You are on page 1of 31

Am J Neurodegener Dis 2013;2(3):145-175

www.AJND.us /ISSN:2165-591X/AJND1307002

Review Article
Pathways to neurodegeneration: mechanistic insights
from GWAS in Alzheimers disease, Parkinsons
disease, and related disorders
Vijay K Ramanan1,2,3, Andrew J Saykin1,2,4,5
1
Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine,
Indianapolis, IN, USA; 2Department of Medical and Molecular Genetics, Indiana University School of Medicine,
Indianapolis, IN, USA; 3Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN,
USA; 4Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis,
IN, USA; 5Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
Received July 11, 2013; Accepted August 25, 2013; Epub September 18, 2013; Published September 30, 2013

Abstract: The discovery of causative genetic mutations in affected family members has historically dominated our
understanding of neurodegenerative diseases such as Alzheimers disease (AD), Parkinsons disease (PD), fronto-
temporal dementia (FTD), and amyotrophic lateral sclerosis (ALS). Nevertheless, most cases of neurodegenerative
disease are not explained by Mendelian inheritance of known genetic variants, but instead are thought to have a
complex etiology with numerous genetic and environmental factors contributing to susceptibility. Although unbiased
genome-wide association studies (GWAS) have identified novel associations to neurodegenerative diseases, most
of these hits explain only modest fractions of disease heritability. In addition, despite the substantial overlap of clini-
cal and pathologic features among major neurodegenerative diseases, surprisingly few GWAS-implicated variants
appear to exhibit cross-disease association. These realities suggest limitations of the focus on individual genetic
variants and create challenges for the development of diagnostic and therapeutic strategies, which traditionally
target an isolated molecule or mechanistic step. Recently, GWAS of complex diseases and traits have focused less
on individual susceptibility variants and instead have emphasized the biological pathways and networks revealed
by genetic associations. This new paradigm draws on the hypothesis that fundamental disease processes may be
influenced on a personalized basis by a combination of variants some common and others rare, some protective
and others deleterious in key genes and pathways. Here, we review and synthesize the major pathways implicated
in neurodegeneration, focusing on GWAS from the most prevalent neurodegenerative disorders, AD and PD. Using
literature mining, we also discover a novel regulatory network that is enriched with AD- and PD-associated genes
and centered on the SP1 and AP-1 (Jun/Fos) transcription factors. Overall, this pathway- and network-driven model
highlights several potential shared mechanisms in AD and PD that will inform future studies of these and other
neurodegenerative disorders. These insights also suggest that biomarker and treatment strategies may require
simultaneous targeting of multiple components, including some specific to disease stage, in order to assess and
modulate neurodegeneration. Pathways and networks will provide ideal vehicles for integrating relevant findings
from GWAS and other modalities to enhance clinical translation.

Keywords: Neurodegeneration, Alzheimers disease (AD), Parkinsons disease (PD), genome-wide association
study (GWAS), single nucleotide polymorphism (SNP), pathway, network, biomarker, omics, complex disease

Introduction Alzheimers disease (AD) [1, 2], -synuclein in


Parkinsons disease (PD) [3], huntingtin protein
Several common themes have driven prevailing in Huntingtons disease (HD) [4], and transac-
notions about neurodegenerative diseases and tive response DNA-binding protein 43 (TDP-43)
their underlying etiology. Pathologically, a fre- in frontotemporal dementia (FTD) and amyo-
quent characteristic of these diseases is the trophic lateral sclerosis (ALS) [5]. The discovery
accumulation and aggregation of abnormal or of genetic mutations causing rare, early onset,
misfolded proteins, as with amyloid- (A) in familial forms of these diseases, as with the
Pathways to neurodegeneration in AD and PD

APP (amyloid precursor protein) gene in AD [6] ways and networks provide mechanistic hypoth-
and the SNCA (-synuclein) gene in PD [7], fur- eses which can guide confirmatory testing in
ther focused attention on mechanisms directly independent human study datasets, cell lines,
connected to disease pathology. However, and animal models. The ability to prioritize
most cases of AD, PD, and other neurodegen- pathways of interest may be particularly impor-
erative diseases cannot be explained by simple tant for approaches with high computational
Mendelian inheritance of genetic mutations in demand. These include whole genome sequ-
isolated disease-specific pathways. These late encing (WGS) studies, which offer enhanced
onset, sporadic forms of disease are thought power to detect rare SNPs and copy number
instead to have a complex etiology, with sus- and other structural variants [31], studies of
ceptibility influenced by lifestyle and environ- disease endophenotypes such as brain imag-
mental factors in addition to as-yet-uncharac- ing [32, 33] or cerebrospinal fluid (CSF) bio-
terized variants in numerous genes [8-12]. markers [34, 35], and studies of molecular
interactions and epistasis [36-38], among oth-
The development of methods for unbiased er approaches.
investigation of the genome initially promised
to address this knowledge gap. Although analy- We propose that pathways and networks pro-
ses of neurodegenerative diseases represent a vide an ideal framework for integrating known
substantial fraction of the more than 1500 neurodegenerative mechanisms and nominat-
published genome-wide association studies ing new targets. Here, we review the major
(GWAS) [13], several limitations of this approach pathways influencing neurodegeneration, foc-
have emerged. Most GWAS-implicated com- using on shared processes implicated by GWAS
mon single nucleotide polymorphisms (SNPs) of the most prevalent neurodegenerative disor-
display modest individual effects on disease ders, AD and PD [39]. As part of a conceptual
risk and together leave substantial heritability model (Figure 1), we discuss these pathways
unexplained [11]. For example, although up to within broader, biologically driven groups repre-
60-80% of AD risk is estimated to derive from senting intracellular mechanisms, influences
genetic factors [14], known genes including the from the local tissue environment and systemic
uniquely large effect of APOE (apolipoprotein E) circulation, and broader factors related to neu-
account for just half of this genetic variance rodevelopment and aging. We also perform net-
[15]. In addition, while major psychiatric disor- work analysis of top AD- and PD-associated
ders have displayed genetic overlap through genes to discover additional novel functional
GWAS [16], similarly robust cross-disorder SNP relationships among multiple candidate genes
associations have not been reported for neuro- and pathways.
degenerative diseases, a surprising finding
given their vast overlap of clinical and patho- Intracellular mechanisms
logical features. As a result, there has been sig-
nificant interest in the development of alterna- Apoptosis
tive perspectives and analytical strategies to
better understand the genetic architecture Although definitions vary, apoptosis is generally
underlying neurodegeneration [17, 18]. understood as a programmed cell death pro-
cess involving caspase activation, maintenance
Recently, biological pathways and networks of organelle integrity, and a lack of cell swelling
have become focal points for harnessing GWAS [40]. Aberrant regulation of apoptosis is one
data [19, 20]. Numerous studies have demon- proposed explanation for the striking loss of
strated that genes functioning in the same hippocampal and cortical neurons in AD and
pathway can collectively influence susceptibili- midbrain dopaminergic neurons in PD [41]. In
ty to neurodegenerative diseases and traits, cultured neurons, A deposition is a direct
even when constituent SNPs do not individually inducer of apoptosis [42], and early onset
exhibit significant association [21-28]. Path- AD-associated mutations in the A processing
ways occupied by top GWAS hits can also genes APP, PSEN1 (presenilin-1), and PSEN2
highlight additional genes with more modest (presenilin-2) can promote apoptosis [43-45].
effects on disease risk but which may provide The largest known genetic risk factor for late-
better targets for biomarker and drug develop- onset AD, the APOE 4 allele [46], may also be
ment [29, 30]. Further, GWAS-implicated path- related to apoptosis through interactions with

146 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

Figure 1. Conceptual model of candidate pathways contributing to neurodegeneration. Candidate pathways influ-
encing the balance of neuronal survival and degeneration are displayed within broader functional groups based
on their major site or mode of action (intracellular mechanisms, local tissue environment influences, systemic
influences, and mechanisms related to neurodevelopment and aging). The pathways and overarching functional
groups in this model are highly related and can have overlapping or interacting components which can collectively
modulate neurodegenerative processes.

A [47]. Interestingly, a recent protein interac- In human PD brains, molecular markers of


tion network analysis identified PDCD4 (pro- apoptosis are abundant in the substantia nigra
grammed cell death 4), which is up-regulated in [58], which contains mostly dopaminergic neu-
AD brains and whose expressed protein inter- rons and is the primary site of atrophy and
acts with ApoE and presenilin-2, as a potential pathology in the disease [39]. The hallmark
regulator of neuronal death in AD that may pathological feature of PD is the presence of
bridge genetic risk factors for early- and late- intracellular inclusions known as Lewy bodies,
onset disease [48]. which are mainly composed of insoluble aggre-
gates of -synuclein [3]. SNCA is associated
Several major AD GWAS-implicated genes [49- with both early- and late-onset PD [7, 59] and
51] also have putative roles in apoptosis. CLU accumulation of -synuclein in cultured dopa-
(clusterin) is proposed to interact with BCL-2 minergic neurons results in apoptosis [60].
protein family members to regulate apoptosis Other PD-related genes with potential roles in
[52, 53], and neuroimaging studies suggest apoptosis include LRRK2 (leucine-rich repeat
CLU-associated brain atrophy may be particu- kinase 2) [61, 62], MAPT (microtubule-associat-
larly evident in early stages of disease [54]. ed protein tau) [63], and PARK2 (parkinson pro-
Another BCL-2 interacting gene, HRK (harakiri) tein 2, E3 ubiquitin protein ligase) [64].
[55], was identified in a large GWAS meta-anal-
ysis of magnetic resonance imaging (MRI) hip- Development of anti-apoptotic and other neuro-
pocampal volume, a key AD endophenotype protective drugs for AD and PD is still in early
[56]. Sequence homology and functional stud- stages and may ultimately require targeting of
ies also indicate that ABCA7 (ATP-binding cas- multiple genes and sub-pathways [65, 66].
sette transporter A7) is required for efficient Such therapies will also need to address the
clearance of apoptotic cells [57]. These diverse evolving understanding of epidemiologic and
roles suggest that AD-associated genetic varia- mechanistic relationships between neurode-
tion may have pleiotropic influences on apop- generation and cancer, particularly since many
totic mechanisms. cancers are marked by down-regulation of

147 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

apoptosis in contrast to the up-regulation seen nisms for these processes are well-known to
in neurodegeneration [67, 68]. Nevertheless, yield metabolites with the potential to promote
the heavy footprint of apoptotic functions neurodegenerative oxidative stress and DNA
among known AD and PD risk loci is encourag- damage [75]. However, mitochondria also play
ing for this direction of study. important roles in other functions that can
modulate neurodegeneration, such as apopto-
Autophagy sis and endocytosis, and several key AD- and
PD-related proteins are localized to mitochon-
Autophagy is a highly regulated mechanism for dria or the interface between mitochondria and
degradation of unnecessary or dysfunctional the endoplasmic reticulum [40, 76]. The inter-
cellular components [4]. Controlled activation section of multiple pathways with mitochondri-
of autophagy may provide a strategy for clear- al function makes this organelle an important
ance of long-lived, aggregated, or dysfunctional target for strategies to combat neurodege-
proteins which contribute to neurodegenera- neration.
tion [40]. In human brains, autophagy is tran-
scriptionally down-regulated during normal In AD, the APOE 4 allele is thought to cause
aging but is up-regulated in AD, suggesting a mitochondrial dysfunction through altering the
possible attempted compensatory response to interaction capabilities of its encoded proteins
A accumulation [69]. In mice, deletion of lipid- and receptor-binding regions [77]. Genes
PD-related LRRK2 yields impaired autophagy involved in actin pathways, such as CD2AP
and augmented accumulation of -synuclein (CD2-associated protein), may directly impact
[62]. Variants in GBA (glucosidase-, acid) are mitochondrial fission, fusion, and transport
also associated with PD [59, 70], and the accu- along axons due to the dynamic actin remodel-
mulation of -synuclein in mutant GBA cell ing and stabilization required for these pro-
lines can be reversed with administration of the cesses [78]. Impairment of mitochondrial fis-
autophagy inducer rapamycin [71]. sion, fusion, and axonal transport can promote
abnormal hyperphosphorylation of MAPT
An important caveat of these findings is that (microtubule-associated protein tau), leading to
other potentially related outcome measures the accumulation of dysfunctional mitochon-
may be relevant for interpretation. For example, dria and the induction of apoptosis due to poor
increased levels of apoptotic effectors such as cellular energetics [79-81]. Components of the
caspase-3 have been detected after pharma- mitochondrial membrane are also important for
cologic inhibition of autophagy in an AD mouse normal functioning through regulation of molec-
model [72]. Whether this represents possible ular flux. For example, the AD risk genes
cross-talk between autophagy and apoptosis to TOMM40 (translocase of outer mitochondrial
respond to cellular stress or indicates that membrane 40 homolog) and TSPO (transloca-
autophagy itself is an alternate mechanism for tor protein of outer membrane, 18 kDa) are
programmed cell death remains controversial essential for mitochondrial import of proteins
[40, 73]. Genetic analyses for epistasis (gene- and cholesterol, respectively [82, 83].
gene interactions) within and between these
pathways may provide alternative strategies for In PD models, SNCA overexpression leads to
addressing these issues. Nevertheless, the the excess -synuclein associating with the
potential for complex relationships between mitochondrial membrane and inducing cyto-
autophagic and other pathways involved in pro- chrome c release and oxidative stress [84]. Two
tein stress and response suggest that in vivo other genes associated with both early- and
modulation of autophagy as a therapy for neu- late-onset PD, PARK2 and PINK1 (PTEN-
rodegeneration may require fine-tuning to induced putative kinase 1) [85], code for pro-
broader genetic and environmental profiles teins that regulate axonal transport of healthy
[69, 74]. mitochondria and autophagy of old or dysfunc-
tional mitochondria (also known as mitophagy)
Mitochondrial dysfunction [86, 87]. Another cause of early-onset PD,
PARK7 (parkinson protein 7; also known as
Mitochondria are primarily tasked with cellular DJ1), appears to work in concert with PARK2
energy production through catabolism of sug- and PINK1 as a sensor of oxidative stress and
ars, fats, and proteins. The underlying mecha- a regulator of mitophagy [84, 88].

148 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

Interestingly, two compounds used to create that modulates oxidative stress and its res-
experimental models of PD exert their toxic ponses might affect susceptibility to neurode-
effects in mitochondria. Exposure to MPTP generation.
(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)
was initially proposed as an environmental The PD-associated genes PARK2, PARK7, and
cause of PD [89]. Since this discovery, injection PINK1 may represent one molecular axis con-
of MPTP has been used to generate numerous tributing to disease risk through regulation of
cellular and animal models for PD [90]. In the oxidative stress. For example, PARK7 knock-
brain, the MAO-B (monoamine oxidase B) down is known to yield hypersensitivity to oxi-
enzyme converts MPTP into MPP+ (1-methyl- dative stress in mouse and fly brains [95], while
4-phenylpyridinium), which interferes with com- the administration of ROS scavengers and the
plex I of the mitochondrial electron transport overexpression of PINK1 and PARK2 have been
chain to fatally deplete ATP levels and cause shown to rescue the effects of PARK7 loss [96].
neuronal death [90]. The pesticide rotenone is In AD, disease-associated variants in CLU may
also used to generate PD experimental models inhibit the normal role of clusterin as a protec-
and similarly interferes with electron transport tive factor against oxidative stress have been
chain function [91]. proposed to inhibit the normal role of clusterin
as a sensor and chaperone of ROS [97].
The extensive involvement of mitochondrial Variants in GSTO2 (glutathione S-transferase
stressors and protectors in AD and PD also sug- omega-2), which codes for a subunit of glutathi-
gests that changes in mitochondrial DNA might one transferase, have also been associated
be additional markers of disease. Increased with decreased levels of glutathione which
levels of mutations in mitochondrial DNA have increase levels of ROS as well as AD suscepti-
been identified in both diseases [92]. However, bility [98]. Two other genes related to oxidative
it is not yet clear whether these mutations stress have been identified in large studies of
affect specific functions or overall mitochondri- AD-related endophenotypes, including the
al health, and it additionally remains to be dis- associations of MTFR1 (mitochondrial fission
covered if specific mitochondrial DNA variants regulator 1) with cognitive decline [99] and
are involved in early-stage disease pathogene- MSRB3 (methionine sulfoxide reductase B3)
sis or if mutations simply provide a measure of with hippocampal volume [56].
ongoing mitochondrial disturbances.
Oxidative stress and DNA damage repair path-
Oxidative DNA damage and repair ways have also been proposed as points of
overlap that might explain the decreased inci-
Oxidative stress refers to an imbalance dence of cancer in individuals with AD or PD
between levels of toxic reactive oxygen species other [67, 100-102]. It is possible that increased
(ROS) and the activity of mechanisms such as levels of oxidative stress which predispose to
the glutathione system and DNA repair path- neurodegeneration may also harm precancer-
ways to detoxify ROS to less reactive interme- ous cells which would otherwise proceed to
diates or to reverse ROS-induced cellular dam- unlimited replication. Other mechanisms, such
age [93]. Mitochondria are the major cellular as alternative splicing of genes involved in oxi-
source of ROS, and therefore dysfunction of dative metabolism and DNA repair, may also
mitochondrial components is a significant con- contribute to age- and neurodegenerative dis-
tributor to oxidative stress and its downstream ease-associated changes in the brain [103]
effects on the structures of DNA, proteins, and that oppose the development of cancer.
lipids. For example, oxidative damage to Additional study at the population and molecu-
-synuclein can change the proteins targeting lar levels will be needed to clarify these poten-
sequence to affect its cellular localization and tial mechanisms.
can promote its aggregation [84], and similar
mechanisms initiated by oxidative stress have Ubiquitin-proteasome system
been proposed to affect A as well as other
proteins implicated in age-related and neurode- The ubiquitin-proteasome system is responsi-
generative changes [94]. As a result, there is ble for targeted degradation of misfolded,
significant interest in whether genetic variation aggregated, or otherwise abnormal proteins.

149 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

The first step in activating this pathway involves Local tissue environment
ubiquitin labeling of a protein to direct it to
cylindrical proteasomes in the nucleus, endo- Cell adhesion
plasmic reticulum, and other compartments,
which recognize ubiquitin-labeled proteins and Cell adhesion involves the binding of a cell to
contain protease enzymes for protein degrada- another cell or to an extracellular surface. In
tion. In contrast to autophagy, which can also healthy brains, cell adhesion pathways are
degrade proteins in addition to whole organ- important for maintenance of synaptic contacts
elles, ubiquitin-mediated proteasomal degra- and blood-brain barrier integrity as well as effi-
dation is thought to be highly selective [104]. cient neurotransmission and intracellular sig-
naling [114]. Altered expression of cell adhe-
For AD, PD, and other neurodegenerative dis- sion genes is a consistent finding in AD and PD
eases marked by accumulation and aggrega- [115-118]. In particular, APOE 4 may promote
tion of specific abnormal proteins, ubiquitin- neurodegeneration through sequestering tar-
proteasome pathways represent natural candi- gets of RELN (reelin), a protease which signals
dates for modulating pathology. Ubiquitin- through APOER2 (apolipoprotein E, receptor 2)
positive inclusions in neurons and glial cells are and NMDA receptors to enhance synaptic
also frequently identified in AD, PD, HD, FTD, strength and plasticity [119, 120]. Depletion of
and other neurodegenerative disorders and reelin levels in key AD brain regions is thought
may be a sequelae of dysfunction in protea- to be an early event in the disease [121].
somal pathways due to variation in genes Genetic variation in RELN is also associated
including GRN (progranulin) and MAPT among with AD pathology in cognitively normal older
others [105]. Early stages of AD additionally individuals [122], reinforcing the potential role
exhibit altered expression of ubiquitin-protea- of cell adhesion as an early driver of neurode-
some pathway genes in astrocytes, which sup- generative changes.
port neuronal function and help maintain
Several studies propose relationships between
homeostasis in the brain [106]. More broadly,
the A and cell adhesion pathways, including
ubiquitin-mediated protein degradation may be
the cleavage of the key synaptic adhesion mol-
neuroprotective in modest quantities but may
ecule N-cadherin by presenilin-1 and -2 [123]
stimulate bulk autophagy or BCL-2-dependent
as well as the interaction of NCAM140 (neuro-
apoptosis at overwhelming or chronic levels [3,
nal cell adhesion molecule 140) with APP to
107, 108].
regulate neuronal outgrowth [124]. Recent
GWAS of imaging endophenotypes have also
Interestingly, activation of PD risk genes with
identified suggestive associations of cell adhe-
direct roles in ubiquitin-proteasome pathways
sion genes, including ITGA1 (integrin-1) and
may have beneficial effects in multiple neuro-
ITGA6 (integrin-6) with florbetapir positron
degenerative diseases. For example, UCHL1
emission tomography (PET) cerebral A burden
(ubiquitin thiolesterase) activation was sug-
[125] and CDH8 (cadherin 8, type 2) with hip-
gested to reverse AD-associated changes in
pocampal volume [126].
neuronal dendrite structure through signaling
of pathways related to cognition [109, 110]. In In addition, pathway analyses have discovered
addition, PARK2 overexpression is proposed to collective effects on risk among cell adhesion
promote clearance of A in AD cell culture mod- genes in AD and PD. An innovative study inte-
els [111], modulate functional levels of SYT11 grating PD case-control GWAS and genome-
(synaptotagmin) and other regulators of neuro- wide expression data for nearly 3500 individu-
transmission [112], and increase lifespan and als found enrichment of association for numer-
reduce levels of damaged proteins and mito- ous adhesion pathways, including four of the
chondria in aging fly brains [113]. These find- top five results (axon guidance, focal adhesion,
ings corroborate the potential protective effect cell adhesion molecules, adherens junction)
of ubiquitin-mediated degradation in combat- [127]. In AD, cell adhesion pathways have also
ing neurodegeneration and highlight the over- displayed enrichment of association using
lapping molecular systems involved in autopha- case-control GWAS [128] and quantitative trait
gy, mitochondrial regulation, and the ubiquitin- GWAS of episodic memory impairment [23].
proteasome system. Although cell adhesion genes and pathways are

150 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

often large and therefore carry risks of false PD-related mutations accentuates -synuclein
positive associations [19, 129], the similarity of load and toxicity [142]. The closely related gene
findings across these three methodologically AAK1 (AP-2 associated kinase 1) [143] has also
diverse studies is striking and provides further been associated through GWAS with age of PD
support of the hypothesis that adhesion mech- onset [144]. The prevalence of disease risk
anisms can contribute to neurodegeneration. genes and potential drug targets in endocytic
pathways is likely to spur continued interest in
Endocytosis the coming years.

The process known as endocytosis, where Neurotransmission


extracellular molecules are engulfed into mem-
brane-bound vesicles for internalization, is Neurotransmitters are endogenous substanc-
important for gathering nutrients, facilitating es used to relay signals across a synapse.
molecular interactions and protein degradation Although overshadowed in recent years by pro-
in a protected environment, and recycling teinopathy-related theories, initial hypotheses
ligands and receptors [130]. Several AD- and about AD and PD focused on disease-associat-
PD-associated genes have central roles in ed neurotransmitter deficits. The selective loss
endocytic pathways. For example, APOE is of brain acetylcholine-signaling neurons under-
required for microglia to degrade A following stood to be crucial for learning and memory
endocytosis, and APOE allelic variation affects drove the hypothesis that AD manifested from
the efficiency of this degradation in animal a cholinergic deficit [145]. Similarly, the loss of
models [131]. SORL1 (sortilin 1), whose asso- dopaminergic neurons from the substantia
ciations to AD were recently confirmed using nigra understood to be important for motor
exome sequencing [132] and GWAS meta-anal- functioning led to the hypothesis that dysfunc-
ysis [133], directs APP to endocytic pathways tion of dopaminergic neurotransmission was a
for recycling and is crucial in preventing the primary cause of PD [146, 147]. As a result,
sorting of APP to alternative pathways which modulation of cholinergic or dopaminergic neu-
generate A [134, 135]. In PD, LRRK2 similarly rotransmission forms the basis of several sym-
regulates the recycling and/or degradation of ptomatic therapies for AD and PD [148, 149].
-synuclein [136, 137] and is a key influence
Genetic and molecular studies support a role
on the endocytic formation of synaptic vesicles
for neurotransmitter mechanisms in neurode-
containing neurotransmitters [138].
generative disease. Pathways related to calci-
Promising strategies for therapeutic targeting um signaling, which are important for presynap-
of endocytic pathways in AD have recently tic neurotransmitter release and postsynaptic
emerged. In an AD mouse model, the retinoid signal transduction involving cyclic AMP (cAMP),
acid receptor (RXR) agonist bexarotene was protein kinase A (PKA), and cAMP response ele-
found to transcriptionally induce APOE to ment binding protein (CREB), have displayed
enhance clearance of A and the reversal of association to AD and PD [23, 127, 128, 150,
cognitive deficits [139]. The yeast homolog of 151]. The gene COMT (catechol-O-methyltrans-
PICALM (phosphatidylinositol binding clathrin ferase) encodes an enzyme that degrades
assembly protein) is also proposed to be an A dopamine and other catecholamine neu-
toxicity modifier [140]. Thus far, these new find- rotransmitters, and COMT variants have been
ings and their therapeutic implications have not associated with dopamine levels in early PD
yet been replicated or validated in other [152] and may contribute to cognitive and psy-
systems. chiatric deficits in AD through interactions with
estrogen [153, 154]. Further, in addition to its
Targeting of endocytic pathways may also be a effects on mitochondria, MPP+ gains entry to
viable approach to combat PD. The PD-asso- cells via the dopamine transporter and inhibits
ciated gene GAK (cyclin G associated kinase) synthesis of dopamine and other catechol-
[59] is a key mediator of endocytic vesicle traf- amines [155, 156]. Variation in multiple genes
ficking by regulating interactions with adaptor also contributes to elevated glutamate levels in
proteins and later driving disassembly of the multiple sclerosis (MS), which is classically
vesicle clathrin coat [141]. In cell culture, under- marked by demyelination and neuroinflamma-
expression of GAK through knockdown or tion [157].

151 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

Cholinesterase inhibitors, which attempt to eases [171-173] and other studies not finding
increase active levels of acetylcholine in the associations [174, 175]. Recent GWAS of CJD
synaptic cleft by inhibiting the enzymes that have also implicated other genes, suggesting
degrade acetylcholine, are a first line symptom- that larger pathways related to protein confor-
atic therapy for AD [158]. An initial imaging mational states and prions may be active in
study in humans identified a correlation bet- neurodegeneration [176-178]. More broadly, a
ween plasma activity of acetylcholinesterase better understanding of the forces contributing
and brain A levels [159]. Recently, a larger to protein conformation and susceptibility to
study of 555 individuals discovered a genome- aggregation and transmissibility would be a
wide significant association of variants at the crucial for unlocking novel diagnostic and ther-
BCHE (butyrylcholinesterase) locus with brain apeutic approaches for neurodegenerative dis-
A levels [160]. Butyrylcholinesterase is eases [179]. Genetic variation affecting several
enriched in senile A plaques [161] and several related pathways, including translational mach-
additional lines of evidence point to potential inery, endoplasmic reticulum function, chaper-
mechanistic connections among BCHE, APOE, one-mediated folding assistance and transpor-
and A [162-165]. Further, some have suggest- tation, and secondary, tertiary, and quaternary
ed that cholinesterase inhibitors which prefer- protein structural interactions, might represent
entially target butyrylcholinesterase may have plausible candidates for association testing to
disease-modifying effects in AD [166, 167]. clarify these mechanisms.
Future work to understand the genetic relation-
ships between the cholinergic and A pathways Systemic environment
and their impact on response to drug treat-
ments will be important to improve risk stratifi- Inflammation and immune dysfunction
cation and therapeutic targeting.
Published literature on AD and PD includes
Prions and transmissible factors robust evidence of disturbances in inflamma-
tion and immune pathways. Increased levels of
Prion protein is a membrane-associated, prote- pro-inflammatory cytokines are common find-
ase-sensitive glycoprotein that is typically ings in blood, cerebrospinal fluid (CSF), and
enriched in lipid rafts consisting of tightly post-mortem brain tissue in both diseases
packed signaling and trafficking molecules [180-183], and non-steroidal anti-inflammatory
[168]. As with other misfolded proteins, mis- drugs have been proposed to have protective
folded prion protein is normally susceptible to effects [184, 185]. Active debate has endured
proteasome-mediated and other forms of pro- on whether inflammation and immune dysregu-
tein degradation. However, accumulation of lation are contributors to neurodegeneration or
misfolded prion protein through inhibition of are instead secondary to ongoing cell death. In
protein degradation pathways has been pro- particular, a fundamental question remains
posed to lead to the formation of protease- outstanding in neurodegenerative disorders: is
resistant, aggregated, infectious (i.e., transmis- inflammation deleterious, protective, or dis-
sible) particles which can be released to neigh- ease stage-dependent?
boring cells and promote misfolded protein
states in those cells [169]. This mechanism is Studies of microglia, the resident immune sys-
thought to underlie the development of fa- tem macrophages in the brain and CSF, provide
tal degenerative transmissible spongiform some clues for resolving these issues. Post-
encephalopathies such as Creutzfeld-Jakob mortem tissue analyses as well as newer in
disease (CJD), and more controversially has vivo PET imaging methods have identified an
been proposed as a unifying factor promoting abundance of activated microglia in AD and PD
neurodegeneration across multiple neurode- brains [186]. Both A and -synuclein are
generative diseases including AD, PD, and ALS known to activate microglia, stimulating the
[170]. release of inflammatory cytokines and activa-
tion of inflammation-mediating enzymes such
So far, genetic association tests of this hypoth- as matrix metalloproteinases (MMPs) [186-
esis have been mixed, with some studies iden- 188]. Activated microglia also express NLRP3
tifying moderate associations of PRNP (prion (nucleotide-binding domain and leucine-rich
protein) variants with neurodegenerative dis- repeat family, pyrin domain containing 3), a

152 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

component of larger structures known as LRRK2 both mediates microglial-induced


inflammasomes which promote several inflam- inflammation [205, 206] and is a target of IFN-
matory processes including the maturation of (interferon gamma), suggesting an additional
IL-1 (interleukin 1, beta) [189]. In animal mod- role in the immune response to pathogens
els, IL-1 exacerbates AD and PD progression [207]. Similarly, the AD-associated gene CR1
[190, 191], and the protective effect of NLRP3 (complement component receptor 1) [49, 208-
knockout in AD mice likely reflects these under- 212] encodes a receptor which may regulate
lying mechanisms [192]. both inflammatory processes as well as classi-
cal complement pathways of innate immunity
Nevertheless, the role of microglia and their to eliminate synaptic connections [213]. The
secreted products may not be unidirectional. common involvement of inflammation and
For example, activated microglia are also immune mechanisms is not limited to AD and
unique among central nervous system cells in PD and appears to extend to ALS [214], MS
expressing CX3CR1 (chemokine receptor 1), a [27], FTD [215], and psychiatric disorders
receptor for the cell survival promoting chemo- [216].
kine known as fractalkine [193]. In PD and ALS
mouse models, CX3CR1 knockout resulted in These findings suggest that fulfilling the prom-
more extensive neuronal loss [194], suggesting ise of therapies targeting these pathways in
that augmentation of signaling through this neurodegenerative disease might be quite
microglial product may be required for therapy. complex [183, 217-219]. Appropriate modula-
In addition, microglia may have divergent roles tion of inflammatory and immune mechanisms
across the course of neurodegenerative dis- may require combinatorial regulation of multi-
eases. Whereas activation of microglia to stim- ple factors, with some being activated and oth-
ulate phagocytosis of aggregated disease- ers deactivated depending on disease stage
related proteins may be protective during early and an individuals genetic profile.
disease stages [195, 196], chronic activation
of microglia may enhance production of differ- Lipid, metabolic, and endocrine factors
ent cytokines which impair phagocytosis and
other cell survival-related processes [197]. Recent epidemiological and molecular studies
are converging to support the hypothesis that
Genetic associations in inflammation- and loss of lipid homeostasis can prominently con-
immune-related pathways may have similar tribute to neurodegeneration. Findings that ath-
implications. Variants in IL1B (interleukin 1, erosclerosis and other cardiovascular diseases
beta) and TNFA (tumor necrosis factor, alpha) are impacted by APOE 4 and can increase the
have been associated with AD and PD and may risk of AD [220] are complemented by studies
contribute to altered cytokine levels and inflam- suggesting that statin use to lower circulating
matory signaling [198, 199]. Meanwhile, cholesterol may modestly reduce the risk of AD
AD-associated variants in CLU [97] and TREM2 and PD [221, 222]. Importantly, neuronal mem-
(triggering receptor expressed on myeloid cells branes contain substantial amounts of choles-
2) [200-203] may impair the normal anti-inflam- terol and other lipids, and disturbances in lipid
matory functions of these genes. TREM2 is pre- pathways have been frequently proposed to
dominately expressed on microglia, and recent impact synaptic signaling and neuronal plastic-
expression analyses of post-mortem AD human ity and degeneration [223-226].
and mouse brain tissue identified perturba-
tions of networks regulated by the TREM2 As the major lipoprotein of the brain, ApoE
ligand TYROBP (protein tyrosine kinase binding transports key lipids and associated proteins to
protein) and enriched with genes functioning in cells for uptake via receptor-mediated endocy-
phagocytosis [204], highlighting the potential tosis [220]. The degree of lipidation in ApoE is
importance of microglia and their expressed an important factor in maintaining lipid homeo-
products in modulating neurodegenerative stasis and in mediating interactions with A
processes. which can promote its endocytic clearance,
and APOE allelic variants may affect both pro-
Other genes appear to bridge inflammation and cesses [227]. Strikingly, two other AD GWAS-
innate immune responses. For example, the implicated genes have primary roles in lipid
PD- and Crohns disease-associated gene homeostasis: CLU represents the second major

153 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

lipoprotein of the brain (also known as apolipo- sclerosis, and coronary heart disease [100,
protein J) [6, 228] and ABCA7 codes for a 248], suggesting that part of the impact of
microglia-enriched trans-membrane cholester- APOE on AD may be mediated through vascular
ol and phospholipid transporter [229, 230]. mechanisms. Pathological changes to the
Among PD-related genes, both PARK2 and blood-brain barrier have also been identified in
LRRK2 code for proteins which regulate cellular AD and PD through histological and molecular
uptake of lipid-rich structures [231-233]. analyses and may explain the proposed mod-
est protective effect of caffeine intake in these
Recently, lipidomics analyses of the complete diseases [249-251].
profile of lipids and their metabolites in tissue
samples have provided initial unbiased views Vascular smooth muscle pathways have dis-
of lipid pathway disturbances in AD and PD played genetic associations with AD imaging
[225, 234, 235]. In PD, this approach identified phenotypes [252], and the AD-associated gene
changes in lipid metabolism in human primary CR1 was also found to increase the risk of cere-
visual cortex, a region that does not exhibit sig- bral amyloid angiopathy, a leading cause of
nificant Lewy body pathology but may be impor- intracerebral hemorrhage in older individuals
tant for visual symptoms in PD [235]. These [253]. Although other vascular-related genes
large-scale findings reinforce the concept that such as VEGF (vascular endothelial growth fac-
lipid pathways are highly complex and include tor) have displayed mostly mixed results in
numerous components with the potential for association tests for AD and PD [254, 255],
local and remote impacts on inflammation, oxi- additional studies will be important to deter-
dative stress, vascular, and other pathways. As mine the effects of in situ genetic risk factors
a result, drugs targeting lipid pathways, includ- on vascular functioning and brain plasticity
ing supplementary administration of endoge- [256], relationships of vascular pathways to
nous compounds [236], would be expected to other mechanisms of neurodegeneration [257],
have pleiotropic effects in the context of neuro- and the impact of lifestyle measures such as
degenerative disease which may require modu- healthy diet and exercise on disease onset and
lation based on the functional status of other progression. Comparisons of genetic and envi-
pathways in an individual [237]. ronmental risk factors for AD and PD with those
impacting vascular dementia will also illumi-
Among metabolic disorders, a particularly inter- nate common and discordant features of their
esting relationship is apparent between diabe- underlying pathophysiology [258].
tes and AD. The presence of type 2 diabetes
doubles the risk of AD [238] and metabolic dys- Neurodevelopment and biological aging
regulation, including loss of insulin signaling
through the PI3 kinase and AKT, occurs in the Epigenetic changes
brain in early AD [239]. In addition, models of
insulin resistance or deficiency result in cere- Epigenetic factors provide mechanisms for
bral A buildup while models of A toxicity lead genetic control that do not involve modifica-
to decreased insulin signaling [240]. As a result, tions to an individuals DNA sequence [259].
diabetes and AD may share several drug tar- These heritable changes, including RNA-
gets, including insulin and IGF (insulin-like associated silencing and methylation or acety-
growth factor) stimulation [241, 242], inflam- lation of DNA or histones, can dynamically
mation [243], BCHE [160, 244, 245], and GSK3 respond to environmental stimuli [260] and
(glycogen synthase kinase 3) [246]. also appear to increase in frequency with aging
[261]. Several AD- and PD-related genes are
Vascular changes regulators or targets of epigenetic mecha-
nisms. For example, nuclear -synuclein accu-
Vascular pathology, including increases in ves- mulation inhibits histone acetylation and pro-
sel wall stiffness, changes in endothelial cell motes apoptosis in cell culture [262]. While
adhesion and metabolism, and dysfunction of PD-related SNCA mutations potentiate this
the blood-brain barrier, can promote neurode- effect, inhibition of SIRT2 (sirtuin 2) deacety-
generation through yielding chronic, low perfu- lase activity may reverse SNCA-induced toxicity
sion [247]. Presence of the APOE 4 allele is a [263]. Similarly, inhibition of HDAC2 (histone
well-known risk factor for dyslipidemia, athero- deacetylase 2) facilitates expression of genes

154 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

related to learning and memory and reverses enhancing neuronal survival and functioning to
AD symptoms in mice [264]. Epigenetic path- combat degenerative changes. For example,
ways may also impact A pathology: in mice, exogenous administration of BDNF was obser-
SIRT1 (sirtuin 1) deacetylase activity promotes ved to rescue stress hormone-induced AD-like
the alternative cleavage of APP by ADAM10 memory impairment in rats through activation
(-secretase) to decrease formation of A of several memory-related signaling pathways
[265]. In addition, nucleotide repeat expan- [275]. In addition, SNPs in the dopaminergic
sions in C9orf72 (chromosome 9 open reading neurotrophin gene CDNF (cerebral dopamine
frame 72), which are a major cause of familial neurotrophic factor) have been associated with
FTD, ALS, and related neurodegenerative disor- PD risk [276], and the highly related gene GDNF
ders [266], may exert their pathologic effects (glial cell derived neurotrophic factor) is also
via mechanisms related to RNA-mediated sile- being explored as a potential therapeutic target
ncing or unconventional translation [267, 268]. for PD [277, 278]. It should be noted that neuro-
trophins can be expressed in non-neuronal tis-
Human epigenome-wide studies have not yet sue and may have roles in promoting or inhibit-
been reported for AD or PD. In analyses of can- ing cancer at those sites [279, 280] which will
didate genes related to neuroinflammation and require further evaluation in the context of
synaptic functioning, changes in methylation of potential neurotrophin-related treatment strat-
CpG islands in the promoters of BDNF (brain- egies for neurodegenerative disease.
derived neurotrophic factor), COX2 (cyclooxy-
genase-2), CREB (cyclic AMP response element Telomeres
binding protein), and NFKB (nuclear factor
kappa B) were identified in human post-mor- Telomeres are DNA sequences at the ends of
tem AD frontal cortex [269]. Epigenome-wide chromosomes that provide protection against
studies might discover novel loci contributing to the loss of more proximal genetic material dur-
AD and PD and would be particularly informa- ing DNA replication in mitosis [281]. In germ-
tive for early stages of the disease spectrum, line and some somatic cells, the enzyme telom-
where targeted therapies would likely be most erase is responsible for maintaining telomere
effective, and to capture dynamic changes in length and structure. However, most adult
epigenetic markers longitudinally. somatic cells do not express telomerase and as
Neurotrophic factors a result gradually lose telomere length and
structure with each cycle of mitosis. While reac-
Neurotrophic factors (neurotrophins) are tivation of telomerase contributes to many
secreted growth factors that promote the types of cancer by maintaining a limitless prolif-
development, functioning, and survival of neu- erative ability for tumor cells, excessively short
rons through regulation of gene transcription. telomere length in aging cells is proposed to
Neurotrophins typically affect transcription signal for senescence and apoptosis [281,
through binding receptors at neuron terminals 282].
to stimulate second messenger signaling cas-
cades or to promote their internalization and Although shortened telomere length in periph-
direct transport along the axon to the nucleus eral white blood cells has been associated with
[270]. Diminished signaling and axonal trans- dementia and mortality in older adults, even
port of BDNF and NGF (nerve growth factor) after adjusting for APOE genotype [283], the
have been identified in post-mortem AD brain relationship between telomere length in neu-
tissue [271], and variants in BDNF have been rons and neurodegeneration is not yet clear. In
associated with CSF A levels in AD [272], age one study, neuronal telomere shortening
of onset in familial PD [273], and age-related induced microglial proliferation (microgliosis) in
changes in brain structure and cognitive func- aging mice but reduced microgliosis and A
tion in individuals without frank disease [274], pathology while improving memory and learn-
suggesting a primary role for neurotrophin sig- ing in AD mice [284]. Changes in telomere
naling in susceptibility to neurodegeneration. length have not been widely observed in periph-
eral white blood cells or in the brain in PD or
Novel treatment approaches for augmenting ALS but will likely receive continued scrutiny
neurotrophin signaling appear promising for [285-287]. In particular, several genetic influ-

155 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

ences on telomere length have been identified software algorithm generating a network with
which may provide novel candidates for study this level of interconnectedness by random
in relation to neurodegenerative disease. selection of input genes was exceedingly small
Variants in TERC (telomerase RNA component), (p = 1.14 x 10 -54). Strikingly, numerous genes in
which codes for a component of telomerase, the network exhibit co-regulation by the SP1
have been associated with telomere length in (specificity protein 1) and AP-1 (activating pro-
several human study samples [288, 289], as tein 1) transcription factors. SP1 has been pre-
have genes related to DNA and histone methyl- viously noted to regulate the expression of mul-
ation [290]. In addition, telomere pathways tiple AD-related genes [23, 295]. Elevated lev-
have exhibited enrichment of genetic associa- els of SP1 have been identified in AD human
tion to human longevity in a large cohort study brains and mouse models [296, 297] and may
[291]. These preliminary findings suggest that be induced by inflammation and oxidative
neurodegenerative diseases may be amenable stress [296, 298]. The AP-1 transcription factor
to therapies targeting mechanisms of cellular is composed of heterodimers of several pro-
and biological aging more broadly [282, 292]. teins, including those encoded by the FOS and
JUN proto-oncogenes [299]. AP-1 is an impor-
Network analysis of top AD- and PD- tant regulator of dopaminergic signaling path-
associated genes ways [300, 301] as well as numerous genes
related to autophagy and lysosomal function
To complement the pathway-driven approach,
[302]. Interestingly, animal models indicate
we performed network analysis to identify addi-
that inhibition of SP1 may be neuroprotective in
tional functional relationships between top AD-
AD [297] and inhibition of AP-1 may be neuro-
and PD-associated genes. While pathways are
protective in PD [303]. The connections among
defined by overarching goals and the mechanis-
SP1, AP-1, and AD- and PD-associated genes
tic steps involved, networks can display other
suggest that coordinate modulation of these
types of relationships which may cut across
transcription factors may be a viable strategy
multiple pathways or may indicate novel path-
for combating neurodegeneration.
ways which have not yet been characterized
[19].
This transcriptional network also includes sev-
Due to the numerous pathways implicated in eral additional genes of interest which were not
AD and PD and the pleiotropic effects of many in the initial input list. For example, EGR1 (early
key disease-associated genes, we hypothe- growth response 1) encodes a zinc-finger tran-
sized that regulatory relationships among these scription factor that is important for synaptic
genes might impact multiple pathways. To plasticity [304] and cognitive performance
explore this hypothesis, we performed tran- [305] and whose up-regulation in AD brains
scription factor network analysis using the may promote phosphorylation of tau [306]. The
MetaCore software (GeneGo, Inc.). This transcription factor encoded by HMGB1 (high-
approach incorporates knowledge from pub- mobility group protein 1) can also directly bind
lished literature to relate an input list of genes aggregated -synuclein [307], regulate phago-
to known transcription factors and proximal tar- cytosis of A [308, 309], and promote inflam-
gets such as ligand-receptor interactions. As mation when secreted by activated microglia or
input, we used the top 10 genes from the necrotic neurons [310, 311]. Interactions
AlzGene (APOE, BIN1, CLU, ABCA7, CR1, between HIV-1 TAT (transactivator of transcrip-
PICALM, MS4A6A, CD33, MS4A4E, CD2AP) tion) and genes involved in AD and PD may be
[293] and PDGene (MAPT, SNCA, GBA, LRRK2, involved in HIV-associated cognitive impair-
PM20D1, GAK, MCCC1, STK39, BST1, GPNMB) ment and A pathology [312, 313]. Other genes
[294] databases in addition to a small number of interest in this regulatory network include
of genes (APP, PSEN1, PSEN2, DJ1, HIP1R, MMP9 (matrix metalloproteinase 9) which is
PARK2, SYT11, UCHL) implicated in both involved in synaptic plasticity and A degrada-
Mendelian and sporadic forms of AD or PD. tion [314], IRF3 and IRF7 (interferon regulatory
factors 3 and 7) which regulate interferon-
A network was identified which displays rela- mediated inflammation and immune responses
tionships among 31 factors, including 19 of the [315-318], and LRP1 (low density lipoprotein
28 input genes (Figure 2). The probability of the receptor-related protein 1) which may affect

156 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

Figure 2. Regulatory network centered on the SP1 and AP-1 transcription factors is enriched with top AD and PD
genes. Meta-analytic genetic association data from public databases and supplementary manual curation was
used to generate a list of 13 AD genes and 15 PD genes. Network analysis was performed using MetaCore (GeneGo,
Inc.) to relate these input genes to known transcription factors and proximal targets based on published findings.
A highly interconnected network including 9 AD genes (labeled in blue), 10 PD genes (labeled in red), and 13 ad-
ditional genes (labeled in black) was identified. Many of the input AD and PD genes exhibit co-regulation by the SP1
and AP-1 transcription factors. Other genes of interest were also related to input AD and PD genes and represent a
variety of candidate pathways in neurodegeneration.

several neurodegeneration pathways including Conclusions and future prospects


lipid metabolism, A endocytosis, and inflam-
mation [319-322]. Through a detailed review of GWAS, we identi-
fied numerous pathways common to AD and PD
It should be noted that this analysis is not com- which nominate promising new targets for fur-
prehensive or unbiased. Complementary strat- ther study as well as biomarker and drug devel-
egies, including the use of alternative criteria opment. These findings build on established
for selection or statistical weighting of input notions of complex disease etiology, with mul-
genes as well as other schema for defining net- tiple processes presumed to influence neuro-
work connections, might highlight different degeneration and clinical outcomes in AD, PD,
relationships. Nevertheless, this regulatory net- and related disorders. They also advance the
work generates hypotheses for further investi- understanding of mechanisms likely to be cru-
gation and reflects, at the transcriptional level, cial in maintaining brain structure and function
many of the same pathways implicated by during normal aging, in contrast to changes
genetic studies of AD and PD. More broadly, a seen in AD and PD. These insights suggest that
better understanding of altered transcriptional collaborative efforts to leverage genetic and
regulation patterns through whole genome biomarker data in AD, PD, and related disorders
expression arrays and whole transcriptome would likely provide major stimuli for develop-
sequencing (RNA-seq) would augment GWAS ing unified treatment approaches to combat
findings in neurodegenerative disease and neurodegeneration.
would provide functional information to con-
nect genetic associations with their biochemi- For neurodegenerative and other complex dis-
cal outcomes. eases, accounting for the substantial heritabil-

157 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

Figure 3. Biological pathways and networks: a hub for convergent omics. Numerous large scale omics approaches
are being used to study complex neurodegenerative diseases and endophenotypes in human tissue and animal and
other model systems. Unlike individual genes and other isolated molecules, which may not be present in all model
systems and may have differential sensitivity for detection with various study designs, pathways and networks are
well-conserved and can be evaluated for convergence across diverse methodological approaches. Integration of
findings to identify pathways and networks with consistent relationships to disease is likely to enhance the develop-
ment of diagnostic biomarkers and treatment and prevention strategies.

ity that is not explained by individual GWAS- ing multiple pathways. One possibility is that a
implicated variants is an ongoing challenge [11, combination of clinical biomarkers such as
323]. The pathways and networks identified genotype, blood and CSF analyte, brain imag-
here provide several routes for addressing this ing, cognitive assessment, and medical history
limitation. For example, pathway analysis of data might be required in order to detect the
GWAS data relies on high quality pathway defi- effects of multiple pathways. Since the func-
nitions, and for some biological realms, expert tions of many disease pathways may be dis-
and updated manual curation of pathways can ease stage-specific, high blood levels of a par-
be superior to public databases and enhance ticular cytokine might have different implica-
statistical power for these analyses [19, 20]. tions for risk stratification depending on geno-
Pathways implicated by common SNPs from type, brain structure, and other measures.
GWAS also provide a knowledge-driven frame- Similarly, therapeutic and preventative strate-
work for targeting initial studies with WGS data, gies for neurodegenerative disease may benefit
which is better suited for detection of rare SNPs from drug combinations based on the cocktail
and copy number and other structural variants approaches used for HIV infection and some
but is computationally demanding to store and cancers. It is possible that efficacy, and there-
analyze [31]. Finally, interactions among known fore the choice of particular drugs to include in
variants and lifestyle, environmental, and epi- the cocktail, may depend on an individuals pro-
genetic factors may impact susceptibility [324], file of biomarkers and key genetic variants
and pathways and networks understood to be some of which may be protective and others
involved in pathogenesis may be more likely to deleterious in targeted pathways. The devel-
contain these interactions [36, 38]. opment of advanced statistical models for
analysis of large, multimodal datasets will help
Diagnosis and treatment strategies for neuro- to explore these potentially new paradigms that
degenerative diseases may also need to evolve may facilitate a personalized medicine for neu-
to reflect a complex genetic architecture involv- rodegenerative diseases.

158 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

More broadly, pathways and networks can Address correspondence to: Dr. Andrew J Saykin, IU
serve as vehicles for integrating findings from Health Neuroscience Center, Suite 4100, Indiana
diverse studies of neurodegeneration. There University School of Medicine, 355 West 16th Street,
are many active strategies for large scale omics Indianapolis, IN 46202, USA. Tel: 317-963-7501;
analysis of neurodegenerative disease (Figure Fax: 317-963-7547; E-mail: asaykin@iupui.edu
3), and findings that converge across these
multiple study designs can provide confirmato- References
ry evidence that is crucial for efficient clinical [1] Hardy JA and Higgins GA. Alzheimers disease:
translation. Isolated genes and molecules can the amyloid cascade hypothesis. Science
be challenging to evaluate for convergence 1992; 256: 184-185.
since they may not be represented in all data [2] Karran E, Mercken M and De Strooper B. The
modalities or experimental model systems. In amyloid cascade hypothesis for Alzheimers
contrast, pathways and networks can incorpo- disease: an appraisal for the development of
rate data from multiple biological levels (e.g., therapeutics. Nat Rev Drug Discov 2011; 10:
genes, transcripts, proteins, and metabolites, 698-712.
among others) and may be more likely to be [3] Taylor JP, Hardy J and Fischbeck KH. Toxic pro-
teins in neurodegenerative disease. Science
evolutionarily conserved [325]. For example,
2002; 296: 1991-1995.
recent pathway-based studies integrating [4] Krainc D. Clearance of mutant proteins as a
GWAS and gene expression data have demon- therapeutic target in neurodegenerative dis-
strated enhanced power, reproducibility, and eases. Arch Neurol 2010; 67: 388-392.
connections of top findings to hypothesized dis- [5] Rademakers R, Neumann M and Mackenzie
ease processes [127, 326, 327]. The utility of IR. Advances in understanding the molecular
these studies will increase as present limita- basis of frontotemporal dementia. Nat Rev
tions of pathway-based approaches are Neurol 2012; 8: 423-434.
addressed, including how to incorporate asso- [6] Sleegers K, Lambert JC, Bertram L, Cruts M,
ciations from intergenic regions and from genes Amouyel P and Van Broeckhoven C. The pur-
suit of susceptibility genes for Alzheimers dis-
without known functions. A pathway-based
ease: progress and prospects. Trends Genet
framework also emphasizes that the discovery 2010; 26: 84-93.
of a strongly associated genetic variant repre- [7] Hardy J. Genetic analysis of pathways to Par-
sents a foundation to study functionally related kinson disease. Neuron 2010; 68: 201-206.
genes, since other components in the pathway [8] Gandhi S and Wood NW. Genome-wide asso-
may yield better targets for biomarker and drug ciation studies: the key to unlocking neurode-
development [29, 30, 328]. These advantages generation? Nat Neurosci 2010; 13: 789-794.
will be vital in harnessing the wealth of existing [9] Bertram L and Tanzi RE. The genetic epidemi-
data on neurodegenerative disease to develop ology of neurodegenerative disease. J Clin In-
an integrated understanding of its mechanisms vest 2005; 115: 1449-1457.
[10] McCarthy MI, Abecasis GR, Cardon LR, Gold-
and formulate optimal clinical guidelines.
stein DB, Little J, Ioannidis JP and Hirschhorn
JN. Genome-wide association studies for com-
Acknowledgements plex traits: consensus, uncertainty and chal-
lenges. Nat Rev Genet 2008; 9: 356-369.
This work was supported by the Indiana Clinical [11] Chee Seng K, En Yun L, Yudi P and Kee Seng C.
and Translational Sciences Institute (CTSI) The pursuit of genome-wide association stud-
National Institutes of Health (NIH) grants U54 ies: where are we now? J Hum Genet 2010;
RR025761, RR027710-01, and RR020128, 55: 195-206.
the Indiana University Medical Scientist Training [12] Noorbakhsh F, Overall CM and Power C. Deci-
Program (MSTP) NIH grant GM077229-02, the phering complex mechanisms in neurodegen-
National Science Foundation (NSF) grant IIS- erative diseases: the advent of systems biolo-
gy. Trends Neurosci 2009; 32: 88-100.
11173365, as well as NIH grants U01
[13] Hindorff LA, Junkins HA, Hall PN, Mehta JP and
AG024904, RC2 AG036535, R01 AG19771,
Manolio TA. A Catalog of Published Genome-
P30 AG10133, and R01 LM011360. Wide Association Studies. National Human Ge-
nome Research Institute, http://www.genome.
Disclosure of conflict of interest gov/gwastudies 2011.
[14] Gatz M, Reynolds CA, Fratiglioni L, Johansson
The authors declare no conflict of interest. B, Mortimer JA, Berg S, Fiske A and Pedersen

159 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

NL. Role of genes and environments for ex- phrenia and bipolar disorder susceptibility. Mol
plaining Alzheimer disease. Arch Gen Psychia- Psychiatry 2011; 16: 286-292.
try 2006; 63: 168-174. [25] ODushlaine C, Kenny E, Heron EA, Segurado
[15] Kamboh MI, Demirci FY, Wang X, Minster RL, R, Gill M, Morris DW and Corvin A. The SNP ra-
Carrasquillo MM, Pankratz VS, Younkin SG, tio test: pathway analysis of genome-wide as-
Saykin AJ, Jun G, Baldwin C, Logue MW, Buros sociation datasets. Bioinformatics 2009; 25:
J, Farrer L, Pericak-Vance MA, Haines JL, Sweet 2762-2763.
RA, Ganguli M, Feingold E, DeKosky ST, Lopez [26] Baranzini SE, Galwey NW, Wang J, Khankha-
OL and Barmada MM. Genome-wide associa- nian P, Lindberg R, Pelletier D, Wu W, Uitde-
tion study of Alzheimers disease. Transl Psy- haag BM, Kappos L, Polman CH, Matthews
chiatry 2012; 2: e117. PM, Hauser SL, Gibson RA, Oksenberg JR and
[16] Cross-Disorder Group of the Psychiatric Ge- Barnes MR. Pathway and network-based anal-
nomics Consortium; Smoller JW, Craddock N, ysis of genome-wide association studies in
Kendler K, Lee PH, Neale BM, Nurnberger JI, multiple sclerosis. Hum Mol Genet 2009; 18:
Ripke S, Santangelo S, Sullivan PF. Identifica- 2078-2090.
tion of risk loci with shared effects on five ma- [27] Sawcer S, Hellenthal G, Pirinen M, Spencer C,
jor psychiatric disorders: a genome-wide analy- Patsopoulos N, Moutsianas L, Dilthey A, Su Z,
sis. Lancet 2013 Apr 20; 381: 1371-9. Freeman C, Hunt S, Edkins S, Gray E, Booth D,
[17] Simon-Sanchez J and Singleton A. Genome- Potter SC, Goris A, Band G, Oturai AB, Strange
wide association studies in neurological disor- A, Saarela J, Bellenguez C, Fontaine B, Gillman
ders. Lancet Neurol 2008; 7: 1067-1072. M, Hemmer B, Gwilliam R, Zipp F, Jayakumar A,
[18] Cantor RM, Lange K and Sinsheimer JS. Priori- Martin R, Leslie S, Hawkins S, Giannoulatou E,
tizing GWAS results: A review of statistical Dalfonso S, Blackburn H, Boneschi FM, Liddle
methods and recommendations for their appli- J, Harbo HF, Perez ML, Spurkland A, Waller MJ,
cation. Am J Hum Genet 2010; 86: 6-22. Mycko MP, Ricketts M, Comabella M, Ham-
[19] Ramanan VK, Shen L, Moore JH and Saykin AJ. mond N, Kockum I, McCann OT, Ban M, Whit-
Pathway analysis of genomic data: concepts, taker P, Kemppinen A, Weston P, Hawkins C,
methods, and prospects for future develop- Widaa S, Zajicek J, Dronov S, Robertson N,
ment. Trends Genet 2012; 28: 323-332. Bumpstead SJ, Barcellos LF, Ravindrarajah R,
[20] Wang K, Li M and Hakonarson H. Analysing bi- Abraham R, Alfredsson L, Ardlie K, Aubin C,
ological pathways in genome-wide association Baker A, Baker K, Baranzini SE, Bergamaschi
studies. Nat Rev Genet 2010; 11: 843-854. L, Bergamaschi R, Bernstein A, Berthele A,
[21] Lambert JC, Grenier-Boley B, Chouraki V, Heath Boggild M, Bradfield JP, Brassat D, Broadley
S, Zelenika D, Fievet N, Hannequin D, Pasquier SA, Buck D, Butzkueven H, Capra R, Carroll
F, Hanon O, Brice A, Epelbaum J, Berr C, Dar- WM, Cavalla P, Celius EG, Cepok S, Chiavacci
tigues JF, Tzourio C, Campion D, Lathrop M and R, Clerget-Darpoux F, Clysters K, Comi G, Coss-
Amouyel P. Implication of the immune system burn M, Cournu-Rebeix I, Cox MB, Cozen W,
in Alzheimers disease: evidence from genome- Cree BA, Cross AH, Cusi D, Daly MJ, Davis E, de
wide pathway analysis. J Alzheimers Dis 2010; Bakker PI, Debouverie M, Dhooghe MB, Dixon
20: 1107-1118. K, Dobosi R, Dubois B, Ellinghaus D, Elovaara
[22] Hong MG, Alexeyenko A, Lambert JC, Amouyel I, Esposito F, Fontenille C, Foote S, Franke A,
P and Prince JA. Genome-wide pathway analy- Galimberti D, Ghezzi A, Glessner J, Gomez R,
sis implicates intracellular transmembrane Gout O, Graham C, Grant SF, Guerini FR, Ha-
protein transport in Alzheimer disease. J Hum konarson H, Hall P, Hamsten A, Hartung HP,
Genet 2010; 55: 707-709. Heard RN, Heath S, Hobart J, Hoshi M, Infante-
[23] Ramanan VK, Kim S, Holohan K, Shen L, Nho Duarte C, Ingram G, Ingram W, Islam T, Jagodic
K, Risacher SL, Foroud TM, Mukherjee S, M, Kabesch M, Kermode AG, Kilpatrick TJ, Kim
Crane PK, Aisen PS, Petersen RC, Weiner MW, C, Klopp N, Koivisto K, Larsson M, Lathrop M,
Saykin AJ; Alzheimers Disease Neuroimaging Lechner-Scott JS, Leone MA, Lepp V, Liljedahl
Initiative (ADNI). Genome-wide pathway analy- U, Bomfim IL, Lincoln RR, Link J, Liu J, Lorent-
sis of memory impairment in the Alzheimers zen AR, Lupoli S, Macciardi F, Mack T, Marriott
Disease Neuroimaging Initiative (ADNI) cohort M, Martinelli V, Mason D, McCauley JL, Mentch
implicates gene candidates, canonical path- F, Mero IL, Mihalova T, Montalban X, Motters-
ways, and networks. Brain Imaging Behav head J, Myhr KM, Naldi P, Ollier W, Page A,
2012; 6: 634-648. Palotie A, Pelletier J, Piccio L, Pickersgill T,
[24] ODushlaine C, Kenny E, Heron E, Donohoe G, Piehl F, Pobywajlo S, Quach HL, Ramsay PP, Re-
Gill M, Morris D and Corvin A. Molecular path- unanen M, Reynolds R, Rioux JD, Rodegher M,
ways involved in neuronal cell adhesion and Roesner S, Rubio JP, Rckert IM, Salvetti M,
membrane scaffolding contribute to schizo- Salvi E, Santaniello A, Schaefer CA, Schreiber

160 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

S, Schulze C, Scott RJ, Sellebjerg F, Selmaj KW, [36] McKinney BA and Pajewski NM. Six Degrees of
Sexton D, Shen L, Simms-Acuna B, Skidmore Epistasis: Statistical Network Models for
S, Sleiman PM, Smestad C, Srensen PS, Sn- GWAS. Front Genet 2011; 2: 109.
dergaard HB, Stankovich J, Strange RC, Sulo- [37] Vidal M, Cusick ME and Barabasi AL. Interac-
nen AM, Sundqvist E, Syvnen AC, Taddeo F, tome networks and human disease. Cell 2011;
Taylor B, Blackwell JM, Tienari P, Bramon E, 144: 986-998.
Tourbah A, Brown MA, Tronczynska E, Casas JP, [38] Schadt EE. Molecular networks as sensors and
Tubridy N, Corvin A, Vickery J, Jankowski J, Vil- drivers of common human diseases. Nature
loslada P, Markus HS, Wang K, Mathew CG, 2009; 461: 218-223.
Wason J, Palmer CN, Wichmann HE, Plomin R, [39] Nussbaum RL and Ellis CE. Alzheimers Dis-
Willoughby E, Rautanen A, Winkelmann J, Wit- ease and Parkinsons Disease. N Engl J Med
tig M, Trembath RC, Yaouanq J, Viswanathan 2003; 348: 1356-1364.
AC, Zhang H, Wood NW, Zuvich R, Deloukas P, [40] Bredesen DE, Rao RV and Mehlen P. Cell death
Langford C, Duncanson A, Oksenberg JR, Peri- in the nervous system. Nature 2006; 443:
cak-Vance MA, Haines JL, Olsson T, Hillert J, 796-802.
Ivinson AJ, De Jager PL, Peltonen L, Stewart GJ, [41] Mattson MP. Apoptosis in neurodegenerative
Hafler DA, Hauser SL, McVean G, Donnelly P, disorders. Nat Rev Mol Cell Biol 2000; 1: 120-
Compston A. Genetic risk and a primary role 129.
for cell-mediated immune mechanisms in mul- [42] Loo DT, Copani A, Pike CJ, Whittemore ER,
tiple sclerosis. Nature 2011; 476: 214-219. Walencewicz AJ and Cotman CW. Apoptosis Is
[28] Psychiatric GWAS Consortium Bipolar Disorder Induced by Beta-Amyloid in Cultured Central-
Working Group. Large-scale genome-wide as- Nervous-System Neurons. Proc Natl Acad Sci U
sociation analysis of bipolar disorder identifies S A 1993; 90: 7951-7955.
a new susceptibility locus near ODZ4. Nat Gen- [43] Wolozin B, Iwasaki K, Vito P, Ganjei JK, Lacana
et 2011; 43: 977-983. E, Sunderland T, Zhao B, Kusiak JW, Wasco W
[29] Hirschhorn JN. Genomewide Association Stud- and DAdamio L. Participation of presenilin 2
ies Illuminating Biologic Pathways. N Engl J in apoptosis: enhanced basal activity con-
Med 2009; 360: 1699-1701. ferred by an Alzheimer mutation. Science
[30] Penrod NM, Cowper-Sal-lari R and Moore JH. 1996; 274: 1710-1713.
Systems genetics for drug target discovery. [44] Weidemann A, Paliga K, Durrwang U, Reinhard
Trends Pharmacol Sci 2011; 32: 623-630. FBM, Schuckert O, Evin G and Masters CL. Pro-
[31] Bras J, Guerreiro R and Hardy J. Use of next- teolytic processing of the Alzheimers disease
generation sequencing and other whole-ge- amyloid precursor protein within its cytoplas-
nome strategies to dissect neurological dis- mic domain by caspase-like proteases. J Biol
ease. Nat Rev Neurosci 2012; 13: 453-464. Chem 1999; 274: 5823-5829.
[32] Braskie MN, Ringman JM and Thompson PM. [45] Guo Q, Sebastian L, Sopher BL, Miller MW,
Neuroimaging measures as endophenotypes Glazner GW, Ware CB, Martin GM and Mattson
in Alzheimers disease. Int J Alzheimers Dis MP. Neurotrophic factors [activity-dependent
2011; 2011: 490140. neurotrophic factor (ADNF) and basic fibro-
[33] Kendler KS and Neale MC. Endophenotype: a blast growth factor (bFGF)] interrupt excitotoxic
conceptual analysis. Mol Psychiatry 2010; 15: neurodegenerative cascades promoted by a
789-797. PS1 mutation. Proc Natl Acad Sci U S A 1999;
[34] Cruchaga C, Kauwe JS, Nowotny P, Bales K, 96: 4125-4130.
Pickering EH, Mayo K, Bertelsen S, Hinrichs A; [46] Corder EH, Saunders AM, Strittmatter WJ, Sch-
Alzheimers Disease Neuroimaging Initiative; mechel DE, Gaskell PC, Small GW, Roses AD,
Fagan AM, Holtzman DM, Morris JC, Goate AM. Haines JL and Pericak-Vance MA. Gene dose of
Cerebrospinal fluid APOE levels: an endophe- apolipoprotein E type 4 allele and the risk of
notype for genetic studies for Alzheimers dis- Alzheimers disease in late onset families. Sci-
ease. Hum Mol Genet 2012 Oct 15; 21: 4558- ence 1993; 261: 921-923.
71. [47] Koffie RM, Hashimoto T, Tai HC, Kay KR, Serra-
[35] Jack CR Jr, Vemuri P, Wiste HJ, Weigand SD, no-Pozo A, Joyner D, Hou S, Kopeikina KJ,
Lesnick TG, Lowe V, Kantarci K, Bernstein MA, Frosch MP, Lee VM, Holtzman DM, Hyman BT
Senjem ML, Gunter JL, Boeve BF, Trojanowski and Spires-Jones TL. Apolipoprotein E4 effects
JQ, Shaw LM, Aisen PS, Weiner MW, Petersen in Alzheimers disease are mediated by synap-
RC, Knopman DS; Alzheimers Disease Neuro- totoxic oligomeric amyloid-beta. Brain 2012;
imaging Initiative. Shapes of the Trajectories of 135: 2155-2168.
5 Major Biomarkers of Alzheimer Disease. [48] Soler-Lopez M, Zanzoni A, Lluis R, Stelzl U and
Arch Neurol 2012; 69: 856-867. Aloy P. Interactome mapping suggests new

161 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

mechanistic details underlying Alzheimers dis- Gallacher J, Hull M, Rujescu D, Giegling I,


ease. Genome Res 2011; 21: 364-376. Goate AM, Kauwe JSK, Cruchaga C, Nowotny P,
[49] Lambert JC, Heath S, Even G, Campion D, Morris JC, Mayo K, Sleegers K, Bettens K,
Sleegers K, Hiltunen M, Combarros O, Zelenika Engelborghs S, De Deyn PP, Van Broeckhoven
D, Bullido MJ, Tavernier B, Letenneur L, Bet- C, Livingston G, Bass NJ, Gurling H, McQuillin
tens K, Berr C, Pasquier F, Fievet N, Barberger- A, Gwilliam R, Deloukas P, Al-Chalabi A, Shaw
Gateau P, Engelborghs S, De Deyn P, Mateo I, CE, Tsolaki M, Singleton AB, Guerreiro R,
Franck A, Helisalmi S, Porcellini E, Hanon O, de Muhleisen TW, Nothen MM, Moebus S, Jockel
Pancorbo MM, Lendon C, Dufouil C, Jaillard C, KH, Klopp N, Wichmann HE, Pankratz VS, San-
Leveillard T, Alvarez V, Bosco P, Mancuso M, do SB, Aasly JO, Barcikowska M, Wszolek ZK,
Panza F, Nacmias B, Bossu P, Piccardi P, An- Dickson DW, Graff-Radford NR, Petersen RC,
noni G, Seripa D, Galimberti D, Hannequin D, van Duijn CM, Breteler MMB, Ikram MA, DeSte-
Licastro F, Soininen H, Ritchie K, Blanche H, fano AL, Fitzpatrick AL, Lopez O, Launer LJ, Se-
Dartigues JF, Tzourio C, Gut I, Van Broeckhoven shadri S, Berr C, Campion D, Epelbaum J, Dar-
C, Alperovitch A, Lathrop M and Amouyel P. tigues JF, Tzourio C, Alperovitch A, Lathrop M,
Genome-wide association study identifies vari- Feulner TM, Friedrich P, Riehle C, Krawczak M,
ants at CLU and CR1 associated with Alzheim- Schreiber S, Mayhaus M, Nicolhaus S, Wagen-
ers disease. Nat Genet 2009; 41: 1094-1099. pfeil S, Steinberg S, Stefansson H, Stefansson
[50] Harold D, Abraham R, Hollingworth P, Sims R, K, Snaedal J, Bjornsson S, Jonsson PV, Cho-
Gerrish A, Hamshere ML, Pahwa JS, Moskvina uraki V, Genier-Boley B, Hiltunen M, Soininen
V, Dowzell K, Williams A, Jones N, Thomas C, H, Combarros O, Zelenika D, Delepine M, Bul-
Stretton A, Morgan AR, Lovestone S, Powell J, lido MJ, Pasquier F, Mateo I, Frank-Garcia A,
Proitsi P, Lupton MK, Brayne C, Rubinsztein Porcellini E, Hanon O, Coto E, Alvarez V, Bosco
DC, Gill M, Lawlor B, Lynch A, Morgan K, Brown P, Siciliano G, Mancuso M, Panza F, Solfrizzi V,
KS, Passmore PA, Craig D, McGuinness B, Nacmias B, Sorbi S, Bossu P, Piccardi P, Arosio
Todd S, Holmes C, Mann D, Smith AD, Love S, B, Annoni G, Seripa D, Pilotto A, Scarpini E,
Kehoe PG, Hardy J, Mead S, Fox N, Rossor M, Galimberti D, Brice A, Hannequin D, Licastro F,
Collinge J, Maier W, Jessen F, Schurmann B, Jones L, Holmans PA, Jonsson T, Riemen-
van den Bussche H, Heuser I, Kornhuber J, schneider M, Morgan K, Younkin SG, Owen MJ,
Wiltfang J, Dichgans M, Frolich L, Hampel H, ODonovan M, Amouyel P and Williams J. Com-
Hull M, Rujescu D, Goate AM, Kauwe JSK, Cru- mon variants at ABCA7, MS4A6A/MS4A4E,
chaga C, Nowotny P, Morris JC, Mayo K, EPHA1, CD33 and CD2AP are associated with
Sleegers K, Bettens K, Engelborghs S, De Deyn Alzheimers disease. Nat Genet 2011 May; 43:
PP, Van Broeckhoven C, Livingston G, Bass NJ, 429-35.
Gurling H, McQuillin A, Gwilliam R, Deloukas P, [52] Yu JT and Tan L. The Role of Clusterin in Al-
Al-Chalabi A, Shaw CE, Tsolaki M, Singleton AB, zheimers Disease: Pathways, Pathogenesis,
Guerreiro R, Muhleisen TW, Nothen MM, Moe- and Therapy. Mol Neurobiol 2012; 45: 314-
bus S, Jockel KH, Klopp N, Wichmann HE, Car- 326.
rasquillo MM, Pankratz VS, Younkin SG, Hol- [53] Youle RJ and Strasser A. The BCL-2 protein
mans PA, ODonovan M, Owen MJ and Williams family: opposing activities that mediate cell
J. Genome-wide association study identifies death. Nat Rev Mol Cell Biol 2008; 9: 47-59.
variants at CLU and PICALM associated with [54] Thambisetty M, An Y, Kinsey A, Koka D, Saleem
Alzheimers disease. Nat Genet 2009; 41: M, Guntert A, Kraut M, Ferrucci L, Davatzikos
1088-1093. C, Lovestone S and Resnick SM. Plasma clus-
[51] Hollingworth P, Harold D, Sims R, Gerrish A, terin concentration is associated with longitu-
Lambert JC, Carrasquillo MM, Abraham R, dinal brain atrophy in mild cognitive impair-
Hamshere ML, Pahwa JS, Moskvina V, Dowzell ment. Neuroimage 2012; 59: 212-217.
K, Jones N, Stretton A, Thomas C, Richards A, [55] Sborgi L, Barrera-Vilarmau S, Obregon P and
Ivanov D, Widdowson C, Chapman J, Lovestone de Alba E. Characterization of a novel interac-
S, Powell J, Proitsi P, Lupton MK, Brayne C, Ru- tion between Bcl-2 members Diva and Haraki-
binsztein DC, Gill M, Lawlor B, Lynch A, Brown ri. PLoS One 2010; 5: e15575.
KS, Passmore PA, Craig D, McGuinness B, [56] Bis JC, DeCarli C, Smith AV, van der Lijn F, Criv-
Todd S, Holmes C, Mann D, Smith AD, Beau- ello F, Fornage M, Debette S, Shulman JM,
mont H, Warden D, Wilcock G, Love S, Kehoe Schmidt H, Srikanth V, Schuur M, Yu L, Choi
PG, Hooper NM, Vardy ER, Hardy J, Mead S, SH, Sigurdsson S, Verhaaren BFJ, DeStefano
Fox NC, Rossor M, Collinge J, Maier W, Jessen AL, Lambert JC, Jack CR, Struchalin M,
F, Ruther E, Schurmann B, Heun R, Kolsch H, Stankovich J, Ibrahim-Verbaas CA, Fleischman
van den Bussche H, Heuser I, Kornhuber J, D, Zijdenbos A, den Heijer T, Mazoyer B, Coker
Wiltfang J, Dichgans M, Frolich L, Hampel H, LH, Enzinger C, Danoy P, Amin N, Arfanakis K,

162 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

van Buchem MA, de Bruijn RF, Beiser A, Du- eton in vitro. J Biol Chem 2011; 286: 5055-
fouil C, Huang J, Cavalieri M, Thomson R, Nies- 5068.
sen WJ, Chibnik LB, Gislason GK, Hofman A, [64] Johnson BN, Berger AK, Cortese GP and Lavoie
Pikula A, Amouyel P, Freeman KB, Phan TG, MJ. The ubiquitin E3 ligase parkin regulates
Oostra BA, Stein JL, Medland SE, Vasquez AA, the proapoptotic function of Bax. Proc Natl
Hibar DP, Wright MJ, Franke B, Martin NG, Acad Sci U S A 2012; 109: 6283-6288.
Thompson PM, Nalls MA, Uitterlinden AG, Au R, [65] Sureda FX, Junyent F, Verdaguer E, Auladell C,
Elbaz A, Beare RJ, van Swieten JC, Lopez OL, Pelegri C, Vilaplana J, Folch J, Canudas AM, Za-
Harris TB, Chouraki V, Breteler MMB, De Jager rate CB, Palles M and Camins A. Antiapoptotic
PL, Becker JT, Vernooij MW, Knopman D, Faze- drugs: a therapautic strategy for the preven-
kas F, Wolf PA, van der Lugt A, Gudnason V, tion of neurodegenerative diseases. Curr
Longstreth WT, Brown MA, Bennett DA, van Pharm Des 2011; 17: 230-245.
Duijn CM, Mosley TH, Schmidt R, Tzourio C, [66] Rohn TT. The role of caspases in Alzheimers
Launer LJ, Ikram MA and Seshadri S. Common disease; potential novel therapeutic opportuni-
variants at 12q14 and 12q24 are associated ties. Apoptosis 2010; 15: 1403-1409.
with hippocampal volume. Nature Genetics [67] Tabares-Seisdedos R, Dumont N, Baudot A,
2012; 44: 545-551. Valderas JM, Climent J, Valencia A, Crespo-Fa-
[57] Jehle AW, Gardai SJ, Li S, Linsel-Nitschke P, corro B, Vieta E, Gomez-Beneyto M, Martinez S
Morimoto K, Janssen WJ, Vandivier RW, Wang and Rubenstein JL. No paradox, no progress:
N, Greenberg S, Dale BM, Qin C, Henson PM inverse cancer comorbidity in people with oth-
and Tall AR. ATP-binding cassette transporter er complex diseases. Lancet Oncol 2011; 12:
A7 enhances phagocytosis of apoptotic cells 604-608.
and associated ERK signaling in macrophages. [68] Holohan KN, Lahiri DK, Schneider BP, Foroud T
J Cell Biol 2006; 174: 547-556. and Saykin AJ. Functional microRNAs in Al-
[58] Jenner P and Olanow CW. Understanding cell zheimers disease and cancer: differential
death in Parkinsons disease. Ann Neurol regulation of common mechanisms and path-
1998; 44: S72-84. way. Front Genet 2012; 3: 323.
[59] International Parkinson Disease Genomics [69] Lipinski MM, Zheng B, Lu T, Yan Z, Py BF, Ng A,
Consortium; Nalls MA, Plagnol V, Hernandez Xavier RJ, Li C, Yankner BA, Scherzer CR and
DG, Sharma M, Sheerin UM, Saad M, Simon- Yuan J. Genome-wide analysis reveals mecha-
Sanchez J, Schulte C, Lesage S, Sveinbjorns- nisms modulating autophagy in normal brain
dottir S, Stefansson K, Martinez M, Hardy J, aging and in Alzheimers disease. Proc Natl
Heutink P, Brice A, Gasser T, Singleton AB and Acad Sci U S A 2010; 107: 14164-14169.
Wood NW. Imputation of sequence variants for [70] Sidransky E and Lopez G. The link between the
identification of genetic risks for Parkinsons GBA gene and parkinsonism. Lancet Neurol
disease: a meta-analysis of genome-wide as- 2012; 11: 986-998.
sociation studies. Lancet 2011; 377: 641-649. [71] Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlin-
[60] Xu J, Kao SY, Lee FJ, Song W, Jin LW and son JJ, Kolodziej P, Kahn I, Saftig P, Woulfe J,
Yankner BA. Dopamine-dependent neurotoxic- Rochet JC, Glicksman MA, Cheng SH, Grabows-
ity of alpha-synuclein: a mechanism for selec- ki GA, Shihabuddin LS and Schlossmacher
tive neurodegeneration in Parkinson disease. MG. Acid -glucosidase mutants linked to gau-
Nat Med 2002; 8: 600-606. cher disease, parkinson disease, and lewy
[61] Iaccarino C, Crosio C, Vitale C, Sanna G, Carri body dementia alter -synuclein processing.
MT and Barone P. Apoptotic mechanisms in Ann Neurol 2011; 69: 940-953.
mutant LRRK2-mediated cell death. Hum Mol [72] Yang DS, Kumar A, Stavrides P, Peterson J, Pe-
Genet 2007; 16: 1319-1326. terhoff CM, Pawlik M, Levy E, Cataldo AM and
[62] Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan Nixon RA. Neuronal apoptosis and autophagy
R, Kelleher RJ 3rd and Shen J. Loss of leucine- cross talk in aging PS/APP mice, a model of
rich repeat kinase 2 causes impairment of pro- Alzheimers disease. Am J Pathol 2008; 173:
tein degradation pathways, accumulation of 665-681.
alpha-synuclein, and apoptotic cell death in [73] Kroemer G and Levine B. Autophagic cell
aged mice. Proc Natl Acad Sci U S A 2010; 107: death: the story of a misnomer. Nat Rev Mol
9879-9884. Cell Biol 2008; 9: 1004-1010.
[63] Qureshi HY and Paudel HK. Parkinsonian neu- [74] Harris H and Rubinsztein DC. Control of au-
rotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydro- tophagy as a therapy for neurodegenerative
pyridine (MPTP) and alpha-synuclein muta- disease. Nat Rev Neurol 2012; 8: 108-117.
tions promote Tau protein phosphorylation at [75] Atamna H and Frey WH 2nd. Mechanisms of
Ser262 and destabilize microtubule cytoskel- mitochondrial dysfunction and energy deficien-

163 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

cy in Alzheimers disease. Mitochondrion D, Cedazo-Minguez A and Cookson MR. DJ-1


2007; 7: 297-310. acts in parallel to the PINK1/parkin pathway to
[76] De Strooper B and Scorrano L. Close encoun- control mitochondrial function and autophagy.
ter: mitochondria, endoplasmic reticulum and Hum Mol Genet 2011; 20: 40-50.
Alzheimers disease. EMBO J 2012; 31: 4095- [89] Langston JW, Ballard P, Tetrud JW and Irwin I.
4097. Chronic Parkinsonism in humans due to a
[77] Chang S, ran Ma T, Miranda RD, Balestra ME, product of meperidine-analog synthesis. Sci-
Mahley RW and Huang Y. Lipid- and receptor- ence 1983; 219: 979-980.
binding regions of apolipoprotein E4 fragments [90] Blesa J, Phani S, Jackson-Lewis V and Przed-
act in concert to cause mitochondrial dysfunc- borski S. Classic and new animal models of
tion and neurotoxicity. Proc Natl Acad Sci U S A Parkinsons disease. J Biomed Biotechnol
2005; 102: 18694-18699. 2012; 2012: 845618.
[78] Bruck S, Huber TB, Ingham RJ, Kim K, Nieder- [91] Jenner P. Parkinsons disease, pesticides and
strasser H, Allen PM, Pawson T, Cooper JA and mitochondrial dysfunction. Trends Neurosci
Shaw AS. Identification of a novel inhibitory 2001; 24: 245-247.
actin-capping protein binding motif in CD2-as- [92] Yan MH, Wang X and Zhu X. Mitochondrial de-
sociated protein. J Biol Chem 2006; 281: fects and oxidative stress in Alzheimer disease
19196-19203. and Parkinson disease. Free Radic Biol Med
[79] DuBoff B, Gotz J and Feany MB. Tau promotes 2013 Sep; 62: 90-101.
neurodegeneration via DRP1 mislocalization [93] Lin MT and Beal MF. Mitochondrial dysfunction
in vivo. Neuron 2012; 75: 618-632. and oxidative stress in neurodegenerative dis-
[80] Iijima-Ando K, Sekiya M, Maruko-Otake A, eases. Nature 2006; 443: 787-795.
Ohtake Y, Suzuki E, Lu B and Iijima KM. Loss of [94] Squier TC. Oxidative stress and protein aggre-
axonal mitochondria promotes tau-mediated gation during biological aging. Exp Gerontol
neurodegeneration and Alzheimers disease- 2001; 36: 1539-1550.
related tau phosphorylation via PAR-1. PLoS [95] Kim RH, Smith PD, Aleyasin H, Hayley S, Mount
Genet 2012; 8: e1002918. MP, Pownall S, Wakeham A, You-Ten AJ, Kalia
[81] Krstic D and Knuesel I. Deciphering the mech- SK, Horne P, Westaway D, Lozano AM, Anis-
anism underlying late-onset Alzheimer dis- man H, Park DS and Mak TW. Hypersensitivity
ease. Nat Rev Neurol 2013; 9: 25-34. of DJ-1-deficient mice to 1-methyl-4-phenyl-
[82] Rupprecht R, Papadopoulos V, Rammes G, 1,2,3,6-tetrahydropyrindine (MPTP) and oxida-
Baghai TC, Fan J, Akula N, Groyer G, Adams D tive stress. Proc Natl Acad Sci U S A 2005;
and Schumacher M. Translocator protein (18 102: 5215-5220.
kDa) (TSPO) as a therapeutic target for neuro- [96] Irrcher I, Aleyasin H, Seifert EL, Hewitt SJ,
logical and psychiatric disorders. Nat Rev Drug Chhabra S, Phillips M, Lutz AK, Rousseaux
Discov 2010; 9: 971-988. MWC, Bevilacqua L, Jahani-Asl A, Callaghan S,
[83] Caselli RJ, Dueck AC, Huentelman MJ, Lutz MacLaurin JG, Winklhofer KF, Rizzu P, Ripp-
MW, Saunders AM, Reiman EM and Roses AD. stein P, Kim RH, Chen CX, Fon EA, Slack RS,
Longitudinal modeling of cognitive aging and Harper ME, McBride HM, Mak TW and Park DS.
the TOMM40 effect. Alzheimers Dement 2012; Loss of the Parkinsons disease-linked gene
8: 490-495. DJ-1 perturbs mitochondrial dynamics. Hum
[84] Henchcliffe C and Beal MF. Mitochondrial biol- Mol Genet 2010 Oct 1; 19: 3734-46.
ogy and oxidative stress in Parkinson disease [97] Wu ZC, Yu JT, Li Y and Tan L. Clusterin in Al-
pathogenesis. Nat Clin Pract Neurol 2008; 4: zheimers disease. Adv Clin Chem 2012; 56:
600-609. 155-173.
[85] Lesage S and Brice A. Role of mendelian genes [98] Allen M, Zou F, Chai HS, Younkin CS, Miles R,
in sporadic Parkinsons disease. Parkinson- Nair AA, Crook JE, Pankratz VS, Carrasquillo
ism Relat Disord 2012; 18 Suppl 1: S66-70. MM, Rowley CN, Nguyen T, Ma L, Malphrus KG,
[86] Palikaras K and Tavernarakis N. Mitophagy in Bisceglio G, Ortolaza AI, Palusak R, Middha S,
neurodegeneration and aging. Front Genet Maharjan S, Georgescu C, Schultz D, Rakh-
2012; 3: 297. shan F, Kolbert CP, Jen J, Sando SB, Aasly JO,
[87] Liu S, Sawada T, Lee S, Yu W, Silverio G, Alapatt Barcikowska M, Uitti RJ, Wszolek ZK, Ross OA,
P, Millan I, Shen A, Saxton W, Kanao T, Taka- Petersen RC, Graff-Radford NR, Dickson DW,
hashi R, Hattori N, Imai Y and Lu B. Parkinsons Younkin SG and Ertekin-Taner N. Glutathione
disease-associated kinase PINK1 regulates S-transferase omega genes in Alzheimer and
Miro protein level and axonal transport of mito- Parkinson disease risk, age-at-diagnosis and
chondria. PLoS Genet 2012; 8: e1002537. brain gene expression: an association study
[88] Thomas KJ, McCoy MK, Blackinton J, Beilina A, with mechanistic implications. Mol Neurode-
van der Brug M, Sandebring A, Miller D, Maric gener 2012; 7: 13.

164 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

[99] De Jager PL, Shulman JM, Chibnik LB, Keenan and Arancio O. Ubiquitin hydrolase Uch-L1 res-
BT, Raj T, Wilson RS, Yu L, Leurgans SE, Tran D, cues beta-amyloid-induced decreases in syn-
Aubin C, Anderson CD, Biffi A, Corneveaux JJ, aptic function and contextual memory. Cell
Huentelman MJ; Alzheimers Disease Neuro- 2006; 126: 775-788.
imaging Initiative; Rosand J, Daly MJ, Myers AJ, [111] Khandelwal PJ, Herman AM, Hoe HS, Rebeck
Reiman EM, Bennett DA and Evans DA. A ge- GW and Moussa CE. Parkin mediates beclin-
nome-wide scan for common variants affect- dependent autophagic clearance of defective
ing the rate of age-related cognitive decline. mitochondria and ubiquitinated Abeta in AD
Neurobiol Aging 2012; 33: 1017, e1011-1015. models. Hum Mol Genet 2011; 20: 2091-
[100] Bajaj A, Driver JA and Schernhammer ES. Par- 2102.
kinsons disease and cancer risk: a systematic [112] Huynh DP, Scoles DR, Nguyen D and Pulst SM.
review and meta-analysis. Cancer Causes Con- The autosomal recessive juvenile Parkinson
trol 2010; 21: 697-707. disease gene product, parkin, interacts with
[101] Driver JA, Beiser A, Au R, Kreger BE, Splansky and ubiquitinates synaptotagmin XI. Hum Mol
GL, Kurth T, Kiel DP, Lu KP, Seshadri S and Genet 2003; 12: 2587-2597.
Wolf PA. Inverse association between cancer [113] Rana A, Rera M and Walker DW. Parkin overex-
and Alzheimers disease: results from the pression during aging reduces proteotoxicity,
Framingham Heart Study. BMJ 2012; 344: alters mitochondrial dynamics, and extends
e1442. lifespan. Proc Natl Acad Sci U S A 2013; 110:
[102] Devine MJ, Plun-Favreau H and Wood NW. Par- 8638-8643.
kinsons disease and cancer: two wars, one [114] Horwitz AR. The origins of the molecular era of
front. Nat Rev Cancer 2011; 11: 812-823. adhesion research. Nat Rev Mol Cell Biol
[103] Tollervey JR, Wang Z, Hortobagyi T, Witten JT, 2012; 13: 805-811.
Zarnack K, Kayikci M, Clark TA, Schweitzer AC,
[115] Blalock EM, Geddes JW, Chen KC, Porter NM,
Rot G, Curk T, Zupan B, Rogelj B, Shaw CE and
Markesbery WR and Landfield PW. Incipient
Ule J. Analysis of alternative splicing associat-
Alzheimers disease: microarray correlation
ed with aging and neurodegeneration in the
analyses reveal major transcriptional and tu-
human brain. Genome Res 2011; 21: 1572-
mor suppressor responses. Proc Natl Acad Sci
1582.
U S A 2004; 101: 2173-2178.
[104] Kraft C, Peter M and Hofmann K. Selective au-
[116] Grunblatt E, Mandel S, Jacob-Hirsch J, Zelig-
tophagy: ubiquitin-mediated recognition and
son S, Amariglo N, Rechavi G, Li J, Ravid R,
beyond. Nat Cell Biol 2010; 12: 836-841.
[105] Ferrari R, Hardy J and Momeni P. Frontotempo- Roggendorf W, Riederer P and Youdim MB.
ral dementia: from Mendelian genetics to- Gene expression profiling of parkinsonian sub-
wards genome wide association studies. J Mol stantia nigra pars compacta; alterations in
Neurosci 2011; 45: 500-515. ubiquitin-proteasome, heat shock protein, iron
[106] Simpson JE, Ince PG, Shaw PJ, Heath PR, Ra- and oxidative stress regulated proteins, cell
man R, Garwood CJ, Gelsthorpe C, Baxter L, adhesion/cellular matrix and vesicle traffick-
Forster G, Matthews FE, Brayne C, Wharton ing genes. J Neural Transm 2004; 111: 1543-
SB; MRC Cognitive Function and Ageing Neuro- 1573.
pathology Study Group. Microarray analysis of [117] Miller RM, Kiser GL, Kaysser-Kranich TM, Lock-
the astrocyte transcriptome in the aging brain: ner RJ, Palaniappan C and Federoff HJ. Robust
relationship to Alzheimers pathology and dysregulation of gene expression in substantia
APOE genotype. Neurobiol Aging 2011; 32: nigra and striatum in Parkinsons disease.
1795-1807. Neurobiol Dis 2006; 21: 305-313.
[107] Kopito RR and Ron D. Conformational disease. [118] Xu PT, Li YJ, Qin XJ, Scherzer CR, Xu H, Sch-
Nat Cell Biol 2000; 2: E207-209. mechel DE, Hulette CM, Ervin J, Gullans SR,
[108] Scorrano L, Oakes SA, Opferman JT, Cheng EH, Haines J, Pericak-Vance MA and Gilbert JR. Dif-
Sorcinelli MD, Pozzan T and Korsmeyer SJ. BAX ferences in apolipoprotein E3/3 and E4/4 al-
and BAK regulation of endoplasmic reticulum lele-specific gene expression in hippocampus
Ca2+: a control point for apoptosis. Science in Alzheimer disease. Neurobiol Dis 2006; 21:
2003; 300: 135-139. 256-275.
[109] Smith DL, Pozueta J, Gong B, Arancio O and [119] Durakoglugil MS, Chen Y, White CL, Kavalali ET
Shelanski M. Reversal of long-term dendritic and Herz J. Reelin signaling antagonizes beta-
spine alterations in Alzheimer disease models. amyloid at the synapse. Proc Natl Acad Sci U S
Proc Natl Acad Sci U S A 2009; 106: 16877- A 2009; 106: 15938-15943.
16882. [120] Chen Y, Durakoglugil MS, Xian X and Herz J.
[110] Gong B, Cao Z, Zheng P, Vitolo OV, Liu S, Stan- ApoE4 reduces glutamate receptor function
iszewski A, Moolman D, Zhang H, Shelanski M and synaptic plasticity by selectively impairing

165 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

ApoE receptor recycling. Proc Natl Acad Sci U S [130] Zhang M. Endocytic mechanisms and drug dis-
A 2010; 107: 12011-12016. covery in neurodegenerative diseases. Front
[121] Herring A, Donath A, Steiner KM, Widera MP, Biosci 2008; 13: 6086-6105.
Hamzehian S, Kanakis D, Kolble K, ElAli A, Her- [131] Jiang Q, Lee CY, Mandrekar S, Wilkinson B,
mann DM, Paulus W and Keyvani K. Reelin Cramer P, Zelcer N, Mann K, Lamb B, Willson
depletion is an early phenomenon of Alzheim- TM, Collins JL, Richardson JC, Smith JD, Com-
ers pathology. J Alzheimers Dis 2012; 30: ery TA, Riddell D, Holtzman DM, Tontonoz P
963-979. and Landreth GE. ApoE promotes the proteo-
[122] Kramer PL, Xu H, Woltjer RL, Westaway SK, lytic degradation of Abeta. Neuron 2008; 58:
Clark D, Erten-Lyons D, Kaye JA, Welsh-Bohmer 681-693.
KA, Troncoso JC, Markesbery WR, Petersen RC, [132] Pottier C, Hannequin D, Coutant S, Rovelet-
Turner RS, Kukull WA, Bennett DA, Galasko D, Lecrux A, Wallon D, Rousseau S, Legallic S,
Morris JC and Ott J. Alzheimer disease pathol- Paquet C, Bombois S, Pariente J, Thomas-An-
ogy in cognitively healthy elderly: a genome- terion C, Michon A, Croisile B, Etcharry-Bouyx
wide study. Neurobiol Aging 2011; 32: 2113- F, Berr C, Dartigues JF, Amouyel P, Dauchel H,
2122. Boutoleau-Bretonniere C, Thauvin C, Frebourg
[123] Kopan R and Ilagan MX. Gamma-secretase: T, Lambert JC, Campion D and Collaborators
proteasome of the membrane? Nat Rev Mol PG. High frequency of potentially pathogenic
Cell Biol 2004; 5: 499-504. SORL1 mutations in autosomal dominant ear-
[124] Chen KP and Dou F. Selective interaction of ly-onset Alzheimer disease. Mol Psychiatry
amyloid precursor protein with different iso- 2012; 17: 875-879.
forms of neural cell adhesion molecule. J Mol [133] Reitz C, Cheng R, Rogaeva E, Lee JH, Tokuhiro
Neurosci 2012; 46: 203-209. S, Zou F, Bettens K, Sleegers K, Tan EK, Kimu-
[125] Wang Y, West JD, MaGee TR, Mcdonald BC, Ri- ra R, Shibata N, Arai H, Kamboh MI, Prince JA,
sacher SL, Shen L, Ramanan VK, Kim S, ONeill Maier W, Riemenschneider M, Owen M, Harold
DP, Farlow MR, Ghetti B and Saykin AJ. Hippo- D, Hollingworth P, Cellini E, Sorbi S, Nacmias B,
campal subfield atrophy on 3T MRI in prodro- Takeda M, Pericak-Vance MA, Haines JL,
mal Alzheimers disease and older adults with Younkin S, Williams J, van Broeckhoven C, Far-
cognitive complaints. Poster Presentation, Al- rer LA, St George-Hyslop PH, Mayeux R; Genet-
zheimers Association International Confer- ic and Environmental Risk in Alzheimer Dis-
ence (AAIC) in Vancouver, British Colombia, ease 1 Consortium. Meta-analysis of the
Canada, July 14-19, 2012. 2012. association between variants in SORL1 and
[126] Saykin AJ, Shen L, Foroud TM, Potkin SG, Alzheimer disease. Arch Neurol 2011; 68: 99-
Swaminathan S, Kim S, Risacher SL, Nho K, 106.
Huentelman MJ, Craig DW, Thompson PM, [134] Rogaeva E, Meng Y, Lee JH, Gu Y, Kawarai T,
Stein JL, Moore JH, Farrer LA, Green RC, Ber- Zou F, Katayama T, Baldwin CT, Cheng R,
tram L, Jack CR Jr, Weiner MW; Alzheimers Hasegawa H, Chen F, Shibata N, Lunetta KL,
Disease Neuroimaging Initiative. Alzheimers Pardossi-Piquard R, Bohm C, Wakutani Y, Cup-
Disease Neuroimaging Initiative biomarkers as ples LA, Cuenco KT, Green RC, Pinessi L,
quantitative phenotypes: Genetics core aims, Rainero I, Sorbi S, Bruni A, Duara R, Friedland
progress, and plans. Alzheimers Dement RP, Inzelberg R, Hampe W, Bujo H, Song YQ,
2010; 6: 265-273. Andersen OM, Willnow TE, Graff-Radford N, Pe-
[127] Edwards YJ, Beecham GW, Scott WK, Khuri S, tersen RC, Dickson D, Der SD, Fraser PE,
Bademci G, Tekin D, Martin ER, Jiang Z, Mash Schmitt-Ulms G, Younkin S, Mayeux R, Farrer
DC, ffrench-Mullen J, Pericak-Vance MA, LA and St George-Hyslop P. The neuronal sorti-
Tsinoremas N and Vance JM. Identifying con- lin-related receptor SORL1 is genetically asso-
sensus disease pathways in Parkinsons dis- ciated with Alzheimer disease. Nat Genet
ease using an integrative systems biology ap- 2007; 39: 168-177.
proach. PLoS One 2011; 6: e16917. [135] Golde TE, Estus S, Younkin LH, Selkoe DJ and
[128] Liu G, Jiang Y, Wang P, Feng R, Jiang N, Chen X, Younkin SG. Processing of the amyloid protein
Song H and Chen Z. Cell adhesion molecules precursor to potentially amyloidogenic deriva-
contribute to Alzheimers disease: multiple tives. Science 1992; 255: 728-730.
pathway analyses of two genome-wide associ- [136] Alegre-Abarrategui J and Wade-Martins R. Par-
ation studies. J Neurochem 2012; 120: 190- kinson disease, LRRK2 and the endocytic-au-
198. tophagic pathway. Autophagy 2009; 5: 1208-
[129] Holmans P. Statistical methods for pathway 1210.
analysis of genome-wide data for association [137] Alegre-Abarrategui J, Christian H, Lufino MM,
with complex genetic traits. Adv Genet 2010; Mutihac R, Venda LL, Ansorge O and Wade-
72: 141-179. Martins R. LRRK2 regulates autophagic activi-

166 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

ty and localizes to specific membrane microdo- [147] Edwards RH. Neural degeneration and the
mains in a novel human genomic reporter transport of neurotransmitters. Ann Neurol
cellular model. Hum Mol Genet 2009; 18: 1993; 34: 638-645.
4022-4034. [148] Corbett A, Pickett J, Burns A, Corcoran J, Dun-
[138] Matta S, Van Kolen K, da Cunha R, van den nett SB, Edison P, Hagan JJ, Holmes C, Jones E,
Bogaart G, Mandemakers W, Miskiewicz K, De Katona C, Kearns I, Kehoe P, Mudher A, Pass-
Bock PJ, Morais VA, Vilain S, Haddad D, Del- more A, Shepherd N, Walsh F and Ballard C.
broek L, Swerts J, Chavez-Gutierrez L, Esposito Drug repositioning for Alzheimers disease.
G, Daneels G, Karran E, Holt M, Gevaert K, Nat Rev Drug Discov 2012; 11: 833-846.
Moechars DW, De Strooper B and Verstreken [149] Antonini A and Albin RL. Dopaminergic treat-
P. LRRK2 controls an EndoA phosphorylation ment and nonmotor features of Parkinson dis-
cycle in synaptic endocytosis. Neuron 2012; ease: The horse lives. Neurology 2013; 80:
75: 1008-1021. 784-785.
[139] Cramer PE, Cirrito JR, Wesson DW, Lee CYD, [150] Ho A and Shen J. Presenilins in synaptic func-
Karlo JC, Zinn AE, Casali BT, Restivo JL, Goebel tion and disease. Trends Mol Med 2011; 17:
WD, James MJ, Brunden KR, Wilson DA and 617-624.
Landreth GE. ApoE-Directed Therapeutics Rap- [151] Sweatt JD. Mechanisms of Memory. 2nd Edi-
idly Clear -Amyloid and Reverse Deficits in AD tion. Academic Press, 2009.
Mouse Models. Science 2012 Mar 23; 335: [152] Wu K, OKeeffe D, Politis M, OKeeffe GC, Rob-
1503-6. bins TW, Bose SK, Brooks DJ, Piccini P and
[140] Treusch S, Hamamichi S, Goodman JL, Mat- Barker RA. The catechol-O-methyltransferase
lack KES, Chung CY, Baru V, Shulman JM, Par- Val(158)Met polymorphism modulates fronto-
rado A, Bevis BJ, Valastyan JS, Han H, Lindha- cortical dopamine turnover in early Parkin-
gen-Persson M, Reiman EM, Evans DA, Bennett sons disease: a PET study. Brain 2012; 135:
DA, Olofsson A, DeJager PL, Tanzi RE, Caldwell 2449-2457.
KA, Caldwell GA and Lindquist S. Functional [153] Serretti A and Olgiati P. Catechol-o-methyl-
Links Between A Toxicity, Endocytic Traffick- transferase and Alzheimers disease: a review
ing, and Alzheimers Disease Risk Factors in of biological and genetic findings. CNS Neurol
Yeast. Science 2011 Dec 2; 334: 1241-5. Disord Drug Targets 2012; 11: 299-305.
[141] Lee DW, Zhao X, Zhang F, Eisenberg E and [154] Spence RD and Voskuhl RR. Neuroprotective
Greene LE. Depletion of GAK/auxilin 2 inhibits
effects of estrogens and androgens in CNS in-
receptor-mediated endocytosis and recruit-
flammation and neurodegeneration. Front
ment of both clathrin and clathrin adaptors. J
Neuroendocrinol 2012; 33: 105-115.
Cell Sci 2005; 118: 4311-4321.
[155] Goldstein M and Lieberman A. The role of the
[142] Dumitriu A, Pacheco CD, Wilk JB, Strathearn
regulatory enzymes of catecholamine synthe-
KE, Latourelle JC, Goldwurm S, Pezzoli G, Ro-
sis in Parkinsons disease. Neurology 1992;
chet JC, Lindquist S and Myers RH. Cyclin-G-
42: 8-12; discussion 41-18.
associated kinase modifies alpha-synuclein
[156] Gainetdinov RR, Fumagalli F, Jones SR and
expression levels and toxicity in Parkinsons
Caron MG. Dopamine transporter is required
disease: results from the GenePD Study. Hum
for in vivo MPTP neurotoxicity: evidence from
Mol Genet 2011; 20: 1478-1487.
mice lacking the transporter. J Neurochem
[143] Henderson DM and Conner SD. A novel AAK1
1997; 69: 1322-1325.
splice variant functions at multiple steps of the
endocytic pathway. Mol Biol Cell 2007; 18: [157] Baranzini SE, Srinivasan R, Khankhanian P,
2698-2706. Okuda DT, Nelson SJ, Matthews PM, Hauser
[144] Latourelle JC, Pankratz N, Dumitriu A, Wilk JB, SL, Oksenberg JR and Pelletier D. Genetic vari-
Goldwurm S, Pezzoli G, Mariani CB, DeStefano ation influences glutamate concentrations in
AL, Halter C, Gusella JF, Nichols WC, Myers RH, brains of patients with multiple sclerosis. Brain
Foroud T; PROGENI Investigators, Coordinators 2010; 133: 2603-2611.
and Molecular Genetic Laboratories; GenePD [158] Mayeux R. Clinical practice. Early Alzheimers
Investigators, Coordinators and Molecular Ge- disease. N Engl J Med 2010; 362: 2194-2201.
netic Laboratories. Genomewide association [159] Alkalay A, Rabinovici GD, Zimmerman G, Agar-
study for onset age in Parkinson disease. BMC wal N, Kaufer D, Miller BL, Jagust WJ and
Med Genet 2009; 10: 98. Soreq H. Plasma acetylcholinesterase activity
[145] Davies P and Maloney AJ. Selective loss of cen- correlates with intracerebral beta-amyloid
tral cholinergic neurons in Alzheimers dis- load. Curr Alzheimer Res 2013; 10: 48-56.
ease. Lancet 1976; 2: 1403. [160] Ramanan VK, Risacher SL, Nho K, Kim S,
[146] Hoehn MM and Yahr MD. Parkinsonism: onset, Swaminathan S, Shen L, Foroud TM, Hakonar-
progression and mortality. Neurology 1967; son H, Huentelman MJ, Aisen PS, Petersen RC,
17: 427-442. Green RC, Jack CR, Koeppe RA, Jagust WJ,

167 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

Weiner MW and Saykin AJ. APOE and BCHE as 129 in idiopathic Parkinsons disease. J Neu-
modulators of cerebral amyloid deposition: a ral Transm 2006; 113: 331-337.
florbetapir PET genome-wide association [174] Combarros O, Sanchez-Guerra M, Llorca J, Al-
study. Mol Psychiatry 2013 Feb 19; [Epub varez-Arcaya A, Berciano J, Pena N and Fernan-
ahead of print]. dez-Viadero C. Polymorphism at codon 129 of
[161] Mesulam MM and Geula C. Butyrylcholinester- the prion protein gene is not associated with
ase reactivity differentiates the amyloid sporadic AD. Neurology 2000; 55: 593-595.
plaques of aging from those of dementia. Ann [175] Jeong BH, Lee KH, Lee YJ, Kim Y, Choi EK, Kim
Neurol 1994; 36: 722-727. YH, Cho YS, Carp R and Kim YS. Lack of asso-
[162] Darvesh S, Hopkins DA and Geula C. Neurobi- ciation between PRNP 1368 polymorphism
ology of butyrylcholinesterase. Nat Rev Neuro- and Alzheimers disease or vascular dementia.
sci 2003; 4: 131-138. BMC Med Genet 2009; 10: 32.
[163] Darreh-Shori T, Forsberg A, Modiri N, An- [176] Mead S, Poulter M, Uphill J, Beck J, Whitfield J,
dreasen N, Blennow K, Kamil C, Ahmed H, Alm- Webb TEF, Campbell T, Adamson G, Deriziotis
kvist O, Lngstrm B and Nordberg A. Differen- P, Tabrizi SJ, Hummerich H, Verzilli C, Alpers
tial levels of apolipoprotein E and butyrylcho- MP, Whittaker JC and Collinge J. Genetic risk
linesterase show strong association with path- factors for variant CreutzfeldtJakob disease:
ological signs of Alzheimers disease in the a genome-wide association study. Lancet Neu-
brain in vivo. Neurobiol Aging 2011 Dec; 32: rol 2009; 8: 57-66.
2320, e15-32. [177] Mead S, Uphill J, Beck J, Poulter M, Campbell T,
[164] Darvesh S, Cash MK, Reid GA, Martin E, Mit- Lowe J, Adamson G, Hummerich H, Klopp N,
nitski A and Geula C. Butyrylcholinesterase is Rckert IM, Wichmann HE, Azazi D, Plagnol V,
associated with beta-amyloid plaques in the Pako WH, Whitfield J, Alpers MP, Whittaker J,
transgenic APPSWE/PSEN1dE9 mouse model Balding DJ, Zerr I, Kretzschmar H and Collinge
of Alzheimer disease. J Neuropathol Exp Neu- J. Genome-wide association study in multiple
rol 2012; 71: 2-14. human prion diseases suggests genetic risk
[165] Lane RM and He Y. Butyrylcholinesterase gen- factors additional to PRNP. Hum Mol Genet
otype and gender influence Alzheimers dis- 2012 Apr 15; 21: 1897-906.
ease phenotype. Alzheimers Dement 2013 [178] Lukic A and Mead S. Genome wide association
Mar; 9: e1-73. studies and prion disease. Prion 2011; 5: 154-
[166] Sabbagh MN, Farlow MR, Relkin N and Beach 160.
TG. Do cholinergic therapies have disease- [179] Lansbury PT and Lashuel HA. A century-old de-
modifying effects in Alzheimers disease? Al- bate on protein aggregation and neurodegen-
zheimers Dement 2006 Apr; 2: 118-25. eration enters the clinic. Nature 2006; 443:
[167] Farlow MR, Miller ML and Pejovic V. Treatment 774-779.
options in Alzheimers disease: maximizing [180] Swardfager W, Lanctot K, Rothenburg L, Wong
benefit, managing expectations. Dement Geri- A, Cappell J and Herrmann N. A meta-analysis
atr Cogn Disord 2008; 25: 408-422. of cytokines in Alzheimers disease. Biol Psy-
[168] Caughey B and Baron GS. Prions and their chiatry 2010; 68: 930-941.
partners in crime. Nature 2006; 443: 803- [181] Akama KT and Van Eldik LJ. Beta-amyloid stim-
810. ulation of inducible nitric-oxide synthase in as-
[169] Prusiner SB. Scrapie prions. Annu Rev Micro- trocytes is interleukin-1beta- and tumor necro-
biol 1989; 43: 345-374. sis factor-alpha (TNFalpha)-dependent, and
[170] Prusiner SB. Cell biology. A unifying role for pri- involves a TNFalpha receptor-associated fac-
ons in neurodegenerative diseases. Science tor- and NFkappaB-inducing kinase-dependent
2012; 336: 1511-1513. signaling mechanism. J Biol Chem 2000; 275:
[171] Dermaut B, Croes EA, Rademakers R, Van den 7918-7924.
Broeck M, Cruts M, Hofman A, van Duijn CM [182] Blum-Degen D, Muller T, Kuhn W, Gerlach M,
and Van Broeckhoven C. PRNP Val129 homo- Przuntek H and Riederer P. Interleukin-1 beta
zygosity increases risk for early-onset Alzheim- and interleukin-6 are elevated in the cerebro-
ers disease. Ann Neurol 2003; 53: 409-412. spinal fluid of Alzheimers and de novo Parkin-
[172] Riemenschneider M, Klopp N, Xiang W, Wa- sons disease patients. Neurosci Lett 1995;
genpfeil S, Vollmert C, Muller U, Forstl H, Illig T, 202: 17-20.
Kretzschmar H and Kurz A. Prion protein codon [183] Hirsch EC and Hunot S. Neuroinflammation in
129 polymorphism and risk of Alzheimer dis- Parkinsons disease: a target for neuroprotec-
ease. Neurology 2004; 63: 364-366. tion? Lancet Neurol 2009; 8: 382-397.
[173] Gossrau G, Herting B, Mockel S, Kempe A, [184] Stewart WF, Kawas C, Corrada M and Metter
Koch R, Reichmann H and Lampe JB. Analysis EJ. Risk of Alzheimers disease and duration of
of the polymorphic prion protein gene codon NSAID use. Neurology 1997; 48: 626-632.

168 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

[185] Wahner AD, Bronstein JM, Bordelon YM and ciency impairs microglial accumulation and ac-
Ritz B. Nonsteroidal anti-inflammatory drugs celerates progression of Alzheimer-like dis-
may protect against Parkinson disease. Neu- ease. Nat Med 2007; 13: 432-438.
rology 2007; 69: 1836-1842. [197] Heneka MT, Nadrigny F, Regen T, Martinez-
[186] Prinz M, Priller J, Sisodia SS and Ransohoff Hernandez A, Dumitrescu-Ozimek L, Terwel D,
RM. Heterogeneity of CNS myeloid cells and Jardanhazi-Kurutz D, Walter J, Kirchhoff F,
their roles in neurodegeneration. Nat Neurosci Hanisch UK and Kummer MP. Locus ceruleus
2011; 14: 1227-1235. controls Alzheimers disease pathology by
[187] Lucin KM and Wyss-Coray T. Immune activa- modulating microglial functions through nor-
tion in brain aging and neurodegeneration: too epinephrine. Proc Natl Acad Sci U S A 2010;
much or too little? Neuron 2009; 64: 110-122. 107: 6058-6063.
[188] Lee EJ, Woo MS, Moon PG, Baek MC, Choi IY, [198] Ramos EM, Lin MT, Larson EB, Maezawa I,
Kim WK, Junn E and Kim HS. -Synuclein Acti- Tseng LH, Edwards KL, Schellenberg GD, Han-
vates Microglia by Inducing the Expressions of sen JA, Kukull WA and Jin LW. Tumor necrosis
Matrix Metalloproteinases and the Subse- factor alpha and interleukin 10 promoter re-
quent Activation of Protease-Activated Recep- gion polymorphisms and risk of late-onset Al-
tor-1. J Immunol 2010 Jul 1; 185: 615-23. zheimer disease. Arch Neurol 2006; 63: 1165-
[189] Halle A, Hornung V, Petzold GC, Stewart CR, 1169.
Monks BG, Reinheckel T, Fitzgerald KA, Latz E, [199] Wahner AD, Sinsheimer JS, Bronstein JM and
Moore KJ and Golenbock DT. The NALP3 in- Ritz B. Inflammatory cytokine gene polymor-
flammasome is involved in the innate immune phisms and increased risk of Parkinson dis-
response to amyloid-[beta]. Nat Immunol ease. Arch Neurol 2007; 64: 836-840.
2008; 9: 857-865. [200] Guerreiro R, Wojtas A, Bras J, Carrasquillo M,
[190] Kitazawa M, Cheng D, Tsukamoto MR, Koike Rogaeva E, Majounie E, Cruchaga C, Sassi C,
MA, Wes PD, Vasilevko V, Cribbs DH and LaFer-
Kauwe JS, Younkin S, Hazrati L, Collinge J, Po-
la FM. Blocking IL-1 Signaling Rescues Cogni-
cock J, Lashley T, Williams J, Lambert JC,
tion, Attenuates Tau Pathology, and Restores
Amouyel P, Goate A, Rademakers R, Morgan K,
Neuronal -Catenin Pathway Function in an Al-
Powell J, St George-Hyslop P, Singleton A, Har-
zheimers Disease Model. J Immunol 2011
dy J; Alzheimer Genetic Analysis Group. TREM2
Dec 15; 187: 6539-49.
variants in Alzheimers disease. N Engl J Med
[191] Pott Godoy MC, Tarelli R, Ferrari CC, Sarchi MI
2013; 368: 117-127.
and Pitossi FJ. Central and systemic IL-1 exac-
[201] Jonsson T, Stefansson H, Steinberg S, Jonsdot-
erbates neurodegeneration and motor symp-
tir I, Jonsson PV, Snaedal J, Bjornsson S, Hut-
toms in a model of Parkinsons disease. Brain
tenlocher J, Levey AI, Lah JJ, Rujescu D, Ham-
2008; 131: 1880-1894.
pel H, Giegling I, Andreassen OA, Engedal K,
[192] Heneka MT, Kummer MP, Stutz A, Delekate A,
Ulstein I, Djurovic S, Ibrahim-Verbaas C, Hof-
Schwartz S, Vieira-Saecker A, Griep A, Axt D,
man A, Ikram MA, van Duijn CM, Thorsteins-
Remus A, Tzeng TC, Gelpi E, Halle A, Korte M,
dottir U, Kong A and Stefansson K. Variant of
Latz E and Golenbock DT. NLRP3 is activated
TREM2 associated with the risk of Alzheimers
in Alzheimers disease and contributes to pa-
disease. N Engl J Med 2013; 368: 107-116.
thology in APP/PS1 mice. Nature 2013; 493:
674-678. [202] Hamerman JA, Jarjoura JR, Humphrey MB, Na-
[193] White GE and Greaves DR. Fractalkine: a survi- kamura MC, Seaman WE and Lanier LL. Cut-
vors guide: chemokines as antiapoptotic me- ting edge: inhibition of TLR and FcR responses
diators. Arterioscler Thromb Vasc Biol 2012; in macrophages by triggering receptor ex-
32: 589-594. pressed on myeloid cells (TREM)-2 and DAP12.
[194] Cardona AE, Pioro EP, Sasse ME, Kostenko V, J Immunol 2006; 177: 2051-2055.
Cardona SM, Dijkstra IM, Huang D, Kidd G, [203] Turnbull IR, Gilfillan S, Cella M, Aoshi T, Miller
Dombrowski S, Dutta R, Lee JC, Cook DN, Jung M, Piccio L, Hernandez M and Colonna M. Cut-
S, Lira SA, Littman DR and Ransohoff RM. Con- ting edge: TREM-2 attenuates macrophage ac-
trol of microglial neurotoxicity by the fractal- tivation. J Immunol 2006; 177: 3520-3524.
kine receptor. Nat Neurosci 2006; 9: 917-924. [204] Zhang B, Gaiteri C, Bodea LG, Wang Z, McEl-
[195] Bamberger ME, Harris ME, McDonald DR, wee J, Podtelezhnikov AA, Zhang C, Xie T, Tran
Husemann J and Landreth GE. A cell surface L, Dobrin R, Fluder E, Clurman B, Melquist S,
receptor complex for fibrillar beta-amyloid me- Narayanan M, Suver C, Shah H, Mahajan M,
diates microglial activation. J Neurosci 2003; Gillis T, Mysore J, MacDonald ME, Lamb JR,
23: 2665-2674. Bennett DA, Molony C, Stone DJ, Gudnason V,
[196] El Khoury J, Toft M, Hickman SE, Means TK, Myers AJ, Schadt EE, Neumann H, Zhu J and
Terada K, Geula C and Luster AD. Ccr2 defi- Emilsson V. Integrated systems approach iden-

169 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

tifies genetic nodes and networks in late-onset Bennett DA and De Jager PL. CR1 is associated
Alzheimers disease. Cell 2013; 153: 707-720. with amyloid plaque burden and age-related
[205] Moehle MS, Webber PJ, Tse T, Sukar N, Stan- cognitive decline. Ann Neurol 2011; 69: 560-
daert DG, DeSilva TM, Cowell RM and West AB. 569.
LRRK2 inhibition attenuates microglial inflam- [210] Wijsman EM, Pankratz ND, Choi Y, Rothstein
matory responses. J Neurosci 2012; 32: 1602- JH, Faber KM, Cheng R, Lee JH, Bird TD, Ben-
1611. nett DA, Diaz-Arrastia R, Goate AM, Farlow M,
[206] Kim B, Yang MS, Choi D, Kim JH, Kim HS, Seol Ghetti B, Sweet RA, Foroud TM, Mayeux R; NIA-
W, Choi S, Jou I, Kim EY and Joe EH. Impaired LOAD/NCRAD Family Study Group. Genome-
inflammatory responses in murine Lrrk2- Wide Association of Familial Late-Onset Al-
knockdown brain microglia. PLoS One 2012; 7: zheimers Disease Replicates BIN1 and CLU
e34693. and Nominates CUGBP2 in Interaction with
[207] Gardet A, Benita Y, Li C, Sands BE, Ballester I, APOE. PLoS Genet 2011; 7: e1001308.
Stevens C, Korzenik JR, Rioux JD, Daly MJ, Xavi- [211] Barral S, Bird T, Goate A, Farlow MR, Diaz-Ar-
er RJ and Podolsky DK. LRRK2 Is Invol- rastia R, Bennett DA, Graff-Radford N, Boeve
ved in the IFN- Response and Host Response BF, Sweet RA, Stern Y, Wilson RS, Foroud T, Ott
to Pathogens. J Immunol 2010; 185: 5577- J, Mayeux R; National Institute on Aging Late-
5585. Onset Alzheimers Disease Genetics Study.
[208] Jun G, Naj AC, Beecham GW, Wang LS, Buros J, Genotype patterns at PICALM, CR1, BIN1, CLU,
Gallins PJ, Buxbaum JD, Ertekin-Taner N, Fallin and APOE genes are associated with episodic
MD, Friedland R, Inzelberg R, Kramer P, Rogae- memory. Neurology 2012; 78: 1464-1471.
va E, St George-Hyslop P, Arnold SE, Baldwin [212] Keenan BT, Shulman JM, Chibnik LB, Raj T,
CT, Barber R, Beach T, Bigio EH, Bird TD, Boxer Tran D, Sabuncu MR; Alzheimers Disease
A, Burke JR, Cairns N, Carroll SL, Chui HC, Clark Neuroimaging Initiative; Allen AN, Corneveaux
DG, Cotman CW, Cummings JL, DeCarli C, Diaz- JJ, Hardy JA, Huentelman MJ, Lemere CA, My-
Arrastia R, Dick M, Dickson DW, Ellis WG, Fal- ers AJ, Nicholson-Weller A, Reiman EM, Evans
lon KB, Farlow MR, Ferris S, Frosch MP, Galas- DA, Bennett DA, De Jager PL. A coding variant
ko DR, Gearing M, Geschwind DH, Ghetti B, in CR1 interacts with APOE-epsilon4 to influ-
Gilman S, Giordani B, Glass J, Graff-Radford ence cognitive decline. Hum Mol Genet 2012;
NR, Green RC, Growdon JH, Hamilton RL, Har- 21: 2377-2388.
rell LE, Head E, Honig LS, Hulette CM, Hyman [213] Stephan AH, Barres BA and Stevens B. The
BT, Jicha GA, Jin LW, Johnson N, Karlawish J, complement system: an unexpected role in
Karydas A, Kaye JA, Kim R, Koo EH, Kowall NW,
synaptic pruning during development and dis-
Lah JJ, Levey AI, Lieberman A, Lopez OL, Mack
ease. Annu Rev Neurosci 2012; 35: 369-389.
WJ, Markesbery W, Marson DC, Martiniuk F,
[214] Evans MC, Couch Y, Sibson N and Turner MR.
Masliah E, McKee AC, Mesulam M, Miller JW,
Inflammation and neurovascular changes in
Miller BL, Miller CA, Parisi JE, Perl DP, Peskind
amyotrophic lateral sclerosis. Mol Cell Neuro-
E, Petersen RC, Poon W, Quinn JF, Raskind M,
sci 2013; 53: 34-41.
Reisberg B, Ringman JM, Roberson ED, Rosen-
berg RN, Sano M, Schneider JA, Schneider LS, [215] Yin F, Banerjee R, Thomas B, Zhou P, Qian L,
Seeley W, Shelanski ML, Smith CD, Spina S, Jia T, Ma X, Ma Y, Iadecola C, Beal MF, Nathan
Stern RA, Tanzi RE, Trojanowski JQ, Troncoso C and Ding A. Exaggerated inflammation, im-
JC, Van Deerlin VM, Vinters HV, Vonsattel JP, paired host defense, and neuropathology in
Weintraub S, Welsh-Bohmer KA, Woltjer RL, progranulin-deficient mice. J Exp Med 2010;
Younkin SG, Cantwell LB, Dombroski BA, 207: 117-128.
Saykin AJ, Reiman EM, Bennett DA, Morris JC, [216] Jones KA and Thomsen C. The role of the in-
Lunetta KL, Martin ER, Montine TJ, Goate AM, nate immune system in psychiatric disorders.
Blacker D, Tsuang DW, Beekly D, Cupples LA, Mol Cell Neurosci 2013; 53: 52-62.
Hakonarson H, Kukull W, Foroud TM, Haines J, [217] Vom Berg J, Prokop S, Miller KR, Obst J, Kalin
Mayeux R, Farrer LA, Pericak-Vance MA, Schel- RE, Lopategui-Cabezas I, Wegner A, Mair F,
lenberg GD; Alzheimers Disease Genetics Schipke CG, Peters O, Winter Y, Becher B and
Consortium. Meta-analysis Confirms CR1, CLU, Heppner FL. Inhibition of IL-12/IL-23 signaling
and PICALM as Alzheimer Disease Risk Loci reduces Alzheimers disease-like pathology
and Reveals Interactions With APOE Geno- and cognitive decline. Nat Med 2012; 18:
types. Arch Neurol 2010; 67: 1473-1484. 1812-1819.
[209] Chibnik LB, Shulman JM, Leurgans SE, Schnei- [218] Gao HM, Liu B, Zhang W and Hong JS. Novel
der JA, Wilson RS, Tran D, Aubin C, Buchman anti-inflammatory therapy for Parkinsons dis-
AS, Heward CB, Myers AJ, Hardy JA, Huentel- ease. Trends Pharmacol Sci 2003; 24: 395-
man MJ, Corneveaux JJ, Reiman EM, Evans DA, 401.

170 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

[219] Tan ZS and Seshadri S. Inflammation in the Al- Amar M, Remaley AT and Sack MN. Parkin is a
zheimers disease cascade: culprit or innocent lipid-responsive regulator of fat uptake in mice
bystander? Alzheimers Res Ther 2010; 2: 6. and mutant human cells. J Clin Invest 2011;
[220] Martins IJ, Hone E, Foster JK, Sunram-Lea SI, 121: 3701-3712.
Gnjec A, Fuller SJ, Nolan D, Gandy SE and Mar- [233] Hatano T, Kubo S, Imai S, Maeda M, Ishikawa
tins RN. Apolipoprotein E, cholesterol metabo- K, Mizuno Y and Hattori N. Leucine-rich repeat
lism, diabetes, and the convergence of risk kinase 2 associates with lipid rafts. Hum Mol
factors for Alzheimers disease and cardiovas- Genet 2007; 16: 678-690.
cular disease. Mol Psychiatry 2006; 11: 721- [234] Chan RB, Oliveira TG, Cortes EP, Honig LS, Duff
736. KE, Small SA, Wenk MR, Shui G and Di Paolo
[221] Sparks DL. Alzheimer disease: statins in the G. Comparative lipidomic analysis of mouse
treatment of Alzheimer disease. Nat Rev Neu- and human brain with Alzheimer disease. J
rol 2011; 7: 662-663. Biol Chem 2012; 287: 2678-2688.
[222] Gao X, Simon KC, Schwarzschild MA and [235] Cheng D, Jenner AM, Shui G, Cheong WF,
Ascherio A. Prospective study of statin use and Mitchell TW, Nealon JR, Kim WS, McCann H,
risk of Parkinson disease. Arch Neurol 2012; Wenk MR, Halliday GM and Garner B. Lipid
69: 380-384. pathway alterations in Parkinsons disease pri-
[223] Karasinska JM and Hayden MR. Cholesterol mary visual cortex. PLoS One 2011; 6: e17299.
metabolism in Huntington disease. Nat Rev [236] Ho PP, Kanter JL, Johnson AM, Srinagesh HK,
Neurol 2011; 7: 561-572. Chang EJ, Purdy TM, van Haren K, Wikoff WR,
[224] Anchisi L, Dessi S, Pani A and Mandas A. Cho- Kind T, Khademi M, Matloff LY, Narayana S,
lesterol homeostasis: a key to prevent or slow Hur EM, Lindstrom TM, He Z, Fiehn O, Olsson T,
down neurodegeneration. Front Physiol 2012; Han X, Han MH, Steinman L and Robinson WH.
3: 486. Identification of Naturally Occurring Fatty Acids
[225] Han X, Rozen S, Boyle SH, Hellegers C, Cheng of the Myelin Sheath That Resolve Neuroin-
H, Burke JR, Welsh-Bohmer KA, Doraiswamy flammation. Sci Transl Med 2012; 4: 137ra173.
PM and Kaddurah-Daouk R. Metabolomics in
[237] Willey JZ and Elkind MS. 3-Hydroxy-3-methylgl-
early Alzheimers disease: identification of al-
utaryl-coenzyme A reductase inhibitors in the
tered plasma sphingolipidome using shotgun
treatment of central nervous system diseases.
lipidomics. PLoS One 2011; 6: e21643.
Arch Neurol 2010; 67: 1062-1067.
[226] Wood PL. Lipidomics of Alzheimers disease:
[238] Kroner Z. The relationship between Alzheim-
current status. Alzheimers Res Ther 2012; 4:
ers disease and diabetes: type 3 diabetes?
5.
Altern Med Rev 2009; 14: 373-379.
[227] Holtzman DM, Herz J and Bu G. Apolipoprotein
e and apolipoprotein e receptors: normal biol- [239] Liu Y, Liu F, Grundke-Iqbal I, Iqbal K and Gong
ogy and roles in Alzheimer disease. Cold Spring CX. Deficient brain insulin signalling pathway
Harb Perspect Med 2012; 2: a006312. in Alzheimers disease and diabetes. J Pathol
[228] Calero M, Tokuda T, Rostagno A, Kumar A, Zlo- 2011; 225: 54-62.
kovic B, Frangione B and Ghiso J. Functional [240] Vignini A, Giulietti A, Nanetti L, Raffaelli F, Gi-
and structural properties of lipid-associated usti L, Mazzanti L and Provinciali L. Alzheim-
apolipoprotein J (clusterin). Biochem J 1999; ers disease And Diabetes: New Insights and
344: 375-383. Unifying Therapies. Curr Diabetes Rev 2013
[229] Kim WS, Weickert CS and Garner B. Role of May; 9: 218-27.
ATP-binding cassette transporters in brain lipid [241] de la Monte SM. Contributions of brain insulin
transport and neurological disease. J Neuro- resistance and deficiency in amyloid-related
chem 2008; 104: 1145-1166. neurodegeneration in Alzheimers disease.
[230] Soscia SJ and Fitzgerald ML. The ABCA7 trans- Drugs 2012; 72: 49-66.
porter, brain lipids and Alzheimers disease. [242] Chen DY, Stern SA, Garcia-Osta A, Saunier-Re-
Clinical Lipidology 2013; 8: 97-108. bori B, Pollonini G, Bambah-Mukku D, Blitzer
[231] Biskup S, Moore DJ, Celsi F, Higashi S, West RD and Alberini CM. A critical role for IGF-II in
AB, Andrabi SA, Kurkinen K, Yu SW, Savitt JM, memory consolidation and enhancement. Na-
Waldvogel HJ, Faull RL, Emson PC, Torp R, Ot- ture 2011; 469: 491-497.
tersen OP, Dawson TM and Dawson VL. Local- [243] Lue LF, Andrade C, Sabbagh M and Walker D.
ization of LRRK2 to membranous and vesicu- Is There Inflammatory Synergy in Type II Diabe-
lar structures in mammalian brain. Ann Neurol tes Mellitus and Alzheimers Disease? Int J Al-
2006; 60: 557-569. zheimers Dis 2012; 2012: 918680.
[232] Kim KY, Stevens MV, Akter MH, Rusk SE, [244] Iwasaki T, Yoneda M, Nakajima A and Terauchi
Huang RJ, Cohen A, Noguchi A, Springer D, Bo- Y. Serum Butyrylcholinesterase is Strongly As-
charov AV, Eggerman TL, Suen DF, Youle RJ, sociated with Adiposity, the Serum Lipid Profile

171 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

and Insulin Resistance. Intern Med 2007; 46: [257] Alvarez JI, Dodelet-Devillers A, Kebir H, Ifergan
1633-9. I, Fabre PJ, Terouz S, Sabbagh M, Wosik K,
[245] Meigs JB, Manning AK, Fox CS, Florez JC, Liu C, Bourbonniere L, Bernard M, van Horssen J, de
Cupples LA and Dupuis J. Genome-wide asso- Vries HE, Charron F and Prat A. The Hedgehog
ciation with diabetes-related traits in the Fram- pathway promotes blood-brain barrier integrity
ingham Heart Study. BMC Med Genet 2007; 8 and CNS immune quiescence. Science 2011;
Suppl 1: S16. 334: 1727-1731.
[246] Gao C, Holscher C, Liu Y and Li L. GSK3: a key [258] Schrijvers EM, Schurmann B, Koudstaal PJ,
target for the development of novel treatments van den Bussche H, Van Duijn CM, Hentschel
for type 2 diabetes mellitus and Alzheimer dis- F, Heun R, Hofman A, Jessen F, Kolsch H, Korn-
ease. Rev Neurosci 2012; 23: 1-11. huber J, Peters O, Rivadeneira F, Ruther E, Uit-
[247] Akinyemi R, Mukaetova-Ladinska E, Attems J, terlinden AG, Riedel-Heller S, Dichgans M, Wilt-
Attems J, Ihara M and Kalaria RN. Vascular fang J, Maier W, Breteler MM and Ikram MA.
Risk Factors and Neurodegeneration in Ageing Genome-wide association study of vascular
related Dementias: Alzheimers Disease and dementia. Stroke 2012; 43: 315-319.
Vascular Dementia. Curr Alzheimer Res 2013 [259] Chuang DM, Leng Y, Marinova Z, Kim HJ and
Jul; 10: 642-53. Chiu CT. Multiple roles of HDAC inhibition in
[248] McCarron MO, Delong D and Alberts MJ. APOE neurodegenerative conditions. Trends Neuro-
genotype as a risk factor for ischemic cerebro- sci 2009; 32: 591-601.
vascular disease: a meta-analysis. Neurology [260] Egger G, Liang G, Aparicio A and Jones PA. Epi-
1999; 53: 1308-1311. genetics in human disease and prospects for
epigenetic therapy. Nature 2004; 429: 457-
[249] Desai BS, Monahan AJ, Carvey PM and Hendey
463.
B. Blood-brain barrier pathology in Alzheimers
[261] Fraga MF, Ballestar E, Paz MF, Ropero S, Se-
and Parkinsons disease: implications for drug
tien F, Ballestar ML, Heine-Suner D, Cigudosa
therapy. Cell Transplant 2007; 16: 285-299.
JC, Urioste M, Benitez J, Boix-Chornet M, San-
[250] Chen X, Ghribi O and Geiger JD. Caffeine pro-
chez-Aguilera A, Ling C, Carlsson E, Poulsen P,
tects against disruptions of the blood-brain
Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C
barrier in animal models of Alzheimers and
and Esteller M. Epigenetic differences arise
Parkinsons diseases. J Alzheimers Dis 2010;
during the lifetime of monozygotic twins. Proc
20 Suppl 1: S127-141.
Natl Acad Sci U S A 2005; 102: 10604-10609.
[251] Bartels AL. Blood-brain barrier P-glycoprotein [262] Kontopoulos E, Parvin JD and Feany MB. Al-
function in neurodegenerative disease. Curr pha-synuclein acts in the nucleus to inhibit his-
Pharm Des 2011; 17: 2771-2777. tone acetylation and promote neurotoxicity.
[252] Silver M, Janousova E, Hua X, Thompson PM Hum Mol Genet 2006; 15: 3012-3023.
and Montana G. Identification of gene path- [263] Outeiro TF, Kontopoulos E, Altmann SM, Kufa-
ways implicated in Alzheimers disease using reva I, Strathearn KE, Amore AM, Volk CB, Max-
longitudinal imaging phenotypes with sparse well MM, Rochet JC, McLean PJ, Young AB, Ab-
regression. Neuroimage 2012; 63: 1681- agyan R, Feany MB, Hyman BT and Kazantsev
1694. AG. Sirtuin 2 inhibitors rescue alpha-synuclein-
[253] Biffi A, Shulman JM, Jagiella JM, Cortellini L, mediated toxicity in models of Parkinsons dis-
Ayres AM, Schwab K, Brown DL, Silliman SL, ease. Science 2007; 317: 516-519.
Selim M, Worrall BB, Meschia JF, Slowik A, De [264] Graff J, Rei D, Guan JS, Wang WY, Seo J, Hen-
Jager PL, Greenberg SM, Schneider JA, Ben- nig KM, Nieland TJ, Fass DM, Kao PF, Kahn M,
nett DA and Rosand J. Genetic variation at CR1 Su SC, Samiei A, Joseph N, Haggarty SJ, Delal-
increases risk of cerebral amyloid angiopathy. le I and Tsai LH. An epigenetic blockade of cog-
Neurology 2012; 78: 334-341. nitive functions in the neurodegenerating
[254] Mihci E, Ozkaynak SS, Sallakci N, Kizilay F and brain. Nature 2012; 483: 222-226.
Yavuzer U. VEGF polymorphisms and serum [265] Donmez G, Wang D, Cohen DE and Guarente L.
VEGF levels in Parkinsons disease. Neurosci SIRT1 suppresses beta-amyloid production by
Lett 2011; 494: 1-5. activating the alpha-secretase gene ADAM10.
[255] He D, Lu W, Chang K, Liu Y, Zhang J and Zeng Cell 2010; 142: 320-332.
Z. Vascular endothelial growth factor polymor- [266] Beck J, Poulter M, Hensman D, Rohrer JD, Ma-
phisms and risk of Alzheimers disease: a me- honey CJ, Adamson G, Campbell T, Uphill J,
ta-analysis. Gene 2013; 518: 296-302. Borg A, Fratta P, Orrell RW, Malaspina A, Rowe
[256] Topiwala A and Ebmeier KP. Vascular changes J, Brown J, Hodges J, Sidle K, Polke JM, Houl-
and brain plasticity: a new approach to neuro- den H, Schott JM, Fox NC, Rossor MN, Tabrizi
degenerative diseases. Am J Neurodegener SJ, Isaacs AM, Hardy J, Warren JD, Collinge J
Dis 2012; 1: 152-159. and Mead S. Large C9orf72 hexanucleotide

172 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

repeat expansions are seen in multiple neuro- (CDNF) gene and Parkinson disease. Neurosci
degenerative syndromes and are more fre- Lett 2011; 493: 97-101.
quent than expected in the UK population. Am [277] Lin LF, Doherty DH, Lile JD, Bektesh S and Col-
J Hum Genet 2013; 92: 345-353. lins F. GDNF: a glial cell line-derived neuro-
[267] Mori K, Weng SM, Arzberger T, May S, Rent- trophic factor for midbrain dopaminergic neu-
zsch K, Kremmer E, Schmid B, Kretzschmar rons. Science 1993; 260: 1130-1132.
HA, Cruts M, Van Broeckhoven C, Haass C and [278] Gash DM, Zhang Z, Ovadia A, Cass WA, Yi A,
Edbauer D. The C9orf72 GGGGCC repeat is Simmerman L, Russell D, Martin D, Lapchak
translated into aggregating dipeptide-repeat PA, Collins F, Hoffer BJ and Gerhardt GA. Func-
proteins in FTLD/ALS. Science 2013; 339: tional recovery in parkinsonian monkeys treat-
1335-1338. ed with GDNF. Nature 1996; 380: 252-255.
[268] Taylor JP. Neuroscience. RNA that gets RAN in [279] Patani N, Jiang WG and Mokbel K. Brain-de-
neurodegeneration. Science 2013; 339: rived neurotrophic factor expression predicts
1282-1283. adverse pathological & clinical outcomes in
[269] Rao JS, Keleshian VL, Klein S and Rapoport SI. human breast cancer. Cancer Cell Int 2011;
Epigenetic modifications in frontal cortex from 11: 23.
Alzheimers disease and bipolar disorder pa- [280] Cao L and During MJ. What is the brain-cancer
tients. Transl Psychiatry 2012; 2: e132. connection? Annu Rev Neurosci 2012; 35:
331-345.
[270] Chowdary PD, Che DL and Cui B. Neurotrophin
[281] Calado RT and Young NS. Telomere diseases.
signaling via long-distance axonal transport.
N Engl J Med 2009; 361: 2353-2365.
Annu Rev Phys Chem 2012; 63: 571-594.
[282] Hadley EC, Lakatta EG, Morrison-Bogorad M,
[271] Schindowski K, Belarbi K and Buee L. Neuro-
Warner HR and Hodes RJ. The future of aging
trophic factors in Alzheimers disease: role of
therapies. Cell 2005; 120: 557-567.
axonal transport. Genes Brain Behav 2008; 7
[283] Honig LS, Kang MS, Schupf N, Lee JH and May-
Suppl 1: 43-56.
eux R. Association of shorter leukocyte telo-
[272] Kauwe JS, Wang J, Mayo K, Morris JC, Fagan mere repeat length with dementia and mortal-
AM, Holtzman DM and Goate AM. Alzheimers ity. Arch Neurol 2012; 69: 1332-1339.
disease risk variants show association with ce- [284] Rolyan H, Scheffold A, Heinrich A, Begus-Nah-
rebrospinal fluid amyloid beta. Neurogenetics rmann Y, Langkopf BH, Holter SM, Vogt-
2009; 10: 13-17. Weisenhorn DM, Liss B, Wurst W, Lie DC, Thal
[273] Karamohamed S, Latourelle JC, Racette BA, DR, Biber K and Rudolph KL. Telomere short-
Perlmutter JS, Wooten GF, Lew M, Klein C, Shill ening reduces Alzheimers disease amyloid
H, Golbe LI, Mark MH, Guttman M, Nicholson pathology in mice. Brain 2011; 134: 2044-
G, Wilk JB, Saint-Hilaire M, DeStefano AL, 2056.
Prakash R, Tobin S, Williamson J, Suchowersky [285] Wang H, Chen H, Gao X, McGrath M, Deer D,
O, Labell N, Growdon BN, Singer C, Watts R, De Vivo I, Schwarzschild MA and Ascherio A.
Goldwurm S, Pezzoli G, Baker KB, Giroux ML, Telomere length and risk of Parkinsons dis-
Pramstaller PP, