Skip to main content
Aging Cell logoLink to Aging Cell
. 2020 Jan 25;19(3):e13109. doi: 10.1111/acel.13109

“Amyloid‐beta accumulation cycle” as a prevention and/or therapy target for Alzheimer's disease

Chinthalapally V Rao 1,, Adam S Asch 2, Daniel J J Carr 3, Hiroshi Y Yamada 1,
PMCID: PMC7059149  PMID: 31981470

Abstract

The cell cycle and its regulators are validated targets for cancer drugs. Reagents that target cells in a specific cell cycle phase (e.g., antimitotics or DNA synthesis inhibitors/replication stress inducers) have demonstrated success as broad‐spectrum anticancer drugs. Cyclin‐dependent kinases (CDKs) are drivers of cell cycle transitions. A CDK inhibitor, flavopiridol/alvocidib, is an FDA‐approved drug for acute myeloid leukemia. Alzheimer's disease (AD) is another serious issue in contemporary medicine. The cause of AD remains elusive, although a critical role of latent amyloid‐beta accumulation has emerged. Existing AD drug research and development targets include amyloid, amyloid metabolism/catabolism, tau, inflammation, cholesterol, the cholinergic system, and other neurotransmitters. However, none have been validated as therapeutically effective targets. Recent reports from AD‐omics and preclinical animal models provided data supporting the long‐standing notion that cell cycle progression and/or mitosis may be a valid target for AD prevention and/or therapy. This review will summarize the recent developments in AD research: (a) Mitotic re‐entry, leading to the “amyloid‐beta accumulation cycle,” may be a prerequisite for amyloid‐beta accumulation and AD pathology development; (b) AD‐associated pathogens can cause cell cycle errors; (c) thirteen among 37 human AD genetic risk genes may be functionally involved in the cell cycle and/or mitosis; and (d) preclinical AD mouse models treated with CDK inhibitor showed improvements in cognitive/behavioral symptoms. If the “amyloid‐beta accumulation cycle is an AD drug target” concept is proven, repurposing of cancer drugs may emerge as a new, fast‐track approach for AD management in the clinic setting.

Keywords: Alzheimer's disease (AD), amyloid‐beta (Aβ), brain, cell cycle, chromosome instability (CIN), cohesinopathy, cyclin‐dependent kinase (CDK) inhibitor, mitosis, mouse, Shugoshin 1 (Sgo1)


“Amyloid‐beta accumulation cycle”. Amyloid‐beta accumulation occurs in early latent phase of Alzheimer's disease, triggering its pathology development, yet the mechanism has been elusive. This integrated hypothesis explains that amyloid‐beta accumulation occurs as a vicious cycle and offers insights on how the cycle may be interfered by cell cycle‐ or mitosis‐targeting drugs.

graphic file with name ACEL-19-e13109-g003.jpg

1. ALZHEIMER'S DISEASE REPRESENTS A MAJOR ISSUE IN CONTEMPORARY MEDICINE

Alzheimer's disease (AD) is a progressive, lethal, and incurable disease. While 3%–4% of AD is early‐onset, in which patients show cognitive symptoms in their 40s‐50s, over 96% of AD is late‐onset, in which symptoms (most notoriously, memory loss and cognitive/behavior issues) first appear at age 65 or older. In late‐onset cases, the patients live an average of four to eight years after diagnosis. As AD progresses, the patients gradually lose various social and physical functions, making disease management financially costly and emotionally burdensome for patients and caregivers. However, there has been no clinically effective medicine for AD therapy. Current FDA‐approved AD medications, such as donepezil/Aricept (central acetylcholinesterase inhibitor) and memantine (N‐methyl‐D‐aspartate [NMDA] receptor antagonist), are neuronal function modulators and can temporarily alleviate AD symptoms. These drugs do not address the pathological cause of AD and therefore do not cure AD. Unmet needs for AD drugs remain. Millions of individuals are affected by AD; for example, in the United States, 5.8 million people are living with AD in 2019 (https://www.alz.org/alzheimers-dementia/facts-figures; accessed 8/2/2019). The number of patients with AD is predicted to increase in upcoming decades, making AD an urgent issue in contemporary health care.

2. COMPLEX AD PATHOLOGY LEADS TO TWO MAJOR LATE‐STAGE AD BRAIN PATHOLOGIES: AMYLOID‐BETA “PLAQUES” AND TAU/P‐TAU NEUROFIBRILLARY “TANGLES”

Alzheimer's disease development is accompanied by various pathological markers and events: cellular/intracellular amyloid‐beta accumulation, amyloid‐beta plaque and phospho‐tau tangle pathology, synaptic degeneration, cognitive impairment, and abnormalities in other cellular biomarkers, such as functions of the oxidative stress pathway and mitochondria. Dissecting the order of these pathology and events is critical to identifying AD drug targets (Figure 1; “Complex Pathology of AD”). Among these major pathological changes in AD brains are (a) senile “plaques” composed of an accumulation of amyloid‐beta and (b) neurofibrillary “tangles” that are mainly made of phosphorylated tau (Iqbal & Grundke‐Iqbal, 2008). Accumulated amyloid‐beta is neurotoxic and is widely considered to be the causal protein for AD development, that is, the amyloid‐beta hypothesis (Hardy & Higgins, 1992; Hardy & Selkoe, 2002; Selkoe, 2001) and its successor, the amyloid‐beta oligomer hypothesis (e.g., Cline, Bicca, Viola, & Klein, 2018). Soluble amyloid‐beta 1‐42 can form oligomers that cause synaptic failure, which leads to cognitive impairment and behavioral issues (Coleman, Federoff, & Kurlan, 2004; Klein, Stine, & Teplow, 2004). Thus, amyloid‐beta accumulation with oligomer formation is considered the triggering event for AD development, leading to cascades of events, pathway activations, and other pathologies. The amount of amyloid‐beta is a result of the balance between generation and catabolism; thus, pathways involved in amyloid‐beta generation and catabolism have been of major research interest and are candidates for AD drug targeting (Hardy & Selkoe, 2002; Selkoe & Hardy, 2016).

Figure 1.

Figure 1

“Complex Pathology of AD”. In the clinic, AD is usually diagnosed with cognitive–behavioral symptoms and verified with brain pathology (i.e., brain scan for amyloid‐beta or observed brain shrinkage). Underlying presymptomatic latent pathologies precede the clinical symptoms by decades. Late‐stage AD patients' brains indicate amyloid‐beta plaques, p‐tau tangles, neurodegeneration, neuroinflammation, and brain shrinkage as widespread pathological traits. Understanding cellular/organ mechanisms driving the development of these pathologies (latent processes) is critical to developing drugs for AD intervention and/or therapy. Amyloid‐beta accumulation is considered a key trigger for AD pathology: (a) pathological amyloid‐beta accumulation precedes p‐tau tangles by 10–15 years (Perrin et al., 2009); (b) oligomeric amyloid‐beta is neurotoxic and can impair neuronal functions (Cline et al., 2018); and (c) amyloid‐beta catabolism is influenced by aging, stress, sleep, and glial cell functions, which is consistent with the fact that 96% of AD is late‐onset/age‐associated (see Text). Amyloid‐beta accumulation is considered a result of balance between amyloid‐beta generation (increased by stress, neuronal activity) and catabolism (which is reduced over age, helped by sleep). The mechanistic question of how amyloid‐beta begins to accumulate in middle age is a critical question

Amyloid‐beta is generated from the larger precursor protein amyloid precursor protein (APP) through proteolysis. Proteolytic enzymes involved in amyloid‐beta generation (i.e., the amyloidogenic pathway) include beta‐secretase (BACE) and gamma‐secretase complex (Moussa‐Pacha, Abdin, Omar, Alniss, & Al‐Tel, 2019; Penke, Bogár, & Fülöp, 2017). Amyloid‐beta can be removed through a proteolytic catabolism pathway. Sikanyika, Parkington, Smith, and Kuruppu (2019) listed 10 proteases that can catabolize Aβ1‐42. These proteases include neprilysin, endothelin converting enzyme 1, and insulin‐degrading enzyme. As the APOE4 variant increases the abundance of Aβ and risk of AD, an APOE‐associated cholesterol pathway plays a role in Aβ dyshomeostasis (Selkoe, 2001; Yamazaki, Zhao, Caulfield, Liu, & Bu, 2019). Plaques are usually surrounded by glial cells, which play a role in immunological removal of plaques (Dong, Li, Cheng, & Hou, 2019). Hence, major rational targets for AD drug R&D have been (a) amyloid‐beta itself via an immunotherapeutic approach, (b) amyloidogenic proteases, and (c) amyloid‐beta catabolism. However, these ongoing approaches have not shown success in the clinic yet. In translational studies with mouse models, a newer approach targeting amyloid‐beta oligomer has shown promise. In 6‐ to 7‐month‐old Tg 5xFAD mice, injection of an Aβ oligomer‐specific antibody rescued memory performance (Cline et al., 2018). Monomeric Aβ‐binding peptide RD2, which renders Aβ unable to form oligomer, reversed AD symptoms (cognition, behavior, and Aβ plaque loads) in aged APP/PS1 mice, indicating its potential for therapeutic application (Schemmert et al., 2019).

In late‐onset AD, amyloid‐beta accumulation and senile plaque development begin in middle age. At the early stage, no immediate visible symptoms appear, and the time window is considered presymptomatic or latent. Ten to fifteen years later, neurofibrillary tangles begin to emerge. However, there is also a 3 to 5‐year time window without notable memory or cognitive symptoms (Perrin, Fagan, & Holtzman, 2009). In a later stage of AD, memory/cognitive/behavioral symptoms and amyloid‐beta plaques and neurofibrillary tangles are observed. Notably, plaques and tangles in the AD brain appear with differential localizations; plaques are diffusely distributed over the entire cortex, while tau tangles are seen in brain regions related to clinical symptoms and overlap with areas of hypometabolism (Dronse et al., 2017). As AD progresses, both lesions spread throughout the brain, each following a stereotyped pattern. Senile plaques spread from association cortices (Thal phase 1) to allocortical areas, including the hippocampus (Thal phase 2); then to diencephalic nuclei, striatum, and cholinergic nuclei of the basal forebrain areas (Thal phase 3); several brain stem nuclei (Thal phase 4); and cerebellum (Thal phase 5) (Thal, Rüb, Orantes, & Braak, 2002). Neurofibrillary tangles first appear in the entorhinal cortex (Braak stages I/II), then spread toward limbic structures, including the hippocampus (Braak stages III/IV) and association cortices (Braak stages V/VI) (Braak & Braak, 1991). Additionally, amyloid‐beta accumulation around blood vessels, also known as cerebral amyloid angiopathy, appears.

Cerebral amyloid angiopathy may play a role in cognitive/behavioral symptoms via cerebrovascular system dysfunction (Weber, Patel, & Lutsep, 2018). This complex pathology led to a controversy regarding which pathology (amyloid‐beta plaques, tau/p‐tau tangles, or cerebral amyloid angiopathy) is more critical in AD symptoms and therefore represents a better therapeutic target. As the decades‐long development of AD pathology is associated or concurrent with many biological events and general aging, it has been difficult to pinpoint an effective intervention or therapeutic target. As approaches targeting late‐stage AD pathologies have not been successful, and as early amyloid‐beta accumulation with oligomer formation has begun to be recognized as a critical step with the amyloid‐beta oligomer hypothesis, the need to understand mechanisms leading to amyloid‐beta accumulation is increasing.

3. CURRENT AD DRUG RESEARCH AND DEVELOPMENT TARGETS

Mainstream AD drug research and development efforts have been directed toward (i) components directly involved in AD pathology, such as amyloid‐beta or p‐tau, amyloidogenic proteases, and other amyloid‐beta or tau binding proteins, such as receptor for advanced glycation endproducts (RAGE); (ii) components associated with pathology and predicted to be involved in symptoms, such as neuroinflammation, oxidative stress, and mitochondrial dysfunction; and (iii) medicines that can ease cognitive/behavioral symptoms of AD, including neuronal function modulators and neurotransmitters, although they may be palliative and may not address pathological causes nor lead to fundamental cure.

As of 30 October 2019, the https://Clinicaltrials.gov database lists 2,214 clinical trials involved in AD (https://clinicaltrials.gov/). The Alzforum website, which maintains a database on potential dementia therapeutics, lists 216 therapeutic reagents for AD and mild cognitive impairment (MCI) (https://www.alzforum.org/therapeutics). However, only five reagents (donepezil, galantamine, memantine, rivastigmine, and tacrine) are approved by the US FDA for use as symptomatic relievers of AD, targeting the cholinergic system and other neurotransmitters. No other agent has been validated as a clinically effective target.

4. INTRINSIC AND EXTRINSIC FACTORS THAT AFFECT AMYLOID‐BETA AMOUNT

The earliest step of AD pathology development is cerebral accumulation of amyloid‐beta, the mechanism of which has been elusive. A majority of sporadic AD patients do not carry mutation in familial AD genes (i.e., APP, PSEN1, or PSEN2) (Lanoiselée et al., 2017), suggesting that other drivers are involved in the development of sporadic late‐onset AD cases. Here, we discuss amyloid‐beta accumulation as a result of the disturbed balance between Aβ generation and catabolism (i.e., Aβ dyshomeostasis). Consistently, AD patients indicated impaired clearance rates for Aβ42 and Aβ40, compared with controls (Mawuenyega et al., 2010).

Various intrinsic factors can affect Aβ generation and/or catabolism. Amyloid‐beta can be generated through intensive neuronal activity (i.e., activity‐dependent Aβ generation) (Ovsepian & O'Leary, 2016). Activation of and/or increase in amyloidogenic proteases can play a role in increasing Aβ, as observed in patients with Down syndrome who exhibit early AD‐like dementia and express a large amount of BACE1 (Miners, Morris, Love, & Kehoe, 2011). Systemic factors, such as the effect of oral and gut microbes on systemic inflammation, are also gathering interest (Tremlett, Bauer, Appel‐Cresswell, Finlay, & Waubant, 2017). Sleep affects accumulation and removal of Aβ (Cordone, Annarumma, Rossini, & Gennaro, 2019). Reports indicate that Aβ catabolism decreases over age. Older mice showed a 40% decline in clearance of injected Aβ, which may be caused by a decline in the efficiency of exchange between the subarachnoid cerebrospinal fluid and the brain parenchyma (Kress et al., 2014). Levels of neprilysin, an Aβ‐degrading protease, decreased with age in both normal and AD patients. This decreasing neprilysin level may act as a trigger for AD (Hellström‐Lindahl, Ravid, & Nordberg, 2008). In APP‐SL70 mice, the microglial response to increasing amyloid‐beta was estimated to be overwhelmed with aging (Blume et al., 2018).

In addition to intrinsic factors, extrinsic or environmental factors can play a role. In a mouse model, recurrent activation of brain herpes simplex virus 1 (HSV1) infection led to amyloid‐beta accumulation and other AD pathology (tau phosphorylation, neuroinflammation) (De Chiara et al., 2019), indicating that viral infection can trigger Aβ accumulation and AD pathology. In an AD‐omics study, HHV‐6A and HHV‐7 were identified as prominently associated with human AD across three independent cohorts (Readhead et al., 2018). Reports like these support the theory that pathogens trigger AD (Haas & Lathe, 2018; Itzhaki, 2014; Sochocka, Zwolińska, & Leszek, 2017), as well as the role of amyloid‐beta as a protection mechanism against viral infection (Li, Liu, Zheng, & Huang, 2018). Although AD cannot be completely explained by the pathogen theory alone, pathogens may act as a risk factor or have an impact on a segment of patients with AD. A high rate of HSV1 and other infections was observed in AD patients (Sochocka et al., 2017). Since three subtypes of AD were identified based on the spread of neurofibrillary tangles (Murray et al., 2011), there may be more than one causal process, leading to distinct pathology of AD subtypes.

5. A BRIEF HISTORY OF EARLIER STUDIES OF ANEUPLOIDY AND AD

Since the 1990s, a long‐standing theory has purported that aneuploidy plays a critical role in AD development. In an early thesis noting AD‐like dementia in patients with Down syndrome, Potter (1991) hypothesized that (i) aneuploidy (chromosome 21 trisomy) is causal to AD, and that (ii) genes associated with the risk of AD would be involved in the cell cycle, and such genes would lead to the development of aneuploidy when mutated (Potter, 1991). Following hypothesis (i), the link between aneuploidy and AD was explored using cytogenetics. Earlier studies tested the rate of aneuploidy in peripheral blood lymphocytes and fibroblasts from AD patients. Cells from familial and sporadic AD patients were shown to have more micronuclei than controls. The antifungal drug griseofulvin mitigated the increase in micronuclei in cells from patients with AD, indicating an altered response to genotoxic challenge (Petrozzi et al., 2002; Trippi et al., 2002). These results may be attributed to increased DNA damage and impaired DNA repair (Coppedè & Migliore, 2009), or perhaps to DNA replication stress (Yurov, Vorsanova, & Iourov, 2011). Newer studies indicated a higher aneuploidy rate in AD‐affected neurons (Iourov, Vorsanova, Liehr, & Yurov, 2009). In addition, the presence of hyperploid neurons was noted (Arendt, Brückner, Mosch, & Lösche, 2010) [see Section 6.3 “High degree of aneuploidy in patients with AD and mild cognitive impairment (MCI)” for additional references]. Although a single‐cell sequencing report noted conflicting results (van den Bos et al., 2016), and although reported rates of aneuploidy vary widely, collectively there seems to be solid support for increased aneuploidy in somatic cells and neurons of patients with AD. Supporting evidence for hypothesis (ii) includes reports that gene mutations causal to neurodegenerative diseases (APP, MAPT, PSEN for AD [see later sections]; Niemann–Pick C; Granic & Potter, 2013) also cause aneuploidy in neurons. In a later section, “Thirteen among 37 genes on the human AD genetic risk loci are functionally involved in the cell cycle and/or mitosis” (Table 1), we will discuss newer corroborating evidence from contemporary AD‐omics.

Table 1.

Thirteen among 37 genes on human AD genetic risk loci are functionally involved in the cell cycle and/or mitosis

Gene Name Full Name Proposed function (s) and pathway(s) involved* “Cell cycle”/total publications** “Mitosis”/total publications** Reported involvement in cell cycle and/or mitosis (including possible link)
ABCA7 ATP‐binding cassette subfamily A member 7

Transporter,

Lipid metabolism/

homeostasis,

Ceramide transport

2/228 1/228 Ovarian cancer metastasis marker candidate (Elsnerova et al., 2017)
ABI3 ABI family member 3 Inhibits ectopic metastasis of tumor cells and cell migration 13/406 1/406

Tumor suppressor,

Expression reduces growth and induces senescence (Latini et al., 2011)

ACE Angiotensin I‐converting enzyme Generates angiotensin II, a potent vasopressor and aldosterone‐stimulating peptide that controls blood pressure and fluid–electrolyte balance 126/13779 6/13779 Inhibition accelerates endothelial regrowth (Van Belle et al., 1997)
ADAM10 ADAM metallopeptidase domain 10

Metalloproteinase,

Cleaves the membrane‐bound precursor of TNF‐alpha to its mature soluble form,

Cleaves several other cell‐surface proteins (e.g., ephrin‐A2, CD44, CDH2, Notch)

32/1364 1/1364

Silencing inhibits the in vitro and in vivo growth of hepatocellular carcinoma cells (Liu, Zhang, Liu, Ji, & Wang, 2015),

Constitutive activation promotes cell growth and activates the TNF‐α/NFκB pathway in mantle cell lymphoma (Armanious, Gelebart, Anand, Belch, & Lai, 2011)

ADAMTS4 ADAM metallopeptidase with thrombospondin type 1 motif 4

ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) protein family,

Cleaves aggrecan, a cartilage proteoglycan, and may be involved in its turnover

6/578 0/578  
ALPK2 Alpha kinase 2 Kinase that recognizes phosphorylation sites in which the surrounding peptides have an alpha‐helical conformation 0/13 0/13  
APH1B Aph−1 homolog B, gamma‐secretase subunit Functional component of the gamma‐secretase complex, which also contains presenilin and nicastrin, a subunit of APP protease complex 1/37 0/37  
BIN1 Bridging integrator 1 Nucleocytoplasmic adaptor, synaptic vesicle endocytosis, cardiac muscle development 35/413 0/413

Tumor suppressor (Pan et al., 2012),

A corepressor of the transcription factor E2F1, Inhibits cell cycle progression (Folk et al., 2019), Regulates fas/fas ligand‐mediated apoptosis (Esmailzadeh, Huang, Su, Zhou, & Jiang, 2015), Accumulates adjacent to amyloid deposits in vivo (De Rossi et al., 2019)

BZRAP1‐AS1 BZRAP1 antisense RNA 1

Noncoding RNA,

Promoters and enhancers for TSPOAP1‐AS1 gene

0/3 0/3  
CASS4 Cas scaffold protein family member 4 Docking protein that plays a role in tyrosine kinase‐based signaling, related to cell adhesion and cell spreading 3/25 0/25  
CD33 CD33 molecule Lectin of the SIGLEC (Sialic acid‐binding immunoglobulin‐like) family, cell–cell interaction, Transmembrane receptor expressed on cells of myeloid lineage 241/3168 4/3168

Marker for acute myeloid leukemia subtype, CD33 inhibition in myeloid cells causes apoptosis (Mingari, Vitale, Romagnani, Falco, & Moretta, 2001),

Anti‐CD33 antibody conjugates are being tested for CD33 + AML in clinic (Kobayashi et al., 2009).

CD2AP CD2‐associated protein Scaffolding molecule that regulates the actin cytoskeleton, receptor endocytosis, and cytokinesis 17/403 1/403

Involved in cytokinesis

(Monzo et al., 2005)

CELF1 CUGBP Elav‐like family member 1

May regulate pre‐mRNA alternative splicing, mRNA editing, and translation;

May be a specific regulator of miRNA biogenesis;

Defects affect myotonic dystrophy via RNA toxicity

21/234 0/234

Upregulated in glioma,

Promotes glioma cell proliferation by suppression of CDKN1B (Xia et al., 2015)

CLNK Cytokine‐dependent hematopoietic cell linker Regulation of immunoreceptor signaling 0/22 0/22  
CLU

Clusterin

(aka. apolipoprotein J)

A chaperon (secreted and cytosolic),

Inhibits formation of amyloid fibrils,

Involved in cell death, tumor progression, and neurodegenerative disorders

119/1759 7/1759

CLU OP activates PI3K/AKT pathway, overrides Cr(VI)‐induced senescence in hepatocytes (Zhang, Zhang, Xiao, Zhong, and Xiao, 2019),

CLU knockdown sensitizes cancer cells to chemotherapy drugs (Al Nakouzi et al., 2016),

Nuclear CLU is pro‐apoptotic (Shannan, Seifert, Boothman, Tilgen, & Reichrath, 2006),

Secretory CLU is pro‐survival,

High levels of sCLU caused G1 cell cycle arrest in distinct cell types (Yu & Tan, 2012)

CNTNAP2 Contactin‐associated protein‐like 2 Cell adhesion molecules and receptors in nervous system 3/395 0/395  
CR1 Complement C3b/C4b receptor 1

Membrane immune adherence receptor,

Belongs to the receptors involved in complement activation,

Captures and clears complement‐opsonized pathogens by erythrocytes and monocytes/macrophage

88/3263 5/3263  
DSG2 Desmoglein 2 Calcium‐binding transmembrane glycoprotein components of desmosomes and cell–cell junctions 13/310 0/310

Overproduction is poor prognostic marker for HCC (Han et al., 2018),

Knockdown arrests NSCLC cells via CDK2 decrease and p27 increase (Cai et al., 2017)

ECHDC3 Enoyl‐CoA hydratase domain‐containing 3 Fatty acid biosynthesis 0/12 0/12  
EPHA1 EPH receptor A1

Ephrin receptor subfamily of the protein tyrosine kinase,

Nervous system development,

Contact‐dependent bidirectional signaling into neighboring cells

16/246 1/246

Negative regulator of the Ras/MAPK pathway (Miao et al., 2001), exerts antimitogenic functions in a cell‐type‐specific manner,

Knockdown of EPHA1 in ovarian cancer cells inhibited their aggressive traits

FERMT2 Fermitin family member 2

Scaffolding protein,

Enhances integrin‐mediated cell adhesion onto the extracellular matrix and cell spreading,

Binds to membranes enriched in phosphoinositides,

the assembly of focal adhesions

4/64 0/64 Inhibited by Wnt/beta‐catenin, resulting in blockade of myoblast fusion in myoblasts (Suzuki, Pelikan, & Iwata, 2015)
HESX1 HESX homeobox 1 Conserved homeobox protein that is a transcriptional repressor in the developing forebrain and pituitary gland 3/235 0/235  

HLA‐DRB5,

HLA‐DRB1

Major histocompatibility complex, class II, DR beta 5, and DR beta 1

Plays a central role in the immune system by presenting peptides derived from extracellular proteins

0/464 (DRB5)

60/9178 (DRB1)

0/464 (DRB5)

0/9178 (DRB1)

 
INPP5D Inositol polyphosphate−5‐phosphatase D

Hydrolyzes the 5' phosphate from phosphatidylinositol (3,4,5)‐trisphosphate and inositol 1,3,4,5‐tetrakisphosphate, negatively regulating the PI3K (phosphoinositide 3‐kinase) pathways,

Negative regulator of myeloid cell proliferation and survival

7/288 0/288 Overexpression suppressed cell growth, migration, and invasion in vitro and in vivo in NSCLC via PI3K pathway inhibition (Fu et al., 2019)
KAT8 Lysine acetyltransferase 8

Histone acetylase (HAT),

Chromatin architecture,

Embryonic development

32/145 3/145

Important for cancer cell survival (Zhang, Liu, et al., 2013),

Required for DNA damage response and double‐strand break repair to ionizing radiation, RNAi for Rcd1, Rcd5, or MBD‐R2 showed abnormal chromosome segregation in Drosophila (Pavlova et al., 2019)

MEF2C Myocyte enhancer factor 2C

MADS box transcription enhancer factor 2 (MEF2) family,

Controls cardiac morphogenesis and myogenesis,

Essential role in hippocampal‐dependent learning and memory by suppressing the number of excitatory synapses and thus regulating basal and evoked synaptic transmission,

Normal neuronal development

65/1122 4/1122

Regulates the expression of G2/M checkpoint genes (14‐3‐3γ, Gadd45b, and p21) and the subcellular localization of CYCLIN B1 (Badodi, Baruffaldi, Ganassi, Battini, & Molinari, 2015),

Substrate of anaphase‐promoting complex,

Expression of phosphorylation mutant can delay cell cycle in colon cancer cells,

Activates CDK inhibitor p21/CDKN1A and thus inhibits cell cycle transition (Di Giorgio, Gagliostro, Clocchiatti, & Brancolini, 2015),

Acts as effectors of neurogenesis in the brain (Li et al., 2008),

Drives B‐cell receptor (BCR)‐induced proliferation of mature B cells (Wilker et al., 2008)

MS4A6A

MS4A4E

Membrane spanning 4‐domains A6A, A4E May be involved in signal transduction as a component of a multimeric receptor complex

1/60 (MS4A6A)

1/26 (MS4A4E)

0/60 (MS4A6A)

0/26 (MS4A4E)

 
NME8 NME/NM23 family member 8

Ciliary function,

Sperm tail maturation

0/31 0/31  
NYAP1 Neuronal tyrosine‐phosphorylated phosphoinositide−3‐kinase adaptor 1

Regulates neuronal morphogenesis,

Disruption in mice affects brain size and neurite elongation

0/4 0/4  
PTK2B/PYK2 Protein tyrosine kinase 2 beta

Calcium‐induced regulation of ion channels and activation of the map kinase signaling pathway,

Member of the FAK subfamily of protein tyrosine kinases

23/457 4/457

Regulates actin cytoskeleton reorganization in fibroblasts (Du et al., 2001),

p‐PYK2 associates with the oocyte spindle and spindle poles,

May function as a component of the microtubule‐organizing center to regulate spindle assembly during the meiotic process of mouse oocytes (Meng et al., 2018),

Promotes migration and invasion of glioma cells (Lipinski et al., 2005)

SCIMP SLP adaptor and CSK interacting membrane protein

Transmembrane adapter/mediator,

Major histocompatibility complex class II signal transduction,

Immune synapse formation

0/9 0/9  
SLC24A4 Solute carrier family 24 member 4 Potassium‐dependent sodium/calcium exchanger protein family, transporter 0/84 0/84  
SORL1 Sortilin‐related receptor 1

Endocytosis and sorting,

Transmembrane signaling receptor activity and low‐density lipoprotein particle binding

4/334 2/334  
SPI1/PU.1 Spi−1 proto‐oncogene, hematopoietic transcription factor PU.1

ETS‐domain transcription factor that activates gene expression during myeloid and B‐lymphoid cell development,

Binds to the PU‐box, a lymphoid‐specific enhancer,

Differentiation or activation of macrophages or B cells

145/1961 4/1961

PU.1 is essential for mixed‐lineage leukemia (MLL) (Zhou et al., 2014),

Required for the growth of MLL leukemic cells via the promotion of cell cycle progression and inhibition of apoptosis,

Acts as tumor suppressor in myeloma (Ueno et al., 2017)

SUZ12P1 SUZ12 pseudogene 1   0/1 0/1

Prostate cancer biomarker,

Long noncoding RNA promoted the proliferation of and inhibited apoptosis of prostate cancer (Wan et al., 2016)

TREM2 Triggering receptor expressed on myeloid cells 2

Immune response,

Membrane protein that forms a receptor signaling complex with the TYRO protein

9/708 0/708

Promotes microglial survival by activating the Wnt/β‐catenin pathway (Zheng et al., 2017),

TREM2/DAP12 complex regulates inflammatory responses in microglia via the JNK signaling pathway (Zhong et al., 2017),

Overexpression enhances glioma cell proliferation and invasion (Wang et al., 2016).

Acts as a tumor suppressor via Wnt1/β‐catenin and Erk signaling in colorectal carcinoma (Kim et al., (2019)

ZCWPW1 Zinc finger CW‐type and PWWP domain‐containing 1

Function poorly known,

A “bottleneck” gene

0/32 0/32  

A list of 37 AD genetic risk loci (genes identified as frequently mutated in AD‐omics studies) was used. The number of existing publications was examined for each gene, followed by a keyword search with “Gene X and cell cycle” or “Gene X and mitosis.” The process provides an estimate for the current total research activity regarding the gene and for the gene's functional involvement in the cell cycle and/or in mitosis. Genes showing a direct or strong functional connection to the cell cycle and/or mitosis are marked in bold. Thirteen among the 37 AD genetic risk loci have indicated functions in the cell cycle and/or mitosis, suggesting the importance of the cell cycle and/or mitosis in AD development.

*

Based on GeneCards database.

**

Publication numbers as of 5 November 2019, via PubMed keyword search.

In Boveri's 19th‐century theorem, aneuploidy was predicted to cause cancer (Boveri, 2008). Aneuploidy can be caused by external genotoxic challenges (e.g., radiation, chemicals, and virus) or by an internal defect in molecular mechanisms for genome maintenance that are intimately involved in cell cycle regulations. Mainstream mechanistic studies of the cell cycle emerged rather independently from studies of AD. The conceptual framework that the cell cycle is driven by cyclin‐dependent kinases (CDKs) and cyclins emerged by the mid‐1980s/early 1990s (Nurse, 2012). During the same period, major cell cycle driving enzymes and their regulatory components were identified through combined efforts, including genetics work using budding yeast S. cerevisiae, fission yeast S. pombe, fungi A. nidulans, and fruit fly D. melanogaster; biochemistry studies using egg extracts of frog X. laevis; and cell biology studies with cultured cells (see Murray & Hunt, 1993).

Cancer, carcinogenesis, and developmental defects associated with aneuploidy have been the primary disease targets of cell cycle studies. These types of investigations were assumed to be useful for finding methods and targets to manipulate cell cycle and cell growth, which could lead researchers to cancer therapeutics. Over the years, this assumption has been shown to be correct. Many validated cancer therapeutics target machineries that are involved in the cell cycle. In the 1990s–2000s, aneuploidy‐inducing transgenic genomic instability mouse models, such as chromosome instability (CIN) models and microsatellite instability (MIN) models, were developed. The models were used mainly to assess cancer development (Rao & Yamada, 2013; Rao, Yamada, Yao, & Dai, 2009; Simon, Bakker, & Foijer, 2015) and to examine the relationship between aneuploidy and carcinogenesis (Weaver & Cleveland, 2009; Zasadil et al., 2016). However, most cancer assessment studies do not keep mice until old age. It was only recently that experimental results testing the link between genomic instability and AD in aged mouse models started to be reported (Rao, Farooqui, Asch, & Yamada, 2018).

6. THE “AMYLOID‐BETA ACCUMULATION CYCLE”

From the notion that aneuploidy plays a role in AD development, some hypotheses focusing on the role of mitotic cycle re‐entry of neurons evolved. One such hypothesis was the “two‐hit hypothesis” that proposes age and mitotic re‐entry as two key factors (“hits”) for AD development (Webber et al., 2005; Zhu, Lee, Perry, & Smith, 2007; Zhu, Raina, Perry, & Smith, 2004). Based on results from genomic instability mouse models, we proposed a version of the two‐hit hypothesis with an emphasis on the role of prolonged mitosis in accumulating amyloid‐beta, the “three‐hit hypothesis.” The “three‐hit hypothesis” proposes (I) aging, (II) mitotic re‐entry, and (III) prolonged mitosis as three key factors for the development of AD (Rao, Farooqui, Zhang, Asch, & Yamada, 2018). Recent literature led us to an integrative hypothesis, the “amyloid‐beta accumulation cycle,” incorporating interference in mitosis and the aneuploidogenic role of amyloid‐beta (Figure 2). The rationales for the “amyloid‐beta accumulation cycle” are summarized below.

Figure 2.

Figure 2

(a) The “amyloid‐beta accumulation cycle”. Normal neurons or glia are challenged by mitotic signaling, which may be associated with age and the microenvironment, such as high reactive oxygen species (ROS), reduced antioxidants, damaged blood–brain barrier, and fatigued stem cells, or other pathogenic conditions, such as diabetic wounds, pathogen infection, or mutated AD risk gene. Mitogenic signaling causes neurons or glial cells to enter the cell cycle and attempt to go through mitosis. In the cycling cells, aneuploidy, an environmental factor, other mutations in an AD risk gene, or already existing extracellular amyloid‐beta cause cells to go through prolonged mitosis or a quasi‐mitotic state with high mitotic kinase activity, when they accumulate amyloid‐beta, BACE, and p‐tau. If the state is not resolved, mitotic catastrophe occurs, and accumulated amyloid‐beta, BACE, and p‐tau are released to the microenvironment. Released amyloid‐beta, with its prion‐like properties, may function as seeds for subsequent plaque pathology. Extracellular amyloid‐beta can provoke inflammation and mitogenic signaling, and can also cause mitotic errors, prolonged mitosis, and aneuploidy. Thus, age‐ or microenvironment‐provoked mitogenic signaling can trigger a vicious cycle leading to further amyloid‐beta accumulation (the “amyloid‐beta accumulation cycle”) (blue/purple arrows). (b) Cancer drugs that target mitotic re‐entry and/or prolonged mitosis may be valid drugs for managing the “amyloid‐beta accumulation cycle” and AD. The two‐hit hypothesis (Zhu et al., 2007, 2004) proposed age and mitotic re‐entry as crucial events for development of AD pathology. In light of the apparent importance of prolonged mitosis in this process, we proposed the three‐hit hypothesis (Rao, Farooqui, Asch et al., 2018). The “amyloid‐beta accumulation cycle” is an integrated hypothesis that emerged from the three‐hit hypothesis. The “amyloid‐beta accumulation cycle” suggests that a reagent that interferes with amyloid‐beta accumulation could be an AD drug. As the cell cycle and mitosis are validated targets for cancer drugs, repurposing of cancer drugs for AD management may emerge as a viable clinical option in the near future. (c) Cerebral amyloid‐beta protein can accumulate in mice with an unmodified APP gene under certain conditions. Under normal circumstances, wild‐type mice with an unmodified APP gene do not accumulate amyloid‐beta in the brain, even in old age (24 months and older). AD modeling in mice has been dependent on introduction of transgenic mutations in genes involved in familial/early‐onset AD (e.g., APP, PSEN1, and MAPT), representing early‐onset AD models (Jankowsky & Zheng, 2017; Saito & Saido, 2018). A rodent model for sporadic late‐onset AD has been an unmet need. Over 96% of all human AD cases are late‐onset and sporadic, a majority of which carry no mutation in known early‐onset AD genes. Thus, identifying conditions under which amyloid‐beta accumulates is valuable to gain mechanistic insights on AD development and to model late‐onset AD. A progeria mouse model SAMP8 was reported to accumulate amyloid‐beta, yet the causal mutation remains unidentified (Akiguchi et al., 2017). Recent reports began to identify conditions that can cause amyloid‐beta accumulation in the mouse brain with unmodified APP or other known early‐onset AD gene mutations. Examples of amyloid‐beta accumulating conditions include (i) aged Sgo1−/+ mice, a cohesinopathy–chromosome instability mouse model (Rao, Farooqui, Zhang et al., 2018) (photo: Our Aβ IHC results from 18‐ to 24‐month‐old Sgo1−/+ mice. The magnified panel indicates extracellular “released” Aβ), and (ii) HSV1 infection (e.g., De Chiara et al., 2019). Photo: Our Aβ IHC results from HSV1‐infected 12‐month‐old C57BL/6 mice (unpublished). Uninfected mice showed no cerebral amyloid‐beta (not shown). Antibody used for IHC: Cell Signaling Technology β‐Amyloid D54D2 (cat. No. 8243). Although multiple Aβ‐specific commercial antibodies recognized the same band, the exact Aβ species accumulated in Sgo1−/+ model remain to be determined

6.1. Neuronal cell cycle re‐entry occurs in AD

Normally, neurons are terminally differentiated cells and maintain their G0 quiescent state. However, human AD brains show signs of neurons in the mitotic cycle and poly(tetra)ploid 4N cells (Herrup, 2010; Herrup & Arendt, 2002; Herrup, Neve, Ackerman, & Copani, 2004; Neve & McPhie, 2006). Consistently, human AD brains show misregulations in canonical cell cycle driving proteins (e.g., CDC25 phosphatases) and inhibitory proteins (e.g., wee1 kinase) in the direction toward mitosis; in degenerating neurons, Cdc25A and Cdc25B show higher activity, while Wee1 shows lower activity (Ding et al., 2000; Tomashevski, Husseman, Jin, Nochlin, & Vincent, 2001; Vincent et al., 2001). In a 3xTg AD mouse model and in human AD patients, hyperphosphorylated retinoblastoma protein, a marker for G1/S transition, co‐localized with hyperphosphorylated tau, linking aberrant cell cycle progression with tau pathology (Hradek et al., 2015). In the study, Hradek et al. used 19‐month‐old animals, thus leaving a question if the pRb accumulation is age‐associated. Lopes, Blurton‐Jones, Yamasaki, Agostinho, and LaFerla (2009) used two mouse models, (i) 3xTg and (ii) a tetracycline‐regulatable transgenic model of neuronal ablation (CaM/Tet‐DTA mice). Neuronal death in model (ii) did show increases in cell cycle markers, suggesting that cell death and release of cellular contents can activate mitogenic signaling. This model also displayed a significant increase in hyperphosphorylated tau and Abeta, supporting the possibility that cell cycle re‐entry may lead to AD‐like changes even in animals without a previous alteration of the genes related to Abeta or tau.

6.2. Various mitogenic/growth signaling factors are activated in the AD brain

Consistent with Section 6.1, misregulations in various mitogenic/growth signaling factors, including ERK/MAPK, GSK3, PI3K/AKT, and CDK, are reported in the AD brain (Kirouac, Rajic, Cribbs, & Padmanabhan, 2017; Monaco & Vallano, 2005; Morroni, Sita, Tarozzi, Rimondini, & Hrelia, 2016; Sun, Liu, Nguyen, & Bing, 2003; Swatton et al., 2004; Vincent, Jicha, Rosado, & Dickson, 1997). Amyloid‐beta oligomers are synaptotoxic and can activate the signaling axis that involves the tyrosine kinase ephrin receptor A4 (EphA4) and c‐Abl tyrosine kinase. When mutated, c‐Abl can act as an oncogene (Vargas, Cerpa, Muñoz, Zanlungo, & Alvarez, 2018). CDK7 can act as a CDK‐activating kinase and can activate the major Cdk–cyclin substrates. CDK7 expression is age‐dependent and is elevated in hippocampal neurons of AD patients (Zhu et al., 2000). The stress signaling AMPK pathway is also reported to be involved in AD (Caberlotto, Lauria, Nguyen, & Scotti, 2013; Mairet‐Coello & Polleux, 2014; Vingtdeux, Davies, Dickson, & Marambaud, 2011), which may demonstrate a link between energy homeostasis and AD. In a streptozotocin‐induced AD model in rats, AMPK activation by AICAR stopped the cell cycle, improved spatial memory, and ameliorated AD pathology (Du et al., 2015).

Neuroinflammation has been reported in AD. Neuroinflammation markers, such as IFN‐gamma and TNF‐alpha, can also cause cell proliferation. In human AD patients, INF‐gamma, TNF‐alpha, and nitric oxide levels were higher in mild and severe stages than in earlier phases, indicating progressive increases (Belkhelfa et al., 2014). Activated TNF‐alpha and the c‐Jun kinase (JNK) signaling pathway led neuronal cells to progress in the cell cycle toward mitosis, followed by neuronal cell death (Bhaskar et al., 2014). The expression and release of pro‐inflammatory molecules are primarily achieved through glial cells, especially by astrocytes and microglia; glial cell functions are also involved in amyloid‐beta clearance and in neurodegenerative disease conditions, including AD (Pérez‐Nievas & Serrano‐Pozo, 2019; Valles et al., 2019). Consistently, in cerebrospinal fluid, microglia activation marker (soluble triggering receptor expressed on myeloid cells 2 (sTREM)), a marker of microglial inflammatory reaction (monocyte chemoattractant protein‐1 (MCP‐1)), and astroglial activation marker (chitinase‐3‐like protein 1 (YKL‐40)) increased as AD progressed (Nordengen et al., 2019).

6.3. High degree of aneuploidy in patients with AD and mild cognitive impairment (MCI)

Human brains are naturally aneuploidogenic during early development and carry a higher rate of aneuploid cells in adulthood. However, compared with healthy subjects, patients with both familial/early‐onset and sporadic late‐onset AD show widespread aneuploidy in their brains and in peripheral blood cells (Bajic, Spremo‐Potparevic, Zivkovic, Isenovic, & Arendt, 2015; Hou, Song, Croteau, Akbari, & Bohr, 2017; Iourov et al., 2009; Lee, Thomas, & Fenech, 2015; Zivković et al., 2010). This AD‐associated aneuploidy is thought to be due to genomic instability (Andriani, Vijg, & Montagna, 2017; Shepherd, Yang, & Halliday, 2018). This observation suggests the existence of a cell population that has newly entered a flawed cell cycle in patients with AD.

6.4. Aneuploidy can cause intellectual disability and/or general aging

Many individuals with congenital aneuploidy or aneuploidogenic conditions show symptoms of intellectual disability, suggesting a role of genomic stability in maintaining neuronal and higher cognitive/behavioral functions. Down syndrome (DS) is caused by developmental aneuploidy/chromosome 21 trisomy. DS patients develop early‐onset AD‐like dementia in their 40s‐50s (Lott & Head, 2019; Potter, 1991). The conventional interpretation of the AD‐like dementia in DS patients is that it is due to the APP gene on chromosome 21; triplicate copies of the APP gene facilitate amyloid‐beta accumulation and development of AD in DS patients. Yet, aneuploidy can also produce proteotoxic effects and ER stress, and disturb stoichiometry of macromolecular complexes (Brennan et al., 2019; Chunduri & Storchová, 2019), as well as transcriptomic alterations at the cellular and/or organ level, compared with those of nonaneuploid control subjects (Wangsa et al., 2019). Thus, the general effects of aneuploidy at the cellular and organ levels may contribute to AD development, in addition to APP overproduction in patients with DS. A recent report on a mouse model of triplication of chromosome 21 genes other than APP supports the presence of an effect of aneuploidy on amyloid‐beta accumulation. The mice showed increased amyloid‐beta aggregation and deposition of plaques with cognitive deficits, although they showed no increase in APP abundance (Wiseman et al., 2018).

Cohesinopathy, a defect leading to premature mitotic chromosome segregation and chromosome instability, is an aneuploidogenic condition. Congenital mutations causing cohesinopathy in humans result in conditions with intellectual disability, deformity, and proneness to cancers, such as Cornelia de Lange syndrome and Roberts syndrome (Cucco & Musio, 2016; Zhu & Wang, 2019). Other aneuploidogenic conditions include mosaic variegated aneuploidy syndrome, which is caused by a mutation in (a) a mitotic checkpoint component BubR1/Bub1B (type 1); (b) CEP57, which encodes a centrosomal protein (type 2) (Snape et al., 2011); or (c) TRIP13, which is involved in mitotic checkpoint complex silencing via Mad2 liberation (Alfieri, Chang, & Barford, 2018; Kaisari et al., 2019). A reduction in BubR1 protein in mice (BubR1H/H mice) resulted in a systemic and neuronal progeria condition (Choi et al., 2016) and in impairment of adult hippocampal neurogenesis (Yang et al., 2017). The progeric conditions were partly ameliorated by targeted elimination/reduction of p16INK4‐positive cells (Baker et al., 2011), by enhancing Wnt signaling via loss of Dickkopf‐1 (Seib et al., 2013) or via inhibition of secreted frizzled‐related protein 3 (sFRP3) (Cho et al., 2019), both of which are endogenous Wnt antagonists. These findings suggest that (a) there is a role of genomic maintenance and aneuploidy in aging processes, and that (b) cell proliferation in the hippocampus is controlled by Wnt signaling. This hippocampal defect may affect neurocognitive performance.

6.5. AD pathology is associated with mitosis

Amyloid‐beta: Proteases involved in the APP‐to‐amyloid‐beta conversion are increasingly well characterized (Penke et al., 2017; Sikanyika et al., 2019). However, the stage at which APP is converted to amyloid‐beta is a question that is addressed less often. APP can be phosphorylated by multiple kinases, including cdc2, Rho‐associated coiled‐coil kinase 1 (ROCK1), and Polo‐like kinase 2 (Plk2). The phosphorylation affects its proteolytic processing, trafficking, and protein–protein interaction (Muresan & Muresan, 2007; Suzuki & Nakaya, 2008; Vieira, Rebelo, Domingues, da Cruz e Silva, & da Cruz e Silva, 2009). Inhibition of Plk2 reduced Aβ formation, synapse loss, and memory decline in the APP‐swDI AD mouse model (Lee et al., 2019). APP is phosphorylated at Thr668, which occurs during mitosis through mitotic cdc2 kinase (Suzuki et al., 1994). Thr668‐phosphorylated APP was associated with the centrosome (Judge, Hornbeck, Potter, & Padmanabhan, 2011). Lack of APP resulted in delayed G2/mitosis in APP‐/‐ mice (López‐Sánchez, Müller, & Frade, 2005), suggesting a role of APP during mitosis. Moreover, amyloid‐beta generation was increased during mitosis in cycling cultured H4‐15X cells and with antimitotic drug treatments, suggesting that mitotic cells are a source of amyloid‐beta (Judge et al., 2011). Conversely, amyloid‐beta can interfere with mitosis by disrupting mitotic spindles and inhibiting mitotic motors (Borysov, Granic, Padmanabhan, Walczak, & Potter, 2011), suggesting that amyloid‐beta accumulation alone can be aneuploidogenic. Amyloid‐beta oligomer is mitogenic and can trigger the cell cycle (Varvel et al., 2008). Thus, amyloid‐beta accumulation and the aneuploidogenic mitotic state may form an “amyloid‐beta accumulation cycle” (Figure 2a).

Tau: Human neurofibrillary tangles co‐localized with MPM2 antigens, a mitotic marker. The report suggests the involvement of mitosis in generation of tangles (Kondratick & Vandré, 1996). Human neuroblastoma SY5Y cells overexpressing tau provide a model for tauopathy studies. Abnormal tau phosphorylation of the Alzheimer‐type (AT100 immunopositive and Ser422) was observed in these cells during mitosis (Delobel et al., 2002). G2/M blockers (paclitaxel, vinblastine, and vincristine) have a dose‐dependent effect on tau phosphorylation at Ser‐202 and Ser‐396/404 in N2aTau3R cells. The Ser‐201 and Ser‐396/404 phosphorylation on tau are associated with neurofibrillary tangles (Conejero‐Goldberg, Townsend, & Davies, 2008). During mitosis, c‐Jun N‐terminal kinase phosphorylates R406W tau (Tatebayashi et al., 2006). Taken together, these results indicate that mitotic conditions can generate phosphorylated tau, which is associated with neurofibrillary tangles (Pope et al., 1994; Preuss & Mandelkow, 1998; Vincent, Zheng, Dickson, Kress, & Davies, 1998).

6.6. Mutations associated with familial/early‐onset AD can cause mitotic error and aneuploidy, as well as other cell cycle disturbances

Presenilin 1/PSEN1 mutation (e.g., familial AD mutation in presenilin 1 [M146L and M146V]) is linked to familial/early‐onset AD. Overexpression of mutant PSEN1 caused chromosome missegregation and aneuploidy in vivo (mice) and in vitro, with mitotic spindle defects observed (Boeras et al., 2008). PSEN1 P117R mutation is a pathogenic AD mutation that can cause increases in p53 and p21 proteins, G1 phase prolongation, S phase shortening, and decreased apoptosis in human lymphocytes (Bialopiotrowicz et al., 2012). Lymphocytes are proposed to serve as a surrogate indicator for the development of AD, as these cells are responsive to oxidative stress and other challenges, and are indicative of aneuploidy and cell cycle disturbances that mirror the condition of neurons in patients with AD (Wojsiat, Prandelli, Laskowska‐Kaszub, Martín‐Requero, & Wojda, 2015). As mentioned in Section 6.5, amyloid‐beta can also disrupt the mitotic spindle and inhibit mitotic motors, thus causing mitotic defects and aneuploidy (Borysov et al., 2011). The APOE subtype is associated with AD risk. Knockdown of APOE in APOE‐expressing ovarian cancer cells led to G2 cell cycle arrest and apoptosis, suggesting its context‐dependent role in cell cycle progression (Chen et al., 2005).

6.7. Forced cell cycle re‐entry resulted in amyloid‐beta and p‐tau accumulation in mouse brains

Transgenic mice in which cell cycle reactivation in neurons is forced by SV40 T antigen via the tet‐on/off system show signs of mitotic re‐entry (e.g., PCNA, cyclin B1, MPM2) and Aβ deposits and phosphorylated tau in the brain (Park, Hallows, Chakrabarty, Davies, & Vincent, 2007). Expression of SV40 T antigen causes replication stress, mitotic dysfunction, and aneuploidy (Hu, Filippakis, Huang, Yen, & Gjoerup, 2013), suggesting a link among mitotic re‐entry, aneuploidy, and AD pathology.

6.8. Amyloid‐beta can bind to mitotic motors and microtubules, causing mitotic error and aneuploidy, as well as triggering the stress response

Amyloid‐beta can disrupt the mitotic spindle and inhibit mitotic motors (e.g., Eg5, KIF4A, MCAK), causing mitotic defects with prolonged mitosis and aneuploidy (Borysov et al., 2011). The transcriptome of cultured SH‐SY5Y cells expressing P301L tau was most affected in the cell cycle and cell proliferation; proteomic analysis on an amyloid‐beta (1‐42)‐injected mouse model revealed that the stress response and metabolism pathways were most affected (Götz et al., 2008). Amyloid‐beta injection in a rat model caused pro‐apoptotic changes (increased caspase‐3, decreased Bcl2/Bax ratio) and activation of stress/mitogenic signaling (increased pERK, pJNK, and NFkB65kd; decreased IkB) (e.g., Dong, Ji, Han, & Han, 2019). Thus, once accumulated, amyloid‐beta can be aneuploidogenic by itself and can trigger stress response and cell death.

6.9. Infection with AD‐associated pathogens can cause mitotic re‐entry, mitotic errors, and/or prolonged mitosis

Various pathogens, including viruses (HHV1‐6, HCV), bacteria (Chlamydia pneumoniae, Helicobacter pylori), fungi (Candida albicans), and protozoa (Toxoplasma gondii), have been identified as potential AD risk factors (Sochocka et al., 2017). Herpes simplex virus 1/HSV1/HHV1 immediate–early protein Vmw110 was shown to inhibit G1/S transition and progression through mitosis (i.e., prolonged mitosis at pseudo‐prometaphase), which was in part caused by Vmw110‐induced proteasome‐dependent degradation of a centromeric protein CENP‐C (Everett, Earnshaw, Findlay, & Lomonte, 1999; Lomonte & Everett, 1999). Cytomegalovirus CMV/HHV5 infection caused transcriptomic misregulations in cell cycle and mitosis genes, and produced a pseudo‐mitosis state in the infected cells (Hertel & Mocarski, 2004). Chlamydia trachomatis disrupted cytokinesis of the host cells and caused aneuploidy with multinuclei (Sun, Sin, Poirier, & Harrison, 2016). Expression of Helicobacter pylori oncoprotein CagA caused (a) uncontrolled cell proliferation by activating the oncoprotein SHP‐2 and (b) spindle misorientation at the onset of anaphase and chromosomal segregation errors with abnormal division axis (Umeda et al., 2009). Phagocytosed Candida albicans caused macrophages to fail cell division, leading to large multinuclear aneuploids (Lewis, Bain, Lowes, Gow, & Erwig, 2012). Toxoplasma gondii facilitated normally quiescent fibroblasts to enter S phase/mitotic re‐entry, and the effect could be mediated both by direct invasion and by conditioned medium in vitro (Lavine & Arrizabalaga, 2009). These observations of AD‐associated pathogens being able to cause mitotic re‐entry, mitotic errors, and/or prolonged mitosis may help to reconcile the aforementioned “AD is caused by pathogen” theory and the “amyloid‐beta accumulation cycle.”

7. WILL ANEUPLOIDY ALONE BE SUFFICIENT TO CAUSE AMYLOID‐BETA ACCUMULATION?

Cohesinopathy–genomic instability model Shugoshin 1 (Sgo1) haploinsufficient mice (Sgo1−/+ mice) showed spontaneous cerebral amyloid‐beta accumulation in old age (Figure 2c; Rao, Farooqui, Asch, et al., 2018; Rao, Farooqui, Zhang, et al., 2018). Normally, amyloid‐beta accumulation does not occur in mice. The International Mouse Phenotyping Consortium (IMPC) database reports an abnormal behavior phenotype in Sgo1tm1a(EUCOMM)Wtsi allele mice, suggesting the likelihood of AD‐like cognitive function/behavioral issues with Sgo1 defects (http://www.mousephenotype.org/data/genes/MGI:1919665#section-associations). In the Sgo1−/+ mice, we did not observe a higher amount of APP protein. Thus, accumulation of precursor protein APP was unlikely to be the cause of amyloid‐beta accumulation. Amyloidogenic protease BACE and mitotic marker phosphor‐histone H3 co‐localized with amyloid‐beta in amyloid‐beta‐expressing cells, suggesting that mitotic/quasi‐mitotic/mitotic catastrophe cells were responsible for increased amyloid‐beta in aged Sgo1−/+ mice (Rao, Farooqui, Zhang et al., 2018).

However, spindle checkpoint defect–genomic instability model BubR1−/+ mice did not show cerebral amyloid‐beta accumulation (Rao, Farooqui, Zhang et al., 2018), suggesting that aneuploidy alone may not be sufficient to cause amyloid‐beta accumulation in a mouse model. Since a major difference in these two chromosome instability–aneuploidogenic models is spindle checkpoint function and prolonged mitosis, prolonged mitosis was proposed to be one of the three critical factors (the “three‐hit” hypothesis; Figure 2b) for amyloid‐beta accumulation (Rao, Farooqui, Asch et al., 2018). Thus, types of aneuploidy that are accompanied by prolonged mitosis, such as cohesinopathy and amyloid‐beta poisoning, are speculated to further lead to amyloid‐beta accumulation.

Whether tetraploidization, another type of aneuploidy, contributes to AD development is a matter of controversy. Tetraploidization was reported to occur in normal and AD brains to a similar degree (Westra, Barral, & Chun, 2009). This finding suggests that the effects of tetraploidization on AD development are limited. A newer paper, however, reported a correlation between neuronal tetraploidization in the cerebral cortex in mice and reduced cognition and AD‐associated neuropathology, suggesting a causal role of tetraploidization in the development of AD (López‐Sánchez et al., 2017). For the tetraploidization mechanism, as AD brains abundantly express neurotrophin receptor p75NTR and proNGF (nerve growth factor), their involvement in triggering neuronal tetraploidization, subsequent abortive mitosis, cell death, and hence neurodegeneration was suggested (Frade & López‐Sánchez, 2010). Determining the cause–consequence relationship of tetraploidization in AD may not be simple, as they may occur rather simultaneously.

8. THIRTEEN AMONG 37 GENES ON THE HUMAN AD GENETIC RISK LOCI ARE FUNCTIONALLY INVOLVED IN THE CELL CYCLE AND/OR MITOSIS

Analyzing AD brains in a comprehensive and hypothesis‐free manner with a combination of various ‐omics, imaging, and other biomarker analysis techniques has been proposed by the “Alzheimer Precision Medicine Initiative (APMI)” to advance understanding of AD, to identify dysfunctional systems and predictive markers, and to develop remedies against neurodegenerative disorders (Hampel, Toschi, et al., 2018; Hampel, Vergallo, et al., 2018). Genome sequencing projects of human AD patients and meta‐analysis of the reports have revealed genes/loci that are frequently mutated in AD patients, that is, AD genetic risk loci (Beecham et al., 2014; Carrasquillo et al., 2015; Chouraki & Seshadri, 2014; Jansen et al., 2019; Kim, 2018; Kunkle et al., 2019; Lambert et al., 2013; Van Cauwenberghe, Broeckhoven, & Sleegers, 2016; Zhang, Gaiteri, et al., 2013), in addition to known familial AD mutations, such as PSEN1/2, APP, and APOE variants. The genes include 21 previously identified loci: ABCA7, BIN1, CASS4, SORL1, CD33, CD2AP, CELF1, CLU, CR1, DSG2, EPHA1, FERMT2, HLA‐DRB5/HLA‐DRB1, INPP5D, MEF2C, MS4M6A, MS4A4E, NME8, PTK2B/PYK2, SLC24A4, and ZCWPW1. In addition, ADAM10, ACE, NYAP1, SPI1, and ECHDC3 were identified through a recent meta‐analysis (Kunkle et al., 2019). ADAMTS4, HESX1, CLNK, TREM2, CNTAP2, APH1B, KAT8, SCIMP, ABI3, SUZ12P1, ALPK2, and BZRAP‐AS1 were identified with international transethnic cohorts (Jun et al., 2017) (Table 1).

Using genome‐wide association studies (GWASs), Han, Huang, Gao, and Huang (2017) identified functions of the genes and categorized these functions as “regulation of beta‐amyloid formation,” “regulation of neurofibrillary tangle assembly,” “leukocyte‐mediated immunity,” and “protein‐lipid complex assembly” signaling pathways. With the protein–protein interaction network and functional module analyses, they also identified “hub” genes and “bottleneck” genes indicating three subnetworks. The hub genes included APOE, PICALM, BIN1, ABCA7, CD2AP, CLU, CR1, MS4A4E, and MS4A6A, while the bottleneck genes included APOE, TOMM40, NME8, PICALM, CD2AP, ZCWPW1, FAM180B, GAB2, and PTK2B (Han et al., 2017). However, as of 2019, not all genes are well characterized, as indicated by the limited number of publications listed in Table 1.

From supporting evidence of the involvement of the cell cycle and mitotic re‐entry in AD development, we hypothesized that some of the genes identified as AD genetic risk loci are functionally involved in the cell cycle and/or mitosis. We performed a series of literature searches using “cell cycle” or “mitosis” as keywords for each of the genes. The search revealed possible functional involvement in the cell cycle or mitosis regulation for 13 among 37 genes (Table 1). This result provides additional support to the long‐standing hypothesis that human AD development is associated with, influenced by, or caused by misregulations in the cell cycle or mitosis via gene mutations, at least in part. The hypothesis warrants further investigation.

9. EFFECTS OF CELL CYCLE INTERFERING DRUGS ON AD MODELS

The aforementioned reports suggest that two major AD pathological features, plaques and tangles, are caused by or associated with cell cycle misregulation toward mitosis. Existing pharmacological reagents can interfere with the cell cycle and its machineries, leading to the question of whether these drugs affect neuronal health and AD symptoms and pathology.

9.1. Antimitotic drugs can be neuroprotective against tauopathy

With exceptions of mitotic kinase or mitotic motor inhibitors, most antimitotic drugs target microtubule dynamics and mitotic spindles. Taxanes, including paclitaxel/Taxol, are microtubule stabilizers, while vinblastine and vincristine are microtubule destabilizers. Both classes of antimitotic drugs have a demonstrated history of use in cancer chemotherapy (Florian & Mitchison, 2016). Numerous in vitro results suggest the benefits of antimitotics for tauopathy. For example, cultured Aplysia neurons expressing mutant–human–tau indicate morphological signs of neurodegeneration under live confocal imaging platforms. A clinically relevant dose of 10 nM paclitaxel rescued these effects, whereas 100 nM paclitaxel facilitated them (Shemesh & Spira, 2011). In testing drugs in animal models, blood–brain barrier penetration and cerebral drug availability must also be considered (Brunden et al., 2011). In the PS19 tau transgenic mouse model of tauopathy, the brain‐penetrant antimitotic drug epothilone D reduced the burden of tau pathology (Zhang et al., 2012). Another brain‐penetrant microtubule stabilizer, dictyostatin, also produced improvement in CNS/brain measures (Brunden, Lee, Smith, Trojanowski, & Ballatore, 2017; Makani et al., 2016). Note that most antimitotics target microtubule dynamics. The possibility remains that their neuroprotective effects occur through microtubule and microtubule‐binding protein‐mediated signaling and/or axonal transport, rather than cell cycle effects (Brunden et al., 2017; Trojanowski, Smith, Huryn, & Lee, 2005). There are ongoing translational studies and clinical trials. For example, TPI‐287/abeotaxane is a brain‐penetrant microtubule stabilizer that indicated efficacy in PS19 tau transgenic mice. Recent basket clinical trials on different tauopathies to test the safety, tolerability, and potential efficacy of TPI‐287 infusion indicated that (i) TPI‐287 was generally well tolerated, although anaphylactoid reactions occurred in 3/26 of AD patients, but not in 42 4‐repeat tauopathies (4RT) patients, which led to a discussion of a potential difference in sensitivity profiles of AD and 4RT patients; (ii) CSF YKL‐40 level changed in TPI‐287 groups; and (iii) the AD treatment group showed a smaller decline in MMSE scores than did the placebo group, although the difference was not significant (Tsai et al., 2019). Still, antimitotics remain candidate drugs for AD management.

9.2. CDK inhibitors ameliorated AD symptoms in animal AD models

CDK5 is a unique member of the CDK family. Unlike canonical cell cycle driving CDKs, such as CDK1 and CDK2, Cdk5 is inactive in the cell cycle, but is specifically expressed and predominantly active in postmitotic neurons. Its role in AD has long been postulated (Dhavan & Thai, 2001). In physiological conditions, CDK5 binds with its activator, p35, and plays roles in the development of CNS and movements of neurons. However, once neurons experience pathogenic challenges, Cdk5 associates with p25, which is generated from p35 by calpain‐dependent cleavage, and becomes hyperactivated. CDK5/p25 causes aberrant hyperphosphorylation of various substrates that include APP, tau, and neurofilaments. Subsequently, Aβ formation, tau hyperphosphorylation, synaptic plasticity, oxidative stress, and mitochondrial dysfunction occur, followed by neuronal cell apoptosis and neurodegeneration with AD pathology (Liu et al., 2016; Lopes & Agostinho, 2011). CDK5 is also a regulator of other cell cycle regulators, including c‐Jun and p38MAPK (Chang et al., 2010). Misregulation of CDK5 can trigger aberrant activation of cell cycle kinases and phosphatases, leading to neuronal cell death (Chang, Vincent, & Shah, 2012). CDK5 inhibition has a neuroprotective effect (Mushtaq et al., 2016). Diaminothiazoles is a CDK5 and GSK3β inhibitor. Diaminothiazoles decreased PHF‐1 immunoreactivity in two animal models of AD (3xTg‐AD and CK‐p25), showed neuroprotective effects, and helped memory recovery (Zhang, Hernandez, et al., 2013).

Flavopiridol/alvocidib is a potent and specific inhibitor of CDKs 1, 2, 4, and 7 in vitro, showing a clear blockade of cell cycle progression at the G1/S and G2/M boundaries (Senderowicz, 1999). In the hCOX‐2 transgenic mice, overexpression of human COX‐2 in murine primary hippocampal neurons accelerated beta‐amyloid‐mediated apoptosis. The in vitro neuronal damage was prevented by flavopiridol (Xiang et al., 2002). Leggio et al. (2016) tested flavopiridol/alvocidib in an amyloid‐beta‐injected AD mouse model. Aβ‐injected mice showed mitotic cyclin A‐positive cycling neurons in the frontal cortex and the hippocampus, and displayed memory deficits. The cell cycle events and memory deficits were prevented by flavopiridol administered at 0.5 and 1 mg/kg body weight. Zhang, Gaiteri, et al. (2013), reported that (a) a strong association with AD clinical and pathological traits and cell cycle‐enriched module/subnetwork, and (b) the cell cycle‐enriched module/subnetwork showed the second largest loss of gene–gene interactions in AD compared with normal controls, thus was among the most affected. Huang et al. (2019) predicted that denticleless (DTL) is the key driver gene of the cell cycle‐enriched module and investigated the role of DTL‐encoded CDT2 protein in AD pathology. In addition, CDT2 is a part of the cell cycle‐driving CUL4 CRL ubiquitin ligase complex. They reported upregulation of DTL/CDT2 in human patients with AD and increased degradation of p21, a CDK inhibitor protein, in vitro with CDT2 overexpression. In mice overexpressing CDT2, degradation of p21 released CDKs' activity toward cell cycle progression and triggered AD processes, including increases in amyloid‐beta, p‐tau, and BACE; memory deficits; and a gene expression signature similar to that observed in human AD, for example, increases in APOE, CASS4, and CLU. Treatment with CDK2/7/9 inhibitor roscovitine/seliciclib rescued CDT2‐induced cognitive defects in CDT2‐overexpressing mice (Huang et al., 2019).

9.3. Targeting mitogenic signaling

Mitogenic signaling is another potential target for AD drugs. However, few clinical trials for AD have explored mitogenic signaling. Those trials included examinations of the following drugs: lactoferrin, which can modulate p‐AKT/PTEN (Mohamed, Salama, & Schaalan, 2019); neflamapimod, a specific inhibitor of p38MAPK‐alpha (Alam, Blackburn, & Patrick, 2017); bryostatin 1, an activator of protein kinase C epsilon (Nelson et al., 2017); tideglusib, an inhibitor of GSK3 (Lovestone et al., 2015); and lithium, a GSK3 inhibitor (Forlenza et al., 2011). Although this “targeting mitogenic kinase/signaling” approach has not become mainstream in AD research and drug development, the approach has been established in cancer research and drug development, with success against oncogene‐addicted cancers (e.g., imatinib/Gleevec targeting Bcr‐Abl tyrosine kinase, trastuzumab/Herceptin targeting Neu receptor). Due to organ‐specific issues (e.g., blood–brain barrier [BBB]‐mediated drug delivery), cancer drug repurposing for AD therapy may not be straightforward. Still, given the abundance of accumulated resources for targeting mitogenic signaling, the approach may hold great future potential.

10. CONCERNS TO BE ADDRESSED

Cancer treatments can leave cognitive impairment in a segment of cancer patients during the course of treatment or after completion, a phenomenon known as chemobrain (Argyriou, Assimakopoulos, Iconomou, Giannakopoulou, & Kalofonos, 2011). A meta‐analysis of animal model studies indicated a correlation between cognitive decline and chemotherapy drug treatments, especially with the cisplatin, CMF (cyclophosphamide, methotrexate, fluorouracil combination), and MTX (methotrexate) + 5‐FU chemotherapy regimens (Matsos & Johnston, 2019). However, there is currently little evidence that the treatments caused amyloid‐beta accumulation or triggered AD‐like dementia. Instead, the cognitive decline is attributed to damage to the BBB, oxidative stress, and cytokine dysregulation; thus, this chemobrain phenomenon is believed to be mechanistically closer to vascular dementia (Ren et al., 2019; Ren, St Clair, & Butterfield, 2017). Consistently, severe chemobrain is associated with cytotoxic DNA‐damaging drugs, rather than CDK inhibitors. An inverse relationship between cancer and AD is known; that is, cancer survivors have less likelihood of developing AD (Zhang et al., 2015). The inverse relationship is proposed to be related to a balance in cellular tendencies toward cell death or growth (Shafi, 2016). Mechanisms in cell survival/death regulation, that is, p53, Pin1, and the Wnt signaling pathway, were discussed as potential therapeutic manipulation targets (Behrens, Lendon, & Roe, 2009). In addition to innate cellular tendencies, we speculate that the inverse relationship may be in part due to a therapeutic or intervention effect of cancer chemotherapy drug on preclinical AD. When cell cycle‐managing chemotherapy drugs are to be repurposed for AD prevention and/or therapy, we suggest designing clinical trials with careful and conservative dosing, with a less likelihood of BBB damage and/or chemobrain induction.

11. SUMMARY

Increasing evidence, including recent ‐omics data from human patients with AD, points to a critical role of the cell cycle and mitosis, leading to the “amyloid‐beta accumulation cycle,” in the development of AD pathology. CDK inhibitors tested on animal models of AD showed symptomatic relief, corroborating the notion that the cell cycle and mitosis are targets of AD drug research and development. With further support, existing cell cycle and mitosis‐targeting drugs, many of which are clinically used as cancer drugs, may be successfully repurposed as AD drugs in the near future.

CONFLICTS OF INTEREST

No conflicts of interest declared.

AUTHOR CONTRIBUTIONS

H.Y. Yamada contributed all aspects of the project. C.V. Rao and A.S. Asch provided intellectual input and material support. D.J.J. Carr contributed generation of unpublished key data.

ACKNOWLEDGEMENTS

We thank Ms. Kathy Kyler for editorial aid, and Ms. Taylor McCoy and Ms. Elizabeth Cambron for administrative aid. Special thanks to anonymous reviewers for the journal. This work was supported by grants from the U.S. National Institutes of Health to C.V. Rao (NCI R01CA094962; NCI R01CA213987) and research funds from the Stephenson Cancer Center to H.Y. Yamada.

Rao CV, Asch AS, Carr DJJ, Yamada HY. “Amyloid‐beta accumulation cycle” as a prevention and/or therapy target for Alzheimer's disease. Aging Cell. 2020;19:e13109 10.1111/acel.13109

Contributor Information

Chinthalapally V. Rao, Email: cv-rao@ouhsc.edu.

Hiroshi Y. Yamada, Email: Hiroshi-yamada@ouhsc.edu.

REFERENCES

  1. Akiguchi, I. , Pallàs, M. , Budka, H. , Akiyama, H. , Ueno, M. , Han, J. , … Hosokawa, M. (2017). SAMP8 mice as a neuropathological model of accelerated brain aging and dementia: Toshio Takeda's legacy and future directions. Neuropathology, 37, 293–305. 10.1111/neup.12373 [DOI] [PubMed] [Google Scholar]
  2. Al Nakouzi, N. , Wang, C. K. , Beraldi, E. , Jager, W. , Ettinger, S. , Fazli, L. , … Gleave, M. (2016). Clusterin knockdown sensitizes prostate cancer cells to taxane by modulating mitosis. EMBO Molecular Medicine, 8, 761–778. 10.15252/emmm.201506059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alam, J. , Blackburn, K. , & Patrick, D. (2017). Neflamapimod: Clinical phase 2b‐ready oral small molecule inhibitor of p38α to reverse synaptic dysfunction in early Alzheimer's disease. The Journal of Prevention of Alzheimer's Disease, 4, 273–278. 10.14283/jpad.2017.41 [DOI] [PubMed] [Google Scholar]
  4. Alfieri, C. , Chang, L. , & Barford, D. (2018). Mechanism for remodelling of the cell cycle checkpoint protein MAD2 by the ATPase TRIP13. Nature, 559, 274–278. 10.1038/s41586-018-0281-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Andriani, G. A. , Vijg, J. , & Montagna, C. (2017). Mechanisms and consequences of aneuploidy and chromosome instability in the aging brain. Mechanisms of Ageing and Development, 161(Pt A), 19–36. 10.1016/j.mad.2016.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Arendt, T. , Brückner, M. K. , Mosch, B. , & Lösche, A. (2010). Selective cell death of hyperploid neurons in Alzheimer's disease. American Journal of Pathology, 177, 15–20. 10.2353/ajpath.2010.090955 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Argyriou, A. A. , Assimakopoulos, K. , Iconomou, G. , Giannakopoulou, F. , & Kalofonos, H. P. (2011). Either called “chemobrain” or “chemofog,” the long‐term chemotherapy‐induced cognitive decline in cancer survivors is real. Journal of Pain and Symptom Management, 41, 126–139. 10.1016/j.jpainsymman.2010.04.021 [DOI] [PubMed] [Google Scholar]
  8. Armanious, H. , Gelebart, P. , Anand, M. , Belch, A. , & Lai, R. (2011). Constitutive activation of metalloproteinase ADAM10 in mantle cell lymphoma promotes cell growth and activates the TNFα/NFκB pathway. Blood, 117, 6237–6246. 10.1182/blood-2010-10-313940 [DOI] [PubMed] [Google Scholar]
  9. Badodi, S. , Baruffaldi, F. , Ganassi, M. , Battini, R. , & Molinari, S. (2015). Phosphorylation‐dependent degradation of MEF2C contributes to regulate G2/M transition. Cell Cycle, 14, 1517–1528. 10.1080/15384101.2015.1026519 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bajic, V. , Spremo‐Potparevic, B. , Zivkovic, L. , Isenovic, E. R. , & Arendt, T. (2015). Cohesion and the aneuploid phenotype in Alzheimer's disease: A tale of genome instability. Neuroscience and Biobehavioral Reviews, 55, 365–374. 10.1016/j.neubiorev.2015.05.010 [DOI] [PubMed] [Google Scholar]
  11. Baker, D. J. , Wijshake, T. , Tchkonia, T. , LeBrasseur, N. K. , Childs, B. G. , van de Sluis, B. , … van Deursen, J. M. (2011). Clearance of p16Ink4a‐positive senescent cells delays ageing‐associated disorders. Nature, 479, 232–236. 10.1038/nature10600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Beecham, G. W. , Hamilton, K. , Naj, A. C. , Martin, E. R. , Huentelman, M. , Myers, A. J. , … Montine, T. J. (2014). Genome‐wide association meta‐analysis of neuropathologic features of Alzheimer's disease and related dementias. PLoS Genetics, 10, e1004606 10.1016/j.jalz.2016.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Behrens, M. I. , Lendon, C. , & Roe, C. M. (2009). A common biological mechanism in cancer and Alzheimer's disease? Current Alzheimer Research, 6, 196–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Belkhelfa, M. , Rafa, H. , Medjeber, O. , Arroul‐Lammali, A. , Behairi, N. , Abada‐Bendib, M. , … Touil‐Boukoffa, C. (2014). IFN‐γ and TNF‐α are involved during Alzheimer disease progression and correlate with nitric oxide production: A study in Algerian patients. Journal of Interferon and Cytokine Research, 34, 839–847. 10.1089/jir.2013.0085 [DOI] [PubMed] [Google Scholar]
  15. Bhaskar, K. , Maphis, N. , Xu, G. , Varvel, N. H. , Kokiko‐Cochran, O. N. , Weick, J. P. , … Lamb, B. T. (2014). Microglial derived tumor necrosis factor‐α drives Alzheimer's disease‐related neuronal cell cycle events. Neurobiology of Diseases, 62, 273–285. 10.1016/j.nbd.2013.10.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bialopiotrowicz, E. , Szybinska, A. , Kuzniewska, B. , Buizza, L. , Uberti, D. , Kuznicki, J. , & Wojda, U. (2012). Highly pathogenic Alzheimer's disease presenilin 1 P117R mutation causes a specific increase in p53 and p21 protein levels and cell cycle dysregulation in human lymphocytes. Journal of Alzheimer's Disease, 32, 397–415. 10.3233/JAD-2012-121129 [DOI] [PubMed] [Google Scholar]
  17. Blume, T. , Focke, C. , Peters, F. , Deussing, M. , Albert, N. L. , Lindner, S. , … Brendel, M. (2018). Microglial response to increasing amyloid load saturates with aging: A longitudinal dual tracer in vivo μPET‐study. Journal of Neuroinflammation, 15, 307 10.1186/s12974-018-1347-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Boeras, D. I. , Granic, A. , Padmanabhan, J. , Crespo, N. C. , Rojiani, A. M. , & Potter, H. (2008). Alzheimer's presenilin 1 causes chromosome missegregation and aneuploidy. Neurobiology of Aging, 29, 319–328. 10.1016/j.neurobiolaging.2006.10.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Borysov, S. I. , Granic, A. , Padmanabhan, J. , Walczak, C. E. , & Potter, H. (2011). Alzheimer Aβ disrupts the mitotic spindle and directly inhibits mitotic microtubule motors. Cell Cycle, 10, 1397–1410. 10.4161/cc.10.9.15478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Boveri, T. (2008). Concerning the origin of malignant tumours by Theodor Boveri. Translated and annotated by Henry Harris. Journal of Cell Science, 121(Supplement 1), 1–84. 10.1242/jcs.025742 [DOI] [PubMed] [Google Scholar]
  21. Braak, H. , & Braak, E. (1991). Neuropathological stageing of Alzheimer‐related changes. Acta Neuropathologica, 82(4), 239–259. 10.1007/BF00308809 [DOI] [PubMed] [Google Scholar]
  22. Brennan, C. M. , Vaites, L. P. , Wells, J. N. , Santaguida, S. , Paulo, J. A. , Storchova, Z. , … Amon, A. (2019). Protein aggregation mediates stoichiometry of protein complexes in aneuploid cells. Genes & Development, 33, 1031–1047. 10.1101/gad.327494.119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Brunden, K. R. , Lee, V. M. , Smith, A. B. 3rd , Trojanowski, J. Q. , & Ballatore, C. (2017). Altered microtubule dynamics in neurodegenerative disease: Therapeutic potential of microtubule‐stabilizing drugs. Neurobiology of Diseases, 105, 328–335. 10.1016/j.nbd.2016.12.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Brunden, K. R. , Yao, Y. , Potuzak, J. S. , Ferrer, N. I. , Ballatore, C. , James, M. J. , … Lee, V.‐Y. (2011). The characterization of microtubule‐stabilizing drugs as possible therapeutic agents for Alzheimer's disease and related tauopathies. Pharmacological Research, 63, 341–351. 10.1016/j.phrs.2010.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Caberlotto, L. , Lauria, M. , Nguyen, T. P. , & Scotti, M. (2013). The central role of AMP‐kinase and energy homeostasis impairment in Alzheimer's disease: A multifactor network analysis. PLoS ONE, 8(11), e78919 10.1371/journal.pone.0078919 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cai, F. , Zhu, Q. , Miao, Y. , Shen, S. , Su, X. , & Shi, Y. (2017). Desmoglein‐2 is overexpressed in non‐small cell lung cancer tissues and its knockdown suppresses NSCLC growth by regulation of p27 and CDK2. Journal of Cancer Research and Clinical Oncology, 143, 59–69. 10.1007/s00432-016-2250-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Carrasquillo, M. M. , Crook, J. E. , Pedraza, O. , Thomas, C. S. , Pankratz, V. S. , Allen, M. , … Ertekin‐Taner, N. (2015). Late-onset Alzheimer's risk variants in memory decline, incident mild cognitive impairment, and Alzheimer's disease. Neurobiology of Aging, 36, 60–67. 10.1016/j.neurobiolaging.2014.07.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chang, K. H. , de Pablo, Y. , Lee, H. P. , Lee, H. G. , Smith, M. A. , & Shah, K. (2010). Cdk5 is a major regulator of p38 cascade: Relevance to neurotoxicity in Alzheimer's disease. Journal of Neurochemistry, 113, 1221–1229. 10.1111/j.1471-4159.2010.06687 [DOI] [PubMed] [Google Scholar]
  29. Chang, K. H. , Vincent, F. , & Shah, K. (2012). Deregulated Cdk5 triggers aberrant activation of cell cycle kinases and phosphatases inducing neuronal death. Journal of Cell Science, 125, 5124–5137. 10.1242/jcs.108183 [DOI] [PubMed] [Google Scholar]
  30. Chen, Y. C. , Pohl, G. , Wang, T. L. , Morin, P. J. , Risberg, B. , Kristensen, G. B. , … Shih Ie, M. (2005). Apolipoprotein E is required for cell proliferation and survival in ovarian cancer. Cancer Research, 65, 331–337. [PubMed] [Google Scholar]
  31. Cho, C. H. , Yoo, K. H. , Oliveros, A. , Paulson, S. , Hussaini, S. M. Q. , van Deursen, J. M. , & Jang, M. H. (2019). sFRP3 inhibition improves age‐related cellular changes in BubR1 progeroid mice. Aging Cell, 18, e12899 10.1111/acel.12899 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Choi, C.‐I. , Yoo, K. I. , Hussaini, S. , Jeon, B. , Welby, J. , Gan, H. , … Jang, M.‐H. (2016). The progeroid gene BubR1 regulates axon myelination and motor function. Aging (Albany NY), 8, 2667–2688. 10.18632/aging.101032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chouraki, V. , & Seshadri, S. (2014). Genetics of Alzheimer's disease. Advances in Genetics, 87, 245–294. 10.1016/B978-0-12-800149-3.00005-6 [DOI] [PubMed] [Google Scholar]
  34. Chunduri, N. K. , & Storchová, Z. (2019). The diverse consequences of aneuploidy. Nature Cell Biology, 21, 54–62. 10.1038/s41556-018-0243-8 [DOI] [PubMed] [Google Scholar]
  35. Cline, E. N. , Bicca, M. A. , Viola, K. L. , & Klein, W. L. (2018). The amyloid‐β oligomer hypothesis: Beginning of the third decade. Journal of Alzheimer's Disease, 64, S567–S610. 10.3233/JAD-179941 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Coleman, P. , Federoff, H. , & Kurlan, R. (2004). A focus on the synapse for neuroprotection in Alzheimer disease and other dementias. Neurology, 63, 1155–1162. 10.1212/01.WNL.0000140626.48118.0A [DOI] [PubMed] [Google Scholar]
  37. Conejero‐Goldberg, C. , Townsend, K. , & Davies, P. (2008). Effects of cell cycle inhibitors on tau phosphorylation in N2aTau3R cells. Journal of Molecular Neuroscience, 35, 143–150. 10.1007/s12031-008-9044-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Coppedè, F. , & Migliore, L. (2009). DNA damage and repair in Alzheimer's disease. Current Alzheimer Research, 6, 36‐47. [DOI] [PubMed] [Google Scholar]
  39. Cordone, S. , Annarumma, L. , Rossini, P. M. , & De Gennaro, L. (2019). Sleep and β‐Amyloid deposition in Alzheimer disease: Insights on mechanisms and possible innovative treatments. Front Pharmacol, 10, 695 10.3389/fphar.2019.00695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Cucco, F. , & Musio, A. (2016). Genome stability: What we have learned from cohesinopathies. American Journal of Medical Genetics. Part C: Seminars in Medical Genetics, 172, 171–178. 10.1002/ajmg.c.31492 [DOI] [PubMed] [Google Scholar]
  41. De Chiara, G. , Piacentini, R. , Fabiani, M. , Mastrodonato, A. , Marcocci, M. E. , Limongi, D. , … Palamara, A. T. (2019). Recurrent herpes simplex virus‐1 infection induces hallmarks of neurodegeneration and cognitive deficits in mice. PLoS Path, 15, e1007617 10.1371/journal.ppat.1007617 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. De Rossi, P. , Andrew, R. J. , Musial, T. F. , Buggia‐Prevot, V. , Xu, G. , Ponnusamy, M. , … Thinakaran, G. (2019). Aberrant accrual of BIN1 near Alzheimer's disease amyloid deposits in transgenic models. Brain Pathology, 29, 485–501. 10.1111/bpa.12687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Delobel, P. , Flament, S. , Hamdane, M. , Mailliot, C. , Sambo, A.‐V. , Bégard, S. , … Buée, L. (2002). Abnormal Tau phosphorylation of the Alzheimer‐type also occurs during mitosis. Journal of Neurochemistry, 83, 412–420. 10.1046/j.1471-4159.2002.01143.x [DOI] [PubMed] [Google Scholar]
  44. Dhavan, R. , & Tsai, L. H. (2001). A decade of CDK5. Nature Reviews Molecular Cell Biology, 2, 749–759. 10.1038/35096019 [DOI] [PubMed] [Google Scholar]
  45. Di Giorgio, E. , Gagliostro, E. , Clocchiatti, A. , & Brancolini, C. (2015). The control operated by the cell cycle machinery on MEF2 stability contributes to the downregulation of CDKN1A and entry into S phase. Molecular and Cellular Biology, 35, 1633–1647. 10.1128/MCB.01461-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Ding, X. L. , Husseman, J. , Tomashevski, A. , Nochlin, D. , Jin, L. W. , & Vincent, I. (2000). The cell cycle Cdc25A tyrosine phosphatase is activated in degenerating postmitotic neurons in Alzheimer's disease. American Journal of Pathology, 157, 1983–1990. 10.1016/S0002-9440(10)64837-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Dong, P. , Ji, X. , Han, W. , & Han, H. (2019). Oxymatrine attenuates amyloid beta 42 (Aβ1‐42)‐induced neurotoxicity in primary neuronal cells and memory impairment in rats. Canadian Journal of Physiology and Pharmacology, 97, 99–106. 10.1139/cjpp-2018-0299 [DOI] [PubMed] [Google Scholar]
  48. Dong, Y. , Li, X. , Cheng, J. , & Hou, L. (2019). Drug development for Alzheimer's disease: Microglia induced neuroinflammation as a target? International Journal of Molecular Sciences, 20, E558 10.3390/ijms20030558 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Dronse, J. , Fliessbach, K. , Bischof, G. N. , von Reutern, B. , Faber, J. , Hammes, J. , … Drzezga, A. (2017). In vivo patterns of tau pathology, amyloid‐β burden, and neuronal dysfunction in clinical variants of Alzheimer's disease. Journal of Alzheimer's Disease, 55, 465–471. 10.3233/JAD-160316 [DOI] [PubMed] [Google Scholar]
  50. Du, L.‐L. , Chai, D.‐M. , Zhao, L.‐N. , Li, X.‐H. , Zhang, F.‐C. , Zhang, H.‐B. , … Zhou, X.‐W. (2015). AMPK activation ameliorates Alzheimer's disease‐like pathology and spatial memory impairment in a streptozotocin‐induced Alzheimer's disease model in rats. Journal of Alzheimer's Disease, 43, 775–784. 10.3233/JAD-140564 [DOI] [PubMed] [Google Scholar]
  51. Du, Q. S. , Ren, X. R. , Xie, Y. , Wang, Q. , Mei, L. , & Xiong, W. C. (2001). Inhibition of PYK2‐induced actin cytoskeleton reorganization, PYK2 autophosphorylation and focal adhesion targeting by FAK. Journal of Cell Science, 114, 2977–2987. [DOI] [PubMed] [Google Scholar]
  52. Elsnerova, K. , Bartakova, A. , Tihlarik, J. , Bouda, J. , Rob, L. , Skapa, P. , … Vaclavikova, R. (2017). Gene expression profiling reveals novel candidate markers of ovarian carcinoma intraperitoneal metastasis. J Cancer, 8, 3598–3606. 10.7150/jca.20766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Esmailzadeh, S. , Huang, Y. , Su, M. W. , Zhou, Y. , & Jiang, X. (2015). BIN1 tumor suppressor regulates Fas/Fas ligand‐mediated apoptosis through c‐FLIP in cutaneous T‐cell lymphoma. Leukemia, 29, 1402–1413. 10.1038/leu.2015.9 [DOI] [PubMed] [Google Scholar]
  54. Everett, R. D. , Earnshaw, W. C. , Findlay, J. , & Lomonte, P. (1999). Specific destruction of kinetochore protein CENP‐C and disruption of cell division by herpes simplex virus immediate‐early protein Vmw110. EMBO Journal, 18, 1526–1538. 10.1093/emboj/18.6.1526 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Florian, S. , & Mitchison, T. J. (2016). Anti‐microtubule drugs. Methods in Molecular Biology, 1413, 403–421. 10.1007/978-1-4939-3542-0_25 [DOI] [PubMed] [Google Scholar]
  56. Folk, W. P. , Kumari, A. , Iwasaki, T. , Pyndiah, S. , Johnson, J. C. , Cassimere, E. K. , … Sakamuro, D. (2019). Loss of the tumor suppressor BIN1 enables ATM Ser/Thr kinase activation by the nuclear protein E2F1 and renders cancer cells resistant to cisplatin. Journal of Biological Chemistry, 294, 5700–5719. 10.1074/jbc.RA118.005699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Forlenza, O. V. , Diniz, B. S. , Radanovic, M. , Santos, F. S. , Talib, L. L. , & Gattaz, W. F. (2011). Disease‐modifying properties of long‐term lithium treatment for amnestic mild cognitive impairment: Randomised controlled trial. British Journal of Psychiatry, 198, 351–356. 10.1192/bjp.bp.110.080044 [DOI] [PubMed] [Google Scholar]
  58. Frade, J. M. , & López‐Sánchez, N. (2010). A novel hypothesis for Alzheimer disease based on neuronal tetraploidy induced by p75 (NTR). Cell Cycle, 9, 1934–1941. [DOI] [PubMed] [Google Scholar]
  59. Fu, Q. , Huang, Y. , Ge, C. , Li, Z. , Tian, H. , Li, Q. , … Song, X. (2019). SHIP1 inhibits cell growth, migration, and invasion in non–small cell lung cancer through the PI3K/AKT pathway. Oncology Reports, 41, 2337–2350. 10.3892/or.2019.6990 [DOI] [PubMed] [Google Scholar]
  60. Götz, J. , David, D. , Hoerndli, F. , Ke, Y. D. , Schonrock, N. , Wiesner, A. , … Ittner, L. M. (2008). Functional genomics dissects pathomechanisms in tauopathies: Mitosis failure and unfolded protein response. Neuro‐Degenerative Diseases, 5, 179–181. 10.1159/000113696 [DOI] [PubMed] [Google Scholar]
  61. Granic, A. , & Potter, H. (2013). Mitotic spindle defects and chromosome mis‐segregation induced by LDL/cholesterol‐implications for Niemann‐Pick C1, Alzheimer's disease, and atherosclerosis. PLoS ONE, 8, e60718 10.1371/journal.pone.0060718 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Haas, J. G. , & Lathe, R. (2018). Microbes and Alzheimer's disease: New findings call for a paradigm change. Trends in Neurosciences, 41, 570–573. 10.1016/j.tins.2018.07.001 [DOI] [PubMed] [Google Scholar]
  63. Hampel, H. , Toschi, N. , Babiloni, C. , Baldacci, F. , Black, K. L. , Bokde, A. L. W. , … Alzheimer Precision Medicine Initiative (APMI) (2018). Revolution of Alzheimer precision neurology. Passageway of systems biology and neurophysiology. Journal of Alzheimer's Disease, 64, S47–S105. 10.3233/JAD-179932 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hampel, H. , Vergallo, A. , Aguilar, L. F. , Benda, N. , Broich, K. , Cuello, A. C. , … Lista, S. (2018). Precision pharmacology for Alzheimer's disease. Pharmacological Research, 130, 331–365. 10.1016/j.phrs.2018.02.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Han, C. P. , Yu, Y. H. , Wang, A. G. , Tian, Y. , Zhang, H. T. , Zheng, Z. M. , & Liu, Y. S. (2018). Desmoglein‐2 overexpression predicts poor prognosis in hepatocellular carcinoma patients. European Review for Medical and Pharmacological Sciences, 22, 5481–5489. 10.26355/eurrev_201809_15808 [DOI] [PubMed] [Google Scholar]
  66. Han, Z. , Huang, H. , Gao, Y. , & Huang, Q. (2017). Functional annotation of Alzheimer's disease associated loci revealed by GWASs. PLoS ONE, 12, e0179677 10.1371/journal.pone.0179677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hardy, J. A. , & Higgins, G. A. (1992). Alzheimer's disease: The amyloid cascade hypothesis. Science, 256, 184–185. 10.1126/science.1566067 [DOI] [PubMed] [Google Scholar]
  68. Hardy, J. , & Selkoe, D. J. (2002). The amyloid hypothesis of Alzheimer's disease: Progress and problems on the road to therapeutics. Science, 297, 353–356. 10.1126/science.1072994 [DOI] [PubMed] [Google Scholar]
  69. Hellström‐Lindahl, E. , Ravid, R. , & Nordberg, A. (2008). Age‐dependent decline of neprilysin in Alzheimer's disease and normal brain: Inverse correlation with a beta levels. Neurobiology of Aging, 29, 210–221. [DOI] [PubMed] [Google Scholar]
  70. Herrup, K. (2010). The involvement of cell cycle events in the pathogenesis of Alzheimer's disease. Alzheimer's Research & Therapy, 2, 13 10.1186/alzrt37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Herrup, K. , & Arendt, T. (2002). Re‐expression of cell cycle proteins induces neuronal cell death during Alzheimer's disease. Journal of Alzheimer's Disease, 4, 243–247. 10.3233/JAD-2002-4315 [DOI] [PubMed] [Google Scholar]
  72. Herrup, K. , Neve, R. , Ackerman, S. L. , & Copani, A. (2004). Divide and die: Cell cycle events as triggers of nerve cell death. Journal of Neuroscience, 24, 9232–9239. 10.1523/JNEUROSCI.3347-04.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Hertel, L. , & Mocarski, E. S. (2004). Global analysis of host cell gene expression late during cytomegalovirus infection reveals extensive dysregulation of cell cycle gene expression and induction of Pseudomitosis independent of US28 function. Journal of Virology, 78, 11988–12011. 10.1128/JVI.78.21.11988-12011.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Hou, Y. , Song, H. , Croteau, D. L. , Akbari, M. , & Bohr, V. A. (2017). Genome instability in Alzheimer disease. Mechanisms of Ageing and Development, 161(Pt A), 83–94. 10.1016/j.mad.2016.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Hradek, A. C. , Lee, H.‐P. , Siedlak, S. L. , Torres, S. L. , Jung, W. , Han, A. H. , & Lee, H.‐G. (2015). Distinct chronology of neuronal cell cycle re‐entry and tau pathology in the 3xTg‐AD mouse model and Alzheimer's disease patients. Journal of Alzheimer's Disease, 43, 57–65. 10.3233/JAD-141083 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Hu, L. , Filippakis, H. , Huang, H. , Yen, T. J. , & Gjoerup, O. V. (2013). Replication stress and mitotic dysfunction in cells expressing simian virus 40 large T antigen. Journal of Virology, 87, 13179–13192. 10.1128/JVI.02224-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Huang, F. , Wang, M. , Liu, R. , Wang, J.‐Z. , Schadt, E. , Haroutunian, V. , … Wang, X. (2019). CDT2‐controlled cell cycle reentry regulates the pathogenesis of Alzheimer's disease. Alzheimer's & Dementia: The Journal of the Alzheimer's Association, 15, 217–231. 10.1016/j.jalz.2018.08.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Iourov, I. Y. , Vorsanova, S. G. , Liehr, T. , & Yurov, Y. B. (2009). Aneuploidy in the normal, Alzheimer's disease and ataxia‐telangiectasia brain: Differential expression and pathological meaning. Neurobiology of Diseases, 34, 212–220. 10.1016/j.nbd.2009.01.003 [DOI] [PubMed] [Google Scholar]
  79. Iqbal, K. , & Grundke‐Iqbal, I. (2008). Alzheimer neurofibrillary degeneration: Significance, etiopathogenesis, therapeutics and prevention. Journal of Cellular and Molecular Medicine, 12, 38–55. 10.1111/j.1582-4934.2008.00225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Itzhaki, R. F. (2014). Herpes simplex virus type 1 and Alzheimer's disease: Increasing evidence for a major role of the virus. Frontiers in Aging Neuroscience, 6, 202 10.3389/fnagi.2014.00202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Jankowsky, J. L. , & Zheng, H. (2017). Practical considerations for choosing a mouse model of Alzheimer's disease. Molecular Neurodegeneration, 12, 89 10.1186/s13024-017-0231-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Jansen, I. E. , Savage, J. E. , Watanabe, K. , Bryois, J. , Williams, D. M. , Steinberg, S. , … Posthuma, D. (2019). Genome‐wide meta‐analysis identifies new loci and functional pathways influencing Alzheimer's disease risk. Nature Genetics, 51, 404–413. 10.1038/s41588-018-0311-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Judge, M. , Hornbeck, L. , Potter, H. , & Padmanabhan, J. (2011). Mitosis‐specific phosphorylation of amyloid precursor protein at threonine 668 leads to its altered processing and association with centrosomes. Molecular Neurodegeneration, 6, 80 10.1186/1750-1326-6-80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Jun, G. R. , Chung, J. , Mez, J. , Barber, R. , Beecham, G. W. , Bennett, D. A. , … Zhang, X. (2017). Transethnic genome‐wide scan identifies novel Alzheimer's disease loci. Alzheimer's & Dementia: The Journal of the Alzheimer's Association, 13, 727–738. 10.1016/j.jalz.2016.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Kaisari, S. , Shomer, P. , Ziv, T. , Sitry‐Shevah, D. , Miniowitz‐Shemtov, S. , Teichner, A. , & Hershko, A. (2019). Role of Polo‐like kinase 1 in the regulation of the action of p31comet in the disassembly of mitotic checkpoint complexes. Proceedings of the National Academy of Sciences of the United States of America, 116, 11725–11730. 10.1073/pnas.1902970116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Kim, J. H. (2018). Genetics of Alzheimer's disease. Dementia and Neurocognitive Disorders, 17, 131–136. 10.12779/dnd.2018.17.4.131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Kim, S.‐M. , Kim, E.‐M. , Ji, K.‐Y. , Lee, H.‐Y. , Yee, S.‐M. , Woo, S.‐M. , … Kang, H.‐S. (2019). TREM2 acts as a tumor suppressor in colorectal carcinoma through Wnt1/β‐catenin and Erk signaling. Cancers (Basel), 11, E1315 10.3390/cancers11091315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Kirouac, L. , Rajic, A. J. , Cribbs, D. H. , & Padmanabhan, J. (2017). Activation of Ras‐ERK signaling and GSK‐3 by amyloid precursor protein and amyloid beta facilitates neurodegeneration in Alzheimer's disease. eNeuro, 4 pii: ENEURO.0149‐16.2017. 10.1523/ENEURO.0149-16.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Klein, W. L. , Stine, W. B. Jr , & Teplow, D. B. (2004). Small assemblies of unmodified amyloid beta‐protein are the proximate neurotoxin in Alzheimer's disease. Neurobiology of Aging, 25, 569–580. [DOI] [PubMed] [Google Scholar]
  90. Kobayashi, Y. , Tobinai, K. , Takeshita, A. , Naito, K. , Asai, O. , Dobashi, N. , … Ohno, R. (2009). Phase I/II study of humanized anti‐CD33 antibody conjugated with calicheamicin, gemtuzumab ozogamicin, in relapsed or refractory acute myeloid leukemia: Final results of Japanese multicenter cooperative study. International Journal of Hematology, 89, 460–469. 10.1007/s12185-009-0298-1 [DOI] [PubMed] [Google Scholar]
  91. Kondratick, C. M. , & Vandré, D. D. (1996). Alzheimer's disease neurofibrillary tangles contain mitosis‐specific phosphoepitopes. Journal of Neurochemistry, 67, 2405–2416. 10.1046/j.1471-4159.1996.67062405.x [DOI] [PubMed] [Google Scholar]
  92. Kress, B. T. , Iliff, J. J. , Xia, M. , Wang, M. , Wei, H. S. , Zeppenfeld, D. , … Nedergaard, M. (2014). Impairment of paravascular clearance pathways in the aging brain. Annals of Neurology, 76, 845–861. 10.1002/ana.24271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kunkle, B. W. , Grenier‐Boley, B. , Sims, R. , Bis, J. C. , Damotte, V. , Naj, A. C. , … Pericak‐Vance, M. A. (2019). Genetic meta‐analysis of diagnosed Alzheimer's disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nature Genetics, 51, 414–430. 10.1038/s41588-019-0358-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Lambert, J.‐C. , Ibrahim‐Verbaas, C. A. , Harold, D. , Naj, A. C. , Sims, R. , Bellenguez, C. , … Amouyel, P. (2013). Meta‐analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nature Genetics, 45, 1452–1458. 10.1038/ng.2802 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Lanoiselée, H. M. , Nicolas, G. , Wallon, D. , Rovelet‐Lecrux, A. , Lacour, M. , Rousseau, S. … collaborators of the CNR‐MAJ project (2017). APP, PSEN1, and PSEN2 mutations in early‐onset Alzheimer disease: A genetic screening study of familial and sporadic cases. PLoS Med, 14, e1002270 10.1371/journal.pmed.1002270 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Latini, F. R. , Hemerly, J. P. , Freitas, B. C. , Oler, G. , Riggins, G. J. , & Cerutti, J. M. (2011). ABI3 ectopic expression reduces in vitro and in vivo cell growth properties while inducing senescence. BMC Cancer, 11, 11 10.1186/1471-2407-11-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Lavine, M. D. , & Arrizabalaga, G. (2009). Induction of mitotic S‐phase of host and neighboring cells by Toxoplasma gondii enhances parasite invasion. Molecular and Biochemical Parasitology, 164, 95–99. 10.1016/j.molbiopara.2008.11.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Lee, J. S. , Lee, Y. , André, E. A. , Lee, K. J. , Nguyen, T. , Feng, Y. , … Pak, D. T. S. (2019). Inhibition of Polo‐like kinase 2 ameliorates pathogenesis in Alzheimer's disease model mice. PLoS ONE, 14, e0219691 10.1371/journal.pone.0219691 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Lee, S. L. , Thomas, P. , & Fenech, M. (2015). Genome instability biomarkers and blood micronutrient risk profiles associated with mild cognitive impairment and Alzheimer's disease. Mutation Research, 776, 54–83. 10.1016/j.mrfmmm.2014.12.012 [DOI] [PubMed] [Google Scholar]
  100. Leggio, G. M. , Catania, M. V. , Puzzo, D. , Spatuzza, M. , Pellitteri, R. , Gulisano, W. , … Drago, F. (2016). The antineoplastic drug flavopiridol reverses memory impairment induced by Amyloid‐ß1‐42 oligomers in mice. Pharmacological Research, 106, 10–20. 10.1016/j.phrs.2016.02.007 [DOI] [PubMed] [Google Scholar]
  101. Lewis, L. E. , Bain, J. M. , Lowes, C. , Gow, N. A. , & Erwig, L. P. (2012). Candida albicans infection inhibits macrophage cell division and proliferation. Fungal Genetics and Biology, 49, 679–680. 10.1016/j.fgb.2012.05.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Li, H. , Liu, C. C. , Zheng, H. , & Huang, T. Y. (2018). Amyloid, tau, pathogen infection and antimicrobial protection in Alzheimer's disease ‐conformist, nonconformist, and realistic prospects for AD pathogenesis. Transl Neurodegener, 7, 34 10.1186/s40035-018-0139-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Li, H. , Radford, J. C. , Ragusa, M. J. , Shea, K. L. , McKercher, S. R. , Zaremba, J. D. , … Lipton, S. A. (2008). Transcription factor MEF2C influences neural stem/progenitor cell differentiation and maturation in vivo. Proceedings of the National Academy of Sciences of the United States of America, 105, 9397–9402. 10.1073/pnas.0802876105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Lipinski, C. A. , Tran, N. L. , Menashi, E. , Rohl, C. , Kloss, J. , Bay, R. C. , … Loftus, J. C. (2005). The tyrosine kinase Pyk2 promotes migration and invasion of glioma cells. Neoplasia, 7(5), 435–445. 10.1593/neo.04712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Liu, S. L. , Wang, C. , Jiang, T. , Tan, L. , Xing, A. , & Yu, J. T. (2016). The role of Cdk5 in Alzheimer's disease. Molecular Neurobiology, 53, 4328–4342. 10.1007/s12035-015-9369-x [DOI] [PubMed] [Google Scholar]
  106. Liu, S. , Zhang, W. , Liu, K. , Ji, B. , & Wang, G. (2015). Silencing ADAM10 inhibits the in vitro and in vivo growth of hepatocellular carcinoma cancer cells. Mol Med Rep, 11, 597–602. 10.3892/mmr.2014.2652 [DOI] [PubMed] [Google Scholar]
  107. Lomonte, P. , & Everett, R. D. (1999). Herpes simplex virus type 1 immediate‐early protein Vmw110 inhibits progression of cells through mitosis and from G(1) into S phase of the cell cycle. Journal of Virology, 73, 9456–9467. 10.1128/JVI.73.11.9456-9467.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Lopes, J. P. , & Agostinho, P. (2011). Cdk5: Multitasking between physiological and pathological conditions. Progress in Neurobiology, 94, 49–63. 10.1016/j.pneurobio.2011.03.006 [DOI] [PubMed] [Google Scholar]
  109. Lopes, J. P. , Blurton‐Jones, M. , Yamasaki, T. R. , Agostinho, P. , & LaFerla, F. M. (2009). Activation of cell cycle proteins in transgenic mice in response to neuronal loss but not amyloid‐beta and tau pathology. Journal of Alzheimer's Disease, 16, 541–549. 10.3233/JAD-2009-0993 [DOI] [PubMed] [Google Scholar]
  110. López‐Sánchez, N. , Fontán‐Lozano, Á. , Pallé, A. , González‐Álvarez, V. , Rábano, A. , Trejo, J. L. , & Frade, J. M. (2017). Neuronal tetraploidization in the cerebral cortex correlates with reduced cognition in mice and precedes and recapitulates Alzheimer's‐associated neuropathology. Neurobiology of Aging, 56, 50–66. 10.1016/j.neurobiolaging.2017.04.008 [DOI] [PubMed] [Google Scholar]
  111. López‐Sánchez, N. , Müller, U. , & Frade, J. M. (2005). Lengthening of G2/mitosis in cortical precursors from mice lacking beta‐amyloid precursor protein. Neuroscience, 130, 51–60. [DOI] [PubMed] [Google Scholar]
  112. Lott, I. T. , & Head, E. (2019). Dementia in down syndrome: Unique insights for Alzheimer disease research. Nature Reviews Neurology, 15, 135–147. 10.1038/s41582-018-0132-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Lovestone, S. , Boada, M. , Dubois, B. , Hüll, M. , Rinne, J. O. , Huppertz, H.‐J. , … del Ser, T. (2015). A phase II trial of tideglusib in Alzheimer's disease. Journal of Alzheimer's Disease, 45, 75–88. 10.3233/JAD-141959 [DOI] [PubMed] [Google Scholar]
  114. Mairet‐Coello, G. , & Polleux, F. (2014). Involvement of ‘stress‐response’ kinase pathways in Alzheimer's disease progression. Current Opinion in Neurobiology, 27, 110–117. 10.1016/j.conb.2014.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Makani, V. , Zhang, B. , Han, H. , Yao, Y. , Lassalas, P. , Lou, K. , … Brunden, K. R. (2016). Evaluation of the brain‐penetrant microtubule‐stabilizing agent, dictyostatin, in the PS19 tau transgenic mouse model of tauopathy. Acta Neuropathol Commun, 4, 106 10.1186/s40478-016-0378-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Matsos, A. , & Johnston, I. N. (2019). Chemotherapy‐induced cognitive impairments: A systematic review of the animal literature. Neuroscience and Biobehavioral Reviews, 102, 382–399. 10.1016/j.neubiorev.2019.05.001 [DOI] [PubMed] [Google Scholar]
  117. Mawuenyega, K. G. , Sigurdson, W. , Ovod, V. , Munsell, L. , Kasten, T. , Morris, J. C. , … Bateman, R. J. (2010). Decreased clearance of CNS beta‐amyloid in Alzheimer's disease. Science, 330, 1774 10.1126/science.1197623 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Meng, X.‐Q. , Cui, B. , Cheng, D. , Lyu, H. , Jiang, L.‐G. , Zheng, K.‐G. , … Zhou, J. (2018). Activated proline‐rich tyrosine kinase 2 regulates meiotic spindle assembly in the mouse oocyte. Journal of Cellular Biochemistry, 119, 736–747. 10.1002/jcb.26237 [DOI] [PubMed] [Google Scholar]
  119. Miao, H. , Wei, B. R. , Peehl, D. M. , Li, Q. , Alexandrou, T. , Schelling, J. R. , … Wang, B. (2001). Activation of EphA receptor tyrosine kinase inhibits the Ras/MAPK pathway. Nature Cell Biology, 3(5), 527–530. 10.1038/35074604 [DOI] [PubMed] [Google Scholar]
  120. Miners, J. S. , Morris, S. , Love, S. , & Kehoe, P. G. (2011). Accumulation of insoluble amyloid‐β in down's syndrome is associated with increased BACE‐1 and neprilysin activities. Journal of Alzheimer's Disease, 23, 101–108. 10.3233/JAD-2010-101395 [DOI] [PubMed] [Google Scholar]
  121. Mingari, M. C. , Vitale, C. , Romagnani, C. , Falco, M. , & Moretta, L. (2001). p75/AIRM1 and CD33, two sialoadhesin receptors that regulate the proliferation or the survival of normal and leukemic myeloid cells. Immunological Reviews, 181, 260–268. 10.1034/j.1600-065X.2001.1810122.x [DOI] [PubMed] [Google Scholar]
  122. Mohamed, W. A. , Salama, R. M. , & Schaalan, M. F. (2019). A pilot study on the effect of lactoferrin on Alzheimer's disease pathological sequelae: Impact of the p‐Akt/PTEN pathway. Biomedicine & Pharmacotherapy, 111, 714–723. 10.1016/j.biopha.2018.12.118 [DOI] [PubMed] [Google Scholar]
  123. Monaco, E. A. 3rd , & Vallano, M. L. (2005). Role of protein kinases in neurodegenerative disease: Cyclin‐dependent kinases in Alzheimer's disease. Frontiers in Bioscience, 10, 143–159. 10.2741/1516 [DOI] [PubMed] [Google Scholar]
  124. Monzo, P. , Gauthier, N. C. , Keslair, F. , Loubat, A. , Field, C. M. , Le Marchand‐Brustel, Y. , & Cormont, M. (2005). Clues to CD2‐associated protein involvement in cytokinesis. Molecular Biology of the Cell, 16, 2891–2902. 10.1091/mbc.e04-09-0773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Morroni, F. , Sita, G. , Tarozzi, A. , Rimondini, R. , & Hrelia, P. (2016). Early effects of Aβ1‐42 oligomers injection in mice: Involvement of PI3K/Akt/GSK3 and MAPK/ERK1/2 pathways. Behavioral Brain Research, 314, 106–115. 10.1016/j.bbr.2016.08.002 [DOI] [PubMed] [Google Scholar]
  126. Moussa‐Pacha, N. M. , Abdin, S. M. , Omar, H. A. , Alniss, H. , & Al‐Tel, T. H. (2019). BACE1 inhibitors: Current status and future directions in treating Alzheimer's disease. Medicinal Research Reviews, 40(1), 339–384. 10.1002/med.21622 [DOI] [PubMed] [Google Scholar]
  127. Muresan, Z. , & Muresan, V. (2007). The amyloid‐beta precursor protein is phosphorylated via distinct pathways during differentiation, mitosis, stress, and degeneration. Molecular Biology of the Cell, 18, 3835–3844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Murray, A. , & Hunt, T. (1993). The cell cycle: An introduction. New York, NY: Oxford University Pres. [Google Scholar]
  129. Murray, M. E. , Graff‐Radford, N. R. , Ross, O. A. , Petersen, R. C. , Duara, R. , & Dickson, D. W. (2011). Neuropathologically defined subtypes of Alzheimer's disease with distinct clinical characteristics: A retrospective study. The Lancet Neurology, 10, 785–796. 10.1016/S1474-4422(11)70156-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Mushtaq, G. , Greig, N. H. , Anwar, F. , Al‐Abbasi, F. A. , Zamzami, M. A. , Al‐Talhi, H. A. , & Kamal, M. A. (2016). Neuroprotective mechanisms mediated by CDK5 inhibition. Current Pharmaceutical Design, 22, 527–534. 10.2174/1381612822666151124235028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Nelson, T. J. , Sun, M. K. , Lim, C. , Sen, A. , Khan, T. , Chirila, F. V. , & Alkon, D. L. (2017). Bryostatin effects on cognitive function and PKCɛ in Alzheimer's disease phase IIa and expanded access trials. Journal of Alzheimer's Disease, 58, 521–535. 10.3233/JAD-170161 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Neve, R. L. , & McPhie, D. L. (2006). The cell cycle as a therapeutic target for Alzheimer's disease. Pharmacology & Therapeutics, 111, 99–113. 10.1016/j.pharmthera.2005.09.005 [DOI] [PubMed] [Google Scholar]
  133. Nordengen, K. , Kirsebom, B. E. , Henjum, K. , Selnes, P. , Gísladóttir, B. , Wettergreen, M. , … Fladby, T. (2019). Glial activation and inflammation along the Alzheimer's disease continuum. Journal of Neuroinflammation, 16, 46 10.1186/s12974-019-1399-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Nurse, P. (2012). Finding CDK: Linking yeast with humans. Nature Cell Biology, 14, 776 10.1038/ncb2547 [DOI] [PubMed] [Google Scholar]
  135. Ovsepian, S. V. , & O'Leary, V. B. (2016). Neuronal activity and amyloid plaque pathology: An update. Journal of Alzheimer's Disease, 49, 13–19. 10.3233/JAD-150544 [DOI] [PubMed] [Google Scholar]
  136. Pan, K. E. , Liang, X.‐T. , Zhang, H.‐K. , Zhao, J.‐J. , Wang, D.‐D. , Li, J.‐J. , … Xia, J.‐C. (2012). Characterization of bridging integrator 1 (BIN1) as a potential tumor suppressor and prognostic marker in hepatocellular carcinoma. Molecular Medicine, 18, 507–518. 10.2119/molmed.2011.00319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Park, K. H. , Hallows, J. L. , Chakrabarty, P. , Davies, P. , & Vincent, I. (2007). Conditional neuronal simian virus 40 T antigen expression induces Alzheimer‐like tau and amyloid pathology in mice. Journal of Neuroscience, 27, 2969–2978. 10.1523/JNEUROSCI.0186-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Pavlova, G. A. , Popova, J. V. , Andreyeva, E. N. , Yarinich, L. A. , Lebedev, M. O. , Razuvaeva, A. V. , … Gatti, M. (2019). RNAi‐mediated depletion of the NSL complex subunits leads to abnormal chromosome segregation and defective centrosome duplication in Drosophila mitosis. PLoS Genetics, 15, e1008371 10.1371/journal.pgen.1008371 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Penke, B. , Bogár, F. , & Fülöp, L. (2017). β‐amyloid and the pathomechanisms of Alzheimer's disease: A comprehensive view. Molecules, 22, E1692 10.3390/molecules22101692 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Pérez‐Nievas, B. G. , & Serrano‐Pozo, A. (2019). Editorial: The role of glia in Alzheimer's disease. Front Neurol, 9, 1161 10.3389/fneur.2018.01161 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Perrin, R. J. , Fagan, A. M. , & Holtzman, D. M. (2009). Multimodal techniques for diagnosis and prognosis of Alzheimer's disease. Nature, 461, 916–922. 10.1038/nature08538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Petrozzi, L. , Lucetti, C. , Scarpato, R. , Gambaccini, G. , Trippi, F. , Bernardini, S. , … Bonuccelli, U. (2002). Cytogenetic alterations in lymphocytes of Alzheimer's disease and Parkinson's disease patients. Neurol Sci, 23(Suppl 2), S97–S98. 10.1007/s100720200087 [DOI] [PubMed] [Google Scholar]
  143. Pope, W. B. , Lambert, M. P. , Leypold, B. , Seupaul, R. , Sletten, L. , Krafft, G. , & Klein, W. L. (1994). Microtubule‐associated protein tau is hyperphosphorylated during mitosis in the human neuroblastoma cell line SH‐SY5Y. Experimental Neurology, 126(2), 185–194. 10.1006/exnr.1994.1057 [DOI] [PubMed] [Google Scholar]
  144. Potter, H. (1991). Review and hypothesis: Alzheimer disease and Down syndrome–chromosome 21 nondisjunction may underlie both disorders. American Journal of Human Genetics, 48, 1192–1200. [PMC free article] [PubMed] [Google Scholar]
  145. Preuss, U. , & Mandelkow, E. M. (1998). Mitotic phosphorylation of tau protein in neuronal cell lines resembles phosphorylation in Alzheimer's disease. European Journal of Cell Biology, 76, 176–184. 10.1016/S0171-9335(98)80032-0 [DOI] [PubMed] [Google Scholar]
  146. Rao, C. V. , Farooqui, M. , Asch, A. S. , & Yamada, H. Y. (2018). Critical role of mitosis in spontaneous late‐onset Alzheimer's disease; from a Shugoshin 1 cohesinopathy mouse model. Cell Cycle, 17, 2321–2334. 10.1080/15384101.2018.1515554 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Rao, C. V. , Farooqui, M. , Zhang, Y. , Asch, A. S. , & Yamada, H. Y. (2018). Spontaneous development of Alzheimer's disease‐associated brain pathology in a Shugoshin‐1 mouse cohesinopathy model. Aging Cell, 17, e12797 10.1111/acel.12797 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Rao, C. V. , & Yamada, H. Y. (2013). Genomic instability and colon carcinogenesis: From the perspective of genes. Frontiers in Oncology, 3, 130 10.3389/fonc.2013.00130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Rao, C. V. , Yamada, H. Y. , Yao, Y. , & Dai, W. (2009). Enhanced genomic instabilities caused by deregulated microtubule dynamics and chromosome segregation: A perspective from genetic studies in mice. Carcinogenesis, 30, 1469–1474. 10.1093/carcin/bgp081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Readhead, B. , Haure‐Mirande, J.‐V. , Funk, C. C. , Richards, M. A. , Shannon, P. , Haroutunian, V. , … Dudley, J. T. (2018). Multiscale analysis of independent Alzheimer's cohorts finds disruption of molecular, genetic, and clinical networks by human herpesvirus. Neuron, 99, 64–82. 10.1016/j.neuron.2018.05.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Ren, X. , Boriero, D. , Chaiswing, L. , Bondada, S. , St Clair, D. K. , & Butterfield, D. A. (2019). Plausible biochemical mechanisms of chemotherapy‐induced cognitive impairment (“chemobrain”), a condition that significantly impairs the quality of life of many cancer survivors. Biochimica et Biophysica Acta (BBA) – Molecular Basis of Disease, 1865, 1088–1097. 10.1016/j.bbadis.2019.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Ren, X. , St Clair, D. K. , & Butterfield, D. A. (2017). Dysregulation of cytokine mediated chemotherapy induced cognitive impairment. Pharmacological Research, 117, 267–273. 10.1016/j.phrs.2017.01.001 [DOI] [PubMed] [Google Scholar]
  153. Saito, T. , & Saido, T. C. (2018). Neuroinflammation in mouse models of Alzheimer's disease. Clinical and Experimental Neuroimmunology, 9, 211–218. 10.1111/cen3.12475 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Schemmert, S. , Schartmann, E. , Zafiu, C. , Kass, B. , Hartwig, S. , Lehr, S. , … Willbold, D. (2019). Aβ oligomer elimination restores cognition in transgenic Alzheimer's mice with full‐blown pathology. Molecular Neurobiology, 56, 2211–2223. 10.1007/s12035-018-1209-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Seib, D. R. M. , Corsini, N. S. , Ellwanger, K. , Plaas, C. , Mateos, A. , Pitzer, C. , … Martin‐Villalba, A. (2013). Loss of Dickkopf‐1 restores neurogenesis in old age and counteracts cognitive decline. Cell Stem Cell, 12, 204–214. 10.1016/j.stem.2012.11.010 [DOI] [PubMed] [Google Scholar]
  156. Selkoe, D. J. (2001). Alzheimer's disease: Genes, proteins, and therapy. Physiological Reviews, 81, 741–766. 10.1152/physrev.2001.81.2.741 [DOI] [PubMed] [Google Scholar]
  157. Selkoe, D. J. , & Hardy, J. (2016). The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Molecular Medicine, 8, 595–608. 10.15252/emmm.201606210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Senderowicz, A. M. (1999). Flavopiridol: The first cyclin‐dependent kinase inhibitor in human clinical trials. Investigational New Drugs, 17, 313–320. [DOI] [PubMed] [Google Scholar]
  159. Shafi, O. (2016). Inverse relationship between Alzheimer's disease and cancer, and other factors contributing to Alzheimer's disease: A systematic review. BMC Neurol, 16, 236 10.1186/s12883-016-0765-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Shannan, B. , Seifert, M. , Boothman, D. A. , Tilgen, W. , & Reichrath, J. (2006). Clusterin and DNA repair: A new function in cancer for a key player in apoptosis and cell cycle control. Journal of Molecular Histology, 37, 183–188. 10.1007/s10735-006-9052-7 [DOI] [PubMed] [Google Scholar]
  161. Shemesh, O. A. , & Spira, M. E. (2011). Rescue of neurons from undergoing hallmark tau‐induced Alzheimer's disease cell pathologies by the antimitotic drug paclitaxel. Neurobiology of Diseases, 43, 163–175. 10.1016/j.nbd.2011.03.008 [DOI] [PubMed] [Google Scholar]
  162. Shepherd, C. E. , Yang, Y. , & Halliday, G. M. (2018). Region‐ and cell‐specific aneuploidy in brain aging and neurodegeneration. Neuroscience, 374, 326–334. 10.1016/j.neuroscience.2018.01.050 [DOI] [PubMed] [Google Scholar]
  163. Sikanyika, N. L. , Parkington, H. C. , Smith, A. I. , & Kuruppu, S. (2019). Powering amyloid beta degrading enzymes: A possible therapy for Alzheimer's disease. Neurochemical Research, 44, 1289–1296. 10.1007/s11064-019-02756-x [DOI] [PubMed] [Google Scholar]
  164. Simon, J. E. , Bakker, B. , & Foijer, F. (2015). CINcere modelling: What have mouse models for chromosome instability taught us? Recent Results in Cancer Research, 200, 39–60. 10.1007/978-3-319-20291-4_2 [DOI] [PubMed] [Google Scholar]
  165. Snape, K. , Hanks, S. , Ruark, E. , Barros‐Núñez, P. , Elliott, A. , Murray, A. , … Rahman, N. (2011). Mutations in CEP57 cause mosaic variegated aneuploidy syndrome. Nature Genetics, 43, 527–529. 10.1038/ng.822 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Sochocka, M. , Zwolińska, K. , & Leszek, J. (2017). The infectious etiology of Alzheimer's disease. Current Neuropharmacology, 15, 996–1009. 10.2174/1570159X15666170313122937 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Sun, A. , Liu, M. , Nguyen, X. V. , & Bing, G. (2003). P38 MAP kinase is activated at early stages in Alzheimer's disease brain. Experimental Neurology, 183, 394–405. 10.1016/S0014-4886(03)00180-8 [DOI] [PubMed] [Google Scholar]
  168. Sun, H. S. , Sin, A. T. , Poirier, M. B. , & Harrison, R. E. (2016). Chlamydia trachomatis inclusion disrupts host cell cytokinesis to enhance its growth in multinuclear cells. Journal of Cellular Biochemistry, 117, 132–143. 10.1002/jcb.25258 [DOI] [PubMed] [Google Scholar]
  169. Suzuki, A. , Pelikan, R. C. , & Iwata, J. (2015). WNT/β‐catenin signaling regulates multiple steps of myogenesis by regulating step‐specific targets. Molecular and Cellular Biology, 35, 1763–1776. 10.1128/MCB.01180-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Suzuki, T. , & Nakaya, T. (2008). Regulation of amyloid beta‐protein precursor by phosphorylation and protein interactions. The Journal of Biological Chemistry, 283, 29633–29637. 10.1074/jbc.R800003200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Suzuki, T. , Oishi, M. , Marshak, D. R. , Czernik, A. J. , Nairn, A. C. , & Greengard, P. (1994). Cell cycle‐dependent regulation of the phosphorylation and metabolism of the Alzheimer amyloid precursor protein. EMBO Journal, 13, 1114–1122. 10.1002/j.1460-2075.1994.tb06360.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Swatton, J. E. , Sellers, L. A. , Faull, R. L. , Holland, A. , Iritani, S. , & Bahn, S. (2004). Increased MAP kinase activity in Alzheimer's and Down syndrome but not in schizophrenia human brain. European Journal of Neuroscience, 19, 2711–2719. 10.1111/j.0953-816X.2004.03365.x [DOI] [PubMed] [Google Scholar]
  173. Tatebayashi, Y. , Planel, E. , Chui, D.‐H. , Sato, S. , Miyasaka, T. , Sahara, N. , … Takashima, A. (2006). c‐jun N‐terminal kinase hyperphosphorylates R406W tau at the PHF‐1 site during mitosis. The FASEB Journal, 20, 762–764. 10.1096/fj.05-4362fje [DOI] [PubMed] [Google Scholar]
  174. Thal, D. R. , Rüb, U. , Orantes, M. , & Braak, H. (2002). Phases of A beta‐deposition in the human brain and its relevance for the development of AD. Neurology, 58, 1791–1800. [DOI] [PubMed] [Google Scholar]
  175. Tomashevski, A. , Husseman, J. , Jin, L. W. , Nochlin, D. , & Vincent, I. (2001). Constitutive Wee1 activity in adult brain neurons with M phase‐type alterations in Alzheimer neurodegeneration. Journal of Alzheimer's Disease, 3, 195–207. 10.3233/JAD-2001-3205 [DOI] [PubMed] [Google Scholar]
  176. Tremlett, H. , Bauer, K. C. , Appel‐Cresswell, S. , Finlay, B. B. , & Waubant, E. (2017). The gut microbiome in human neurological disease: A review. Annals of Neurology, 81, 369–382. 10.1002/ana.24901 [DOI] [PubMed] [Google Scholar]
  177. Trippi, F. , Botto, N. , Scarpato, R. , Petrozzi, L. , Bonuccelli, U. , Latorraca, S. , … Migliore, L. (2002). Spontaneous and induced chromosome damage in somatic cells of sporadic and familial Alzheimer's disease patients. Mutagenesis, 16, 323–327. 10.1093/mutage/16.4.323 [DOI] [PubMed] [Google Scholar]
  178. Trojanowski, J. Q. , Smith, A. B. , Huryn, D. , & Lee, V. M. (2005). Microtubule‐stabilising drugs for therapy of Alzheimer's disease and other neurodegenerative disorders with axonal transport impairments. Expert Opinion on Pharmacotherapy, 6, 683–686. 10.1517/14656566.6.5.683 [DOI] [PubMed] [Google Scholar]
  179. Tsai, R. M. , Miller, Z. , Koestler, M. , Rojas, J. C. , Ljubenkov, P. A. , Rosen, H. J. , & Boxer, A. L. (2019). Reactions to multiple ascending doses of the microtubule stabilizer TPI‐287 in patients with Alzheimer disease, progressive supranuclear palsy, and corticobasal syndrome. JAMA Neurology, [Epub ahead of print]. doi: 10.1001/jamaneurol.2019.3812 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Ueno, N. , Nishimura, N. , Ueno, S. , Endo, S. , Tatetsu, H. , Hirata, S. , … Okuno, Y. (2017). PU.1 acts as tumor suppressor for myeloma cells through direct transcriptional repression of IRF4. Oncogene, 36, 4481–4497. 10.1038/onc.2017.79 [DOI] [PubMed] [Google Scholar]
  181. Umeda, M. , Murata‐Kamiya, N. , Saito, Y. , Ohba, Y. , Takahashi, M. , & Hatakeyama, M. (2009). Helicobacter pylori CagA causes mitotic impairment and induces chromosomal instability. Journal of Biological Chemistry, 284, 22166–22172. 10.1074/jbc.M109.035766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Valles, S. L. , Iradi, A. , Aldasoro, M. , Vila, J. M. , Aldasoro, C. , Torre, J. D. L. , … Jorda, A. (2019). Function of glia in aging and the brain diseases. Internation Journal of Medical Sciences, 16, 1473–1479. 10.7150/ijms.37769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Van Belle, E. , Meurice, T. , Tio, F. O. , Corseaux, D. , Dupuis, B. , McFadden, E. P. , … Bertrand, M. E. (1997). ACE inhibition accelerates endothelial regrowth in vivo: A possible explanation for the benefit observed with ACE inhibitors following arterial injury. Biochemical and Biophysical Research Communications, 231, 577–581. 10.1006/bbrc.1997.6061 [DOI] [PubMed] [Google Scholar]
  184. Van Cauwenberghe, C. , Van Broeckhoven, C. , & Sleegers, K. (2016). The genetic landscape of Alzheimer disease: Clinical implications and perspectives. Genetics in Medicine, 18, 421–430. 10.1038/gim.2015.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. van den Bos, H. , Spierings, C. J. , Taudt, A. , Bakker, B. , Porubský, D. , Falconer, E. , … & Landsdorp, P. M. (2016). Single-cell whole genome sequencing reveals no evidence for common aneuploidy in normal and Alzheimer’s disease neurons. Genome Biol., 17, 116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Vargas, L. M. , Cerpa, W. , Muñoz, F. J. , Zanlungo, S. , & Alvarez, A. R. (2018). Amyloid‐β oligomers synaptotoxicity: The emerging role of EphA4/c‐Abl signaling in Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis, 1864, 1148–1159. 10.1016/j.bbadis.2018.01.023 [DOI] [PubMed] [Google Scholar]
  187. Varvel, N. H. , Bhaskar, K. , Patil, A. R. , Pimplikar, S. W. , Herrup, K. , & Lamb, B. T. (2008). Abeta oligomers induce neuronal cell cycle events in Alzheimer's disease. The Journal of Neuroscience, 28, 10786–10793. 10.1016/j.nbd.2013.10.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Vieira, S. I. , Rebelo, S. , Domingues, S. C. , da Cruz e Silva, E. F. , & da Cruz e Silva, O. A. B. (2009). S655 phosphorylation enhances APP secretory traffic. Molecular and Cellular Biochemistry, 328, 145–154. 10.1007/s11010-009-0084-7 [DOI] [PubMed] [Google Scholar]
  189. Vincent, I. , Bu, B. , Hudson, K. , Husseman, J. , Nochlin, D. , & Jin, L. (2001). Constitutive Cdc25B tyrosine phosphatase activity in adult brain neurons with M phase‐type alterations in Alzheimer's disease. Neuroscience, 105, 639–650. 10.1016/S0306-4522(01)00219-6 [DOI] [PubMed] [Google Scholar]
  190. Vincent, I. , Jicha, G. , Rosado, M. , & Dickson, D. W. (1997). Aberrant expression of mitotic cdc2/cyclin B1 kinase in degenerating neurons of Alzheimer's disease brain. Journal of Neuroscience, 17, 3588–3598. 10.1523/JNEUROSCI.17-10-03588.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Vincent, I. , Zheng, J. H. , Dickson, D. W. , Kress, Y. , & Davies, P. (1998). Mitotic phosphoepitopes precede paired helical filaments in Alzheimer's disease. Neurobiology of Aging, 19, 287–296. 10.1016/S0197-4580(98)00071-2 [DOI] [PubMed] [Google Scholar]
  192. Vingtdeux, V. , Davies, P. , Dickson, D. W. , & Marambaud, P. (2011). AMPK is abnormally activated in tangle‐ and pre‐tangle‐bearing neurons in Alzheimer's disease and other tauopathies. Acta Neuropathologica, 121(3), 337–349. 10.1007/s00401-010-0759-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Wan, X. , Huang, W. , Yang, S. , Zhang, Y. , Pu, H. , Fu, F. , … Li, Y. (2016). Identification of androgen‐responsive lncRNAs as diagnostic and prognostic markers for prostate cancer. Oncotarget, 7, 60503–60518. 10.18632/oncotarget.11391 [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Wang, X. Q. , Tao, B. B. , Li, B. , Wang, X. H. , Zhang, W. C. , Wan, L. , … Li, S. T. (2016). Overexpression of TREM2 enhances glioma cell proliferation and invasion: A therapeutic target in human glioma. Oncotarget, 7, 2354–2366. 10.18632/oncotarget.6221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Wangsa, D. , Braun, R. , Stuelten, C. H. , Brown, M. , Bauer, K. M. , Emons, G. , … Ried, T. (2019). Induced chromosomal aneuploidy results in global and consistent deregulation of the transcriptome of cancer cells. Neoplasia, 21, 721–729. 10.1016/j.neo.2019.04.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Weaver, B. A. , & Cleveland, D. W. (2009). The role of aneuploidy in promoting and suppressing tumors. Journal of Cell Biology, 185, 935–937. 10.1083/jcb.200905098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Webber, K. M. , Raina, A. K. , Marlatt, M. W. , Zhu, X. , Prat, M. I. , Morelli, L. , … Smith, M. A. (2005). The cell cycle in Alzheimer disease: A unique target for neuropharmacology. Mechanisms of Ageing and Development, 126, 1019–1025. 10.1016/j.mad.2005.03.024 [DOI] [PubMed] [Google Scholar]
  198. Weber, S. A. , Patel, R. K. , & Lutsep, H. L. (2018). Cerebral amyloid angiopathy: Diagnosis and potential therapies. Expert Review of Neurotherapeutics, 18, 503–513. 10.1080/14737175.2018.1480938 [DOI] [PubMed] [Google Scholar]
  199. Westra, J. W. , Barral, S. , & Chun, J. (2009). A reevaluation of tetraploidy in the Alzheimer's disease brain. Neuro‐Degenerative Diseases, 6, 221–229. 10.1159/000236901 [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Wilker, P. R. , Kohyama, M. , Sandau, M. M. , Albring, J. C. , Nakagawa, O. , Schwarz, J. J. , & Murphy, K. M. (2008). Transcription factor Mef2c is required for B cell proliferation and survival after antigen receptor stimulation. Nature Immunology, 9, 603–612. 10.1038/ni.1609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Wiseman, F. K. , Pulford, L. J. , Barkus, C. , Liao, F. , Portelius, E. , Webb, J. J. , … Fisher, E. M. C. (2018). Trisomy of human chromosome 21 enhances amyloid‐β deposition independently of an extra copy of APP. Brain, 141, 2457–2474. 10.1093/brain/awy159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Wojsiat, J. , Prandelli, C. , Laskowska‐Kaszub, K. , Martín‐Requero, A. , & Wojda, U. (2015). Oxidative stress and aberrant cell cycle in Alzheimer's disease lymphocytes: Diagnostic prospects. Journal of Alzheimer's Disease, 46(2), 329–350. 10.3233/JAD-141977 [DOI] [PubMed] [Google Scholar]
  203. Xia, L. , Sun, C. , Li, Q. , Feng, F. , Qiao, E. , Jiang, L. , … Ge, M. (2015). CELF1 is up‐regulated in glioma and promotes glioma cell proliferation by suppression of CDKN1B. International Journal of Biological Sciences, 11, 1314–1324. 10.7150/ijbs.11344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Xiang, Z. , Ho, L. , Valdellon, J. , Borchelt, D. , Kelley, K. , Spielman, L. , & Pasinetti, G. M. (2002). Cyclooxygenase (COX)‐2 and cell cycle activity in a transgenic mouse model of Alzheimer's disease neuropathology. Neurobiology of Aging, 23, 327–334. 10.1016/S0197-4580(01)00282-2 [DOI] [PubMed] [Google Scholar]
  205. Yamazaki, Y. , Zhao, N. , Caulfield, T. R. , Liu, C.‐C. , & Bu, G. (2019). Apolipoprotein E and Alzheimer disease: Pathobiology and targeting strategies. Nature Reviews Neurology, 15(9), 501–518. 10.1038/s41582-019-0228-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Yang, Z. , Jun, H. , Choi, C.‐I I. , Yoo, K. H. , Cho, C. H. , Hussaini, S. M. Q. , … Jang, M.‐H. (2017). Age‐related decline in BubR1 impairs adult hippocampal neurogenesis. Aging Cell, 16, 598–601. 10.1111/acel.12594 [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Yu, J. T. , & Tan, L. (2012). The role of clusterin in Alzheimer's disease: Pathways, pathogenesis, and therapy. Molecular Neurobiology, 45, 314–326. 10.1007/s12035-012-8237-1 [DOI] [PubMed] [Google Scholar]
  208. Yurov, Y. B. , Vorsanova, S. G. , & Iourov, I. Y. (2011). The DNA replication stress hypothesis of Alzheimer's disease. Scientific World Journal, 11, 2602–2612. 10.1100/2011/625690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Zasadil, L. M. , Britigan, E. M. C. , Ryan, S. D. , Kaur, C. , Guckenberger, D. J. , Beebe, D. J. , … Weaver, B. A. (2016). High rates of chromosome missegregation suppress tumor progression but do not inhibit tumor initiation. Molecular Biology of the Cell, 27, 1981–1989. 10.1091/mbc.E15-10-0747 [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Zhang, B. , Carroll, J. , Trojanowski, J. Q. , Yao, Y. , Iba, M. , Potuzak, J. S. , … Brunden, K. R. (2012). The microtubule‐stabilizing agent, epothilone D, reduces axonal dysfunction, neurotoxicity, cognitive deficits, and Alzheimer‐like pathology in an interventional study with aged tau transgenic mice. Journal of Neuroscience, 32, 3601–3611. 10.1523/JNEUROSCI.4922-11.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Zhang, B. , Gaiteri, C. , Bodea, L.‐G. , Wang, Z. , McElwee, J. , Podtelezhnikov, A. A. , … Emilsson, V. . (2013). Integrated systems approach identifies genetic nodes and networks in late‐onset Alzheimer's disease. Cell, 153, 707–720. 10.1016/j.cell.2013.03.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Zhang, Q. , Guo, S. , Zhang, X. , Tang, S. , Shao, W. , Han, X. , … Du, Y. (2015). Inverse relationship between cancer and Alzheimer's disease: A systemic review meta‐analysis. Neurological Sciences, 36, 1987–1994. 10.1007/s10072-015-2282-2 [DOI] [PubMed] [Google Scholar]
  213. Zhang, S. , Liu, X. , Zhang, Y. , Cheng, Y. , & Li, Y. (2013). RNAi screening identifies KAT8 as a key molecule important for cancer cell survival. International Journal of Clinical and Experimental Pathology, 6, 870–877. [PMC free article] [PubMed] [Google Scholar]
  214. Zhang, X. , Hernandez, I. , Rei, D. , Mair, W. , Laha, J. K. , Cornwell, M. E. , … Kosik, K. S. (2013). Diaminothiazoles modify Tau phosphorylation and improve the tauopathy in mouse models. The Journal of Biological Chemistry, 288, 22042–22056. 10.1074/jbc.M112.436402 [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Zhang, Y. , Zhang, Y. , Xiao, Y. , Zhong, C. , & Xiao, F. (2019). Expression of Clusterin suppresses Cr(VI)‐induced premature senescence through activation of PI3K/AKT pathway. Ecotoxicology and Environmental Safety, 183, 109465 10.1016/j.ecoenv.2019.109465 [DOI] [PubMed] [Google Scholar]
  216. Zheng, H. , Jia, L. , Liu, C.‐C. , Rong, Z. , Zhong, L. I. , Yang, L. , … Bu, G. (2017). TREM2 promotes microglial survival by activating Wnt/β‐catenin pathway. Journal of Neuroscience, 37, 1772–1784. 10.1523/JNEUROSCI.2459-16.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Zhong, L. I. , Zhang, Z.‐L. , Li, X. , Liao, C. , Mou, P. , Wang, T. , … Chen, X.‐F. (2017). TREM2/DAP12 complex regulates inflammatory responses in microglia via the JNK signaling pathway. Frontiers in Aging Neuroscience, 9, 204 10.3389/fnagi.2017.00204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Zhou, J. , Wu, J. , Li, B. , Liu, D. , Yu, J. , Yan, X. , … Wang, Q.‐F. (2014). PU.1 is essential for MLL leukemia partially via crosstalk with the MEIS/HOX pathway. Leukemia, 28, 1436–1448. 10.1038/leu.2013.384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Zhu, X. , Lee, H. G. , Perry, G. , & Smith, M. A. (2007). Alzheimer disease, the two‐hit hypothesis: An update. Biochimica et Biophysica Acta, 1772, 494–502. 10.1016/j.bbadis.2006.10.014 [DOI] [PubMed] [Google Scholar]
  220. Zhu, X. , Raina, A. K. , Perry, G. , & Smith, M. A. (2004). Alzheimer's disease: The two‐hit hypothesis. The Lancet Neurology, 3, 219–226. 10.1016/S1474-4422(04)00707-0 [DOI] [PubMed] [Google Scholar]
  221. Zhu, X. , Rottkamp, C. A. , Raina, A. K. , Brewer, G. J. , Ghanbari, H. A. , Boux, H. , & Smith, M. A. (2000). Neuronal CDK7 in hippocampus is related to aging and Alzheimer disease. Neurobiology of Aging, 21, 807–813. 10.1016/S0197-4580(00)00217-7 [DOI] [PubMed] [Google Scholar]
  222. Zhu, Z. , & Wang, X. (2019). Roles of cohesin in chromosome architecture and gene expression. Seminars in Cell & Developmental Biology, 90, 187–193. 10.1016/j.semcdb.2018.08.004 [DOI] [PubMed] [Google Scholar]
  223. Zivkovic, L. , Spremo‐Potparevic, B. , Plecas‐Solarovic, B. , Djelic, N. , Ocic, G. , Smiljkovic, P. , … Bajic, V. (2010). Premature centromere division of metaphase chromosomes in peripheral blood lymphocytes of Alzheimer's disease patients: Relation to gender and age. Journals of Gerontology. Series A, Biological Sciences and Medical Sciences, 65, 1269–1274. 10.1093/gerona/glq148 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Aging Cell are provided here courtesy of Wiley

RESOURCES