trisomy 12 (Boeras et al., 2008; Granic et al., 2010), and about 30% of total aneuploidy in metaphase cells within 48 hours (Boeras et al., 2008). These results indicated that an aneugenic effect of FAD-mutations likely affected all chromosomes with random gains and losses of whole chromosomes, and that chromosome mis-segregation was not restricted only to the cells expressing mutated genes, but extended to nearby, non-transfected cells. We hypothesized that Aβ peptide itself found at increased levels in both familial and sporadic AD might be the probable effector molecule interfering with mitosis and chromosome segregation (Boeras et al., 2008; Potter, 2008). Lastly, immunocytochemistry of PS1-transfected cells revealed several abnormalities in the mitotic spindles, with disarrayed microtubules, multiple centosomes and lagging chromosomes as the most prominent spindle malformations (Boeras et al., 2008).

3.3. Aβ induced aneuploidy and the role of tau

Sequential cleavage of the APP protein with β- and presenilin/γ-secretase enzymes yields more amyloidogenic Aβ42 peptide as a central event in AD pathogenesis. We proposed to test the role of Aβ in genomic instability and trisomy 21 induction. Led by our initial observation that more cells became aneuploid than are transfected and express FAD genes, we examined the aneugenic effect of Aβ peptide in culture. hTERT-HME cells treated with 1μM Aβ40 and Aβ42 develop more than 20% aneuploid metaphases and about 2% trisomy 21 and 12 cells within the 48 hours of exposure compared to 6% and less than 1% of the cells treated with various control peptides (Figure 3, Granic et al., 2010). These results indicated that AD might be a self-propagating disorder in which the product of FAD mutations and trisomy 21, the Aβ peptide, further induces chromosome segregation and generation of trisomy 21 cells.

Figure 3.

Quantitative FISH analysis with a dual color probe revealed an increased induction of trisomy 21 (SpectrumOrange, Vysis) and trisomy 12 (SpectrumGreen) in Aβ treated hTERT-HME cells (A-C). (Used with permission from Granic et al., 2010, Molecular Biology of the Cell).

To investigate the mechanism by which Aβ exerts its aneugenic effect on dividing cells, we analyzed the peptide’s other toxic activities especially those related to microtubule function. Several lines of investigation have indicated that Aβ induces and requires downstream changes/defects in microtubules (MT) to exert its neurodegenerative activity. Numerous in vitro and in vivo studies have shown that Aβ induces phosphorylation of tau (e.g., Small & Duff, 2008), and that Aβ toxicity depends on the presence of tau (Rappaport et al., 2002). Therefore, we investigated the role of tau in Aβ induced aneuploidy. Splenocytes prepared from normal, Tau+/-, Tau -/- mice were treated with Aβ peptide and analyzed for aneuploidy 48 hours later. Knocking out one or even more effectively both copies of Tau induced up to 5% aneuploidy (trisomy 16). However, the Aβ aneugenic effect was greatly attenuated in the cells lacking Tau but not in normal cells, indicating that Aβ induced chromosome mis-segregation requires tau protein and disrupts normal tau-stabilizing microtubule function. In a series of studies, Borysov et al. (2011) have shown that Aβ42 peptide added to Xenopus egg extracts impairs the structure and stability of mitotic spindles, and inhibits three motor kinesins, Eg5, KIF4A, and MCAK, required for normal mitotic spindle function and proper chromosome segregation.

Recently, we have shown that overexperssion of APP prevents the localization of low density lipoprotein receptor (LDLR) from the Golgi to the cell membrane (Abisambra et al., 2010). This latest finding led us to hypothetize that neurons exposed to Aβ in AD brains may also fail to localize other key receptors to the cell membrane, including those for neurotrophins and neurotransmitters, causing neuronal disfunction and dementia. Future studies are under way to confirm if the interference with microtubule function by Aβ will cause defects in neuroplasticity through mis-localization of the receptors away from the plasma membrane, as well as contributing to defective neurogenesis leading to dysfunctional, aneuploid, including trisomy 21 neurons prone to Aβ overproduction and neurodegeneration.

Advertisement

4. How trisomy 21 mosaicism may lead to Alzheimer’s disease

Several potential mechanisms could explain how trisomy 21 mosaicism could lead to AD (Potter, 1991; 2004; 2008). For instance, aneuploidy cells might be prone to cell death and neurodegeneration (Arendt et al., 2010), similar to cortical neurons in DS brains that undergo spontaneous apoptosis (Busciglio & Yankner, 1997). Apoptosis could also indirectly affect APP processing and Aβ levels in mosaic AD brain. The support for the latter hypothesis comes from the finding that embryonic DS brains and adult sera contain a higher ratio of neurotoxic Aβ42 over Aβ40 peptide (Teller et al., 1996). Also, trisomy 21 microglia overexpress inflammatory proteins and begin an inflammatory cascade that promotes Aβ fibrilization (Potter et al., 2001). Finally, aneuploidy in AD may arise from a defect in microtubule function which may lead to poor protein, neurotransmitters and nutrient trafficking (Cash et al., 2003). Trisomy 21 may be both a cause and an effect of microtubule dysfunction generating a feed-forward loop further promoting AD progression.

Advertisement

5. Implications of the trisomy 21 model of Alzheimer’s disease for diagnosis and therapy

The mechanistic implication of the results discussed in this review is that an early step in Alzheimer’s disease pathogenesis may be the development of genomic instability and trisomy 21, contributing to progression of dementia. The search for more effective diagnoses should take into account the events of mitotic defects in peripheral and central tissues of individuals at risk. Chromosome analysis and detection of low levels of trisomy 21 in skin fibroblasts or buccal cells in patients during the pre-clinical phase of dementia could be a potential diagnostic test. Another implication of the data presented above that trisomy 21 in AD may be the initiating event in disease pathogenesis also suggests new approaches to treatments (Potter, 2004). For example, drugs that would repair the mitotic defects and strengthen the fidelity of the chromosome segregation could be searched for and used prophylactically. Further, aneugenic environmental agents that cause chromosome instability could be identified and counteracted with drugs that restore genomic homeostasis. Another more difficult but equally effective approach to therapy would be to detect and remove mis-segregated cells from the body by exploiting their unique cell biology and/or gene expression.

To summarize, the results from several laboratories over the past twenty years have shown that Alzheimer’s patients are prone to genomic instability accumulating about 2-3 fold more trisomy 21 cells throughout the body compared to age matched healthy controls. The precise mechanism by which these abnormal cells arise during the life span of an individual and how or whether they contribute to disease initiation and progression are subjects of active investigation. The better understanding of these novel findings has the potential to contribute to the development of future diagnoses and therapies for Alzheimer’s disease.

Advertisement

6. Summary

Convincing epidemiological and molecular evidence has been accumulating that link pathological changes observed in the brains of both Down syndrome individuals and neurodegeneration in Alzheimer’s disease to chromosomal instability and trisomy 21 mosaicism. The results from several laboratories indicate that errors in mitosis, specifically mis-segregation of somatic chromosomes in peripheral and brain tissues of AD patients may play an important role in both early (familial) and late (sporadic) onset of disease. Here, we proposed a unifying hypothesis for Alzheimer’s and Down syndrome neurodegeneration—development of a mosaic population of aneuploid, including trisomy 21 cells and alternation in genomic stability may lead to classic AD neuropathology observed in both diseases. The evidence for this hypothesis include: a) cells from familial and sporadic AD patients exhibit mis-segregated, including trisomy 21 cells in brain, blood, mucosa and skin, and harbour abnormalities in several aspects of the cell cycle that may contribute to aneuploidy and neurodegeneration; b) overexpression of mutated Alzheimer’s genes, presenilin 1 and APP, in cellular and transgenic mouse models induce aneuploidy, including trisomy 21; and c) Aβ peptide is the likely effector molecule responsible for disruption of proper functioning of microtubules and mitotic spindle integrity leading to mitotic defects and apoptosis. The possibility that many cases of Alzheimer’s disease are mosaic for trisomy 21 opens new approaches for diagnosis and therapy.

Advertisement

Acknowledgments

Our work was supported by the Byrd Alzheimer’s Institute, the Eric Pfeiffer Chair for Research on Alzheimer’s Disease, the Alzheimer’s Association, and the National Institute on Aging grant No. AG25711. We thank Dr. Bruce Lamb for the chromosome 16 BAC probe and Dr. Karen Duff for PS1 transgenic mice. Figure 1 was reprinted from the publication ‘Chromosome missegregation and trisomy 21 mosaicism in Alzheimer's disease‘, Neurobiology of Disease, Vol. 6, No. 3, (December, 1998), pp. 167-179, Geller, L. N., & Potter, H. (1999), copyright (1998), with permission from Elsevier. Figure 2 and Figure 3 were reprinted from the publication ‘Alzheimer Aβ peptide induces chromosome mis-segregation and aneuploidy, including trisomy 21: requirement for tau and APP. Molecular Biology of the Cell, Vol. 21, No. 4, (December, 2009), pp. 511-520, copyright (2009), with the permission from the Molecular Biology of the Cell.

References

  1. 1. AbisambraJ. F.FiorelliT.PadmanabhanJ.et al.2010LDLR expression and localization are altered in mouse and human cell culture models of Alzheimer’s disease. PLoS ONE, 51January, 2010), e8556
  2. 2. AkiyamaH.BargerS.BarnumS.et al.2000Inflammation and Alzheimer’s disease. Neurobiology of Aging, 213May, 2000), 383421
  3. 3. AlonsoA.del C.LiB.Grudke-IqbalIgbalK.2006Polymerization of hyperphosphorylated tau into filaments eliminates its inhibitory activity. Proceedings of the National Academy of Science of the USA, 103June, 2006), 88648869
  4. 4. AmaducciL. A.FratiglioniL.RoccaW. A.et al.1986Risk factors for clinically diagnosed Alzheimer’s disease : a case-control study of an Italian population. Neurology, 367July, 1986), 922931
  5. 5. Alzheimer’s Disease International2010World Alzheimer Report 2010, In: Alzheimer’s Disease International, 12.12.2010, Available from: http://www.alz.co.uk/research/files/WorldAlzheimerReport2010ExecutiveSummary
  6. 6. ArendtT.BrücknerM. K.MoschB.et al.2010Selective cell death of hyperploid neurons in Alzheimer’s disease. The American Journal of Pathology, 1771July, 10), 1520
  7. 7. ArendtT.RodelL.GartnerU.et al.1996Expression of the cyclin-dependent kinase inhibitor p16 in Alzheimer’s disease. Neuroreports, 718November, 1996), 30473049
  8. 8. BendlinB. B.CaressonC. M.GleasonC. E.et al.2010Midlife predictors of Alzheimer’s disease. Maturitas, 652February, 2010), 131137
  9. 9. BertramL.TanziR. E.2005Alzheimer’s disease: one disorder, too many genes? Human Molecular Genetics, 13Spec, 1April, 2004), R135R141
  10. 10. BlennowK.de LeonM.ZetterbergH.2006Alzheimer’s disease. The Lancet, 3689533July, 2006), 387403
  11. 11. BoerasD. I.GranicA.PadmanabhanJ.et al.2008Alzheimer’s presenilin 1 causes chromosome missegregation and aneuploidy. Neurobiology of Aging, 293December, 2006), 319328
  12. 12. BorysovS. I.GranicA.PadmanabhanJ.WalczakC. E.PotterH.2011Alzheimer Aβ disrupts the mitotic spindle and directly inhibits mitotic microtubule motors. Cell Cycle, 109May, 2011), 114
  13. 13. BusciglioJ.YanknerB. A.1996Apoptosis and increased generation of reactive oxygen species in Down syndrome neurons in vitro. Nature, 3786559December, 1995), 776779
  14. 14. CashA. D.AlievG.SiedlakS. L.et al.2003Microtubule reduction in Alzheimer’s disease and aging is independent of tau filament formation. American Journal of Pathology, 1625May, 2003), 16231627
  15. 15. ChandraV.PhiliposeV.BellP. A.et al.1987Case-control study of probable Alzheimer’s disease. Neurology, 378August, 1987), 12951300
  16. 16. CummingsJ. L.2004Alzheimer’s disease. New England Journal of Medicine, 3511July 2004), 5667
  17. 17. EliasM. F.BeiserA.WolfP. A.et al.2000The preclinical phase of Alzheimer disease: A 22-year prospective study of Framingham cohort. Archives of Neurology, 576June, 2000), 808813
  18. 18. EpsteinC. J.1990The consequences of chromosome imbalance. American Journal of Medical Genetics: Supplement, 731-7n.d), 3137
  19. 19. FerriC. P.PrinceM.BrayneC.BrodatyH.et al.2005Alzheimer’s Disease International. Global prevalence of dementia: a Delphi consensus study. Lancet, 3669503December, 2005), 21122117
  20. 20. FitzgeraldP. H.ArcherS. A.MorrisC. M.1986Evidence for the repeated primary non-disjunction of chromosome 21 as a result of premature centromere division (PCD). Human Genetics, 721January, 1986), 5862
  21. 21. FlickerJ.2010Modifiable lifestyle risk factors for Alzheimer’s disease. Journal of Alzheimer’s Disease, 203n.d.), 803811
  22. 22. FloydR. F.HensleyK.2002Oxidative stress in brain aging: Implications for therapeutics of neurodegenerative diseases. Neurobiology of Aging, 235September, 2002), 795 EOF807 EOF
  23. 23. GellerL. N.PotterH.1999Chromosome missegregation and trisomy 21 mosaicism in Alzheimer’s disease. Neurobiology of Disease, 63December, 1998), 167179
  24. 24. GlennerG. G.WangC. W.1984Alzheimer’s disease and Down’s syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Biochemical and Biophysiological Research Communications, 1223August, 1984), 1131 EOF5 EOF
  25. 25. GoldgaberD.LermanM. J.Mc BrideO. W.et al.1987Characterization and chromosomal localization of cDNA encoding brain amyloid of Alzheimer’s disease. Science, 2354791February, 1987), 877880
  26. 26. GranicA.PadmanabhanJ.NordenM.PotterH.2010Alzheimer Aβ peptide induces chromosome mis-segregation and aneuploidy, including trisomy 21: requirement for tau and APP. Molecular Biology of the Cell, 214December, 2009), 511520
  27. 27. GrantW. B.CampbellA.ItzhakiR. F.et al.2002The significance of environmental factors in the etiology of Alzheimer’s disease. Journal of Alzheimer’s Disease, 43June, 2002), 179189
  28. 28. Grundke-IqbalI.IqbalK.QuinlanM.et al.1986Microtubule associated protein tau. A component of Alzheimer paired helical filaments. The Journal of Biological Biochemistry, 26113May, 1986), 60846089
  29. 29. HardyJ.2006Alzheimer’s disease: the amyloid cascade hypothesis: an update and reappraisal. Alzheimers Disease, 93Suppl), 151153
  30. 30. HardyJ.2009The amyloid hypothesis for Alzheimer’s disease: a critical reappraisal. Journal of Neurochemistry, 1104August, 2009), 11291134
  31. 31. HardyJ.GoateA.OwenM.et al.1989Presenile dementia associated with mosaic trisomy 21 in a patient with Down syndrome child. Lancet, 28665September, 1989), 743
  32. 32. HaassC.SelkoeD. J.1993Cellular processing of beta-amyloid precursor protein and the genesis of amyloid beta-peptide. Cell, 756December, 1993), 10391042
  33. 33. HestonL. L.MastriA. R.AndersonV. E.et al.1981Dementia of Alzheimer type. Archives of Genetic Psychiatry, 3810October, 1981), 10811090
  34. 34. HeymanA.WilkinsonW.HurwitzB.et al.1983Alzheimer’s disease; genetic aspects and associated clinical disorders. Annals of Neurology, 145November, 1983), 507515
  35. 35. HiguchiS.MuramatsuT.MatsushitaS.et al.1996Presenilin-1 polymorphism and Alzheimer’s disease. Lancet, 3479009April, 1996), 1186
  36. 36. HondaT.NihonmatsuN.YasutakeK.et al.2000Familial Alzheimer’s disease-associated mutations block translocation of full-length presenilin 1 to the nuclear envelope. Neuroscience Research, 372June, 2000), 101111
  37. 37. HughesT. F.GanguliM.2009Modifiable risk factors for late-life cognitive impairment and dementia. Current Psychiatry Reviews, 52May, 2009), 7392
  38. 38. ItagakiS.Mc GreerP. L.AkiyamaH.et al.1989Relationship of microglia and astrocytes to amyloid deposits of Alzheimer disease. Journal of Immunology, 243October, 1989), 173182
  39. 39. IourovI. Y.VorsanovaS. G.LiehrT.et al.2009Aneuploidy in the normal, Alzheimer’s disease and ataxia-telangiectasia brain: differential expression and pathological meaning. Neurobiology of Disease, 342May, 2009), 212220
  40. 40. JanickiS. M.MonteiroM. J.1999Presenilin overexpression arrests cells in the G1 phase of the cell cycle: arrest potentiated by the Alzheimer’s disease PS2 (N141I) Mutant. American Journal of Pathology, 1551July, 1999), 135144
  41. 41. KulnaneL. S.LehmanE. J.HockB. J.et al.2002Rapid and efficient detection of transgene homozygosity by FISH of mouse fibroblasts. Mammalian Genome, 134April, 2002), 223226
  42. 42. La FerlaF. M.GreenK. N.OddoS.2007Intracellular amyloid-β in Alzheimer’s disease. Nature Reviews Neuroscience, 87July, 2007), 499509
  43. 43. LeeH. G.CasadesusG.ZhuX.et al.2004Challenging the amyloid cascade hypothesis: senile plaques and amyloid-beta as protective adaptations to Alzheimer disease. Annals of the New York Academy of Science, 10191-4June, 2004), 14
  44. 44. LiJ.MaJ.PottreH.1995Identification and expression analysis of a potential familial Alzheimer’s disease gene on chromsome 1 related to AD3. Proceedings of the National Academy of Science of the USA, 9226December, 1995), 1218012184
  45. 45. LiJ.XuM.ZhouH.et al.1997Alzheimer presenilins in the nuclear membrane, interphase kinetochores, and centrosomes suggest a role in chromosome segregation. Cell, 905September, 1997), 917927
  46. 46. LiM.ChenL.LeeD. H.et al.2007The role of intracellular amyloid beta in Alzheimer’s disease. Progress in Neurobiology, 833October, 2007), 131139
  47. 47. LucarelliP.PiciulloA.PalmarinoM.et al.2004Association between presenilin 1-48C/T polymorphism and Down syndrome. Neuroscience Letters, 3671August, 2004), 8891
  48. 48. MattsonM. P.MagnusT.2006Ageing and neuronal vulnerability. Nature Reviews of Neuroscience, 74April, 2006), 278294
  49. 49. MaurerK.HoyerS.2006Alzheimer revisited: differences in origin of the disease carrying his name. Journal of Neuronal Transmission, 11311November, 2006), 16451658
  50. 50. Mc NaughtonD.KnightW.GuerreiroR.et al.2010Duplication of amyloid precursor protein (APP), but not prion protein (PRNP) gene is a significant cause of early onset dementia in a large UK series. Neurobiology of Aging, Epub ahead of print, (December 2010), Article in Press
  51. 51. MiglioreL.BottoN.ScarpatoR.et al.1999Preferential occurrence of chromosome 21 malsegregation in peripheral blood lymphocytes of Alzheimer disease patients. Cytogenetics and Cell Genetics, 871-2n.d.), 4146
  52. 52. MiglioreL.TestaA.ScarpatoR.et al.1997Spontaneous and induced aneuploidy in peripheral blood lymphocytes of patients with Alzheimer’s disease. Human Genetics, 1013December, 1997), 299305
  53. 53. MiglioreL.BoniG.BernardiniF.et al.2006Susceptibility to chromosome malsegregation in lymphocytes of women who had a Down syndrome child in young age. Neurobiology of Aging, 275May, 2006), 710716
  54. 54. MiglioreL.MigheliF.CoppedéF.2009Susceptibility to aneuploidy in young mothers of Down syndrome children. ScientificWorldJournal, 9October, 2009), 10521060
  55. 55. MoschB.MorawskiM.MittagA.et al.2007Aneuploidy and DNA replication in the normal human brain and Alzheimer’s disease. Journal of Neuroscience, 2726June, 2007), 68596867
  56. 56. NagyZ.2005The last neuronal division: a unifying hypothesis for the pathogenesis of Alzheimer’s disease. Journal of Cellular and Molecular Medicine, 93pp. (July-September, 2005), 531 EOF541 EOF
  57. 57. NagyZ.EsiriM. M.CatoA. M.et al.1997Cell cycle markers in the hippocampus in Alzheimer’s disease. Acta Neuropathologica, 941July, 1997), 615
  58. 58. NeveR. L.FinchE. A.DawesL. R.1988Expression of the Alzheimer amyloid precursor gene transcripts in the human brain. Neuron, 18October, 1988), 669677
  59. 59. NizzariM.VeneziaV.BianchiniP.et al.2007Amyloid precursor protein and presenilin 1 interaction studied by FRET in human H4 cells. Annals of the New York Academy of Science, 1096January, 2007), 249257
  60. 60. ObrenovicM. E.RainaA. K.OgawaO.et al.2003Alzheimer’s disease- a new begining, or final exit? In: Cell Cycle Mechanisms and Neuronal Cell death, Copani, A. & Nicoletti, F. (eds.), 7993Landes Bioscience, Austin, TX
  61. 61. OlsonM. I.ShawC. M.1969Presenile dementia and Alzheimer’s disease in mongolism. Brain, 921March, 1969), 147156
  62. 62. PadmanabhanJ.LevyM.PottreH.2011Cell cycle-dependent phosphorylation, distribution, and processing of amyloid precursor protein (In submission).
  63. 63. PettersonD.GardinerK.Kao-TF.et al.1988Mapping of the gene encoding the β-amyloid precursor protein and its relationship to the Down syndrome region of chromosome 21. Proceedings of the National Academy of Science of the USA, 8521November, 1988), 8266-8270
  64. 64. PopeW. B.LambertM. P.LeypoldB.et al.1994Microtubule-associated protein tau is hyperphosphorylated during mitosis in the human neuroblastoma cell line SH-SY5Y. Experimental Neurology, 1262April, 1994), 185194
  65. 65. PotterH.1991Review and hypothesis: Alzheimer disease and Down syndrome chromosome 21 nondisjunction may underlie both disorders. American Journal of Human Genetics, 486June, 1991), 11921200
  66. 66. PotterH.2004Cell cycle and chromosome segregation defects in Alzheimer’s disease. In: Cell Cycle Mechanisms and Neuronal Cell Death, Copani, A. & Nicoletti, F. (eds.), 5578Landes Bioscience, Austin, TX
  67. 67. PotterH.2008Down syndrome and Alzheimer’s disease: two sides of the same coin. Future Neurology, 31n.d.), 2937
  68. 68. PotterH.MaDasJ.etS.al1995Beyond β-protein: New steps in the pathogenic pathway to Alzheimer’s disease. In: Research Advances in Alzheimer’s Disease and Related Disorders, Iqbal, K., Mortimer, J. A., Winblad, B. & Wisniewski, H. M. (eds), 643654John Wiley and Sons Ltd, New York
  69. 69. PotterH.WefesI. M.NilssonL. N. G.2001The inflammation-induced pathological chaperones ACT and apoE are necessary catalysts of Alzheimer amyloid formation. Neurobiology of Aging, 226November, 2001), 923930
  70. 70. RakicP.1985Limits of neurogenesis in primates. Science, 2274690March, 1985), 10541056
  71. 71. RapoportM.DawsonH. N.BinderL. I.et al.2002Tau is essential to beta-amyloid-induced neurotoxicity. Proceedings of the National Academy of Science of the USA, 999April, 2002), 63646369
  72. 72. RiedT.1998Interphase cytogenetics and its role in molecular diagnostics of solid tumors. American Journal of Pathology, 1522325 EOF7 EOF
  73. 73. RingmanJ. M.RaoP. N.LuP. H.et al.2008Mosaicism for trisomy 21 in a patient with young-onset dementia: a case report and brief literature review. Archives of Neurology, 653March, 2008), 412415
  74. 74. RogaevE. I.SherringtonR.RogaevaE. A.et al.1995Familial Alzheimer’s disease in kindreds with missense mutations in a gene on chromsome 1 related to the Alzheimer’s disease type 3 gene. Nature, 3766543August, 1995), 775778
  75. 75. RoweI. F.RidlerM. A. C.GibberdF. B.et al.1989Presenile dementia associated with mosaic trisomy 21 in a patient with a Down syndrome child. Lancet, 28656July, 1989), 229
  76. 76. Rovelet-LecruxA.HannequinD.RauxG.et al.2006APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nature Genetics, 381January, 2006), 1112
  77. 77. ScheffS. W.PriceD. A.SchmittF. A.MufsonE. J.2006Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiology of Aging, 2710October, 2006), 13721384
  78. 78. SchupfN.KapellD.LeeJ. H.OttmanR.MayeuxR.1994Increased risk of Alzheimer’s disease in mothers of adults with Down’s syndrome. Lancet, 3448919August, 1994), 353356
  79. 79. SchumpfB.KapellD.NightingaleJ. H.LeeJ.MohlenhoffBewley. S.et al.2001Specificity of the fivefold increase in AD in mothers of adults with Down syndrome. Neurology, 576September, 2001), 979-984
  80. 80. ScottW. K.GrowdonJ. H.RosesA. D.et al.1996Presenilin-1 polymorphism and Alzheimer’s disease. Lancet, 3479009April, 1996), 11861187
  81. 81. SelkoeD. J.1999Translating cell biology into therapeutic advances in Alzheimer’s disease. Nature,3996738June, 1999), A23A31
  82. 82. SelkoeD. J.2001Alzheimer’s disease: genes, proteins, and therapy. Physiological Reviews,812April, 2001), 742760
  83. 83. SelkoeD. J.2004Cell biology of protein misfolding: the examples of Alzheimer’s and
  84. 84. Parkinson’sdisease.NatureCell.BiologyVol. .ppNo. 1.November.200410541061
  85. 85. SmallS.A.DuffK.2008Linking Abeta and tau in late-onset Alzheimer’s disease: a dual pathway hypothesis. Neuron, 604November, 2008), 534542
  86. 86. SmallB. J.FratiglioniL.ViitanenM.et al.2000The course of cognitive impairment in preclinical Alzheimer’s disease. Three- and 6-year follow-up of a population based sample. Archives of Neurology, 576June, 2000), 839844
  87. 87. SleegersK.BrouwersN.GijselinckI.et al.2006APP duplication is sufficient to cause early onset Alzheimer’s dementia with cerebral amyloid angiopathy. Brain, 12911November, 2006), 29772983
  88. 88. Spremo-PotparevićB.ŽivkovićL.DjelićN.et al.2008Premature centomere division of the X chromosome in neurons in Alzheimer’s disease. Journal of Neurochemistry, 1065September, 2008), 22182223
  89. 89. StokinG. B.GoldsteinS. B.2006Axonal transport and Alzheimer’s disease. Annual Reviews of Biochemistry, 75607-27n.d), 607627
  90. 90. StokinG. B.LilloC.FalzoneT. L.BruschR. G.RockensteinE.et al.2005Axonopathy and transport deficits early in the pathogenesis of Alzheimer’s disease. Science, 3075713February, 2005), 12821288
  91. 91. SuzukiN.CheungT. T.CaiX.D.et al.1994An increased percentage of long amyloid β-protein by familial amyloid β-protein precursor (βAPP717) mutants. Science, 2645163May, 1994), 13361340
  92. 92. SuzukiT.OishiM.MarshakD. R.et al.1994Cell cycle-dependent regulation of the phosphorylation and metabolism of the Alzheimer amyloid precursor protein. The EMBO Journal 135March, 1994), 11141122
  93. 93. TanziR. E.BertramL.2001New frontiers in Alzheimer’s disease genetics. Neuron, 322October, 2001), 181184
  94. 94. TanziR. E.GusellaJ. F.WatkinsP. C.et al.1987Amyloid β-protein gene cDNA, mRNA distributions, and genetic linkage near the Alzheimer locus. Science, 2354791February, 1987), 880884
  95. 95. TellerJ. K.RussoC.de BuskL. M.et al.1996Presence of soluble amyloid β-peptide precedes amyloid plaque fromation in Down syndrome. Nature Medicine, 21January, 1996), 9395
  96. 96. ThomasP.FenechM.2008Chromosome 17 and 21 aneuploidy in buccal cells is increased with ageing and in Alzheimer’s disease. Mutagenesis, 231January, 2008), 5765
  97. 97. TrippiF.BottoN.ScarpatoR.et al.2001Spontaneous and induced chromosomal damage in somatic cells of sporadic and familial Alzheimer’s disease patients. Mutagenesis, 164July, 2001), 321-327
  98. 98. VarvelN. H.BhaskarK.PatilA. R.PimplikarS. W.HerrupK.LambB. T.2008Abeta oligomers induce neuronal cell cycle events in Alzheimer’s disease. Journal of Neuroscience, 2843October, 2008), 1078610793
  99. 99. VincentI.RosadoM.DaviesP.1996Mitotic mechanisms in Alzheimer’s Disease? Journal of Cell Biology, 1323February, 1996), 423425
  100. 100. VitoP.WolozinB.GanjeiJ. K.et al.1996Requirement of the familial Alzheimer’s disease gene PS2 for apoptosis. Opposing effect of ALG-3. Journal of Biological Chemistry, 27149December, 1996), 3102531028
  101. 101. WahrleS.DasP.NyborgA. C.et al.2002Cholesterol dependent gamma-secretase activity is buoyant in cholesterol rich membrane microdomains. Neurobiology of Disease, 91February, 2002), 1123
  102. 102. WisniewskiH. M.RabeA.WisniewskiK. E.1988Neuropathology and dementia in people with Down’s syndrome. In: Molecular Neuropathology of Aging, Davies, P. & Finch, C. (eds.), 399413Cold Spring Harbor Laboratory, New York, USA
  103. 103. WolfeM. S.2003The secretases of Alzheimer’s disease. Current Topics in Developmental Biology, 54233-61n.d.), 233261
  104. 104. WolozinB.IwasakiK.VitoP.et al.1996Participation of presenilin 2 in apoptosis: enhanced basal activity conferred by an Alzheimer mutation. Science; 2745293December, 1996), 17101713
  105. 105. WraggM.HuttonM.TalbotC.1996Genetic association between intronic polymorphism in presenilin-1 gene and late-onset Alzheimer’s disease. Alzheimer’s Disease Collaborative Group. Lancet, 3479000February, 1996), 509512
  106. 106. YangY.GeldmacherD. S.HerrupK.2001DNA replication precedes neuronal cell death in Alzheimer’s disease. Journal of Neuroscience, 218April, 2001), 26612668
  107. 107. YangY.HerrupK.2007Cell division in the CNS: Protective response or lethal event in post-mitotic neurons? Biochimica et Biophysica Acta, 17724April, 2007), 457-466
  108. 108. YangY.VarvelN. H.LambB. T.HerrupK.2006Ectopic cell cycle events link human Alzheimer’s disease and amyloid precursor protein transgenic mouse models. Journal of Neuroscience, 263January, 2006), 775784
  109. 109. ZitnikG.WangL.MartinG. M.et al.2006Localization of eddogenous APP/APP-proteolytic products are consistent with microtubular transport. Journal of Molecular Neuroscience, 311n.d.), 5968

Written By

Antoneta Granic and Huntington Potter

Submitted: 08 April 2011 Published: 29 August 2011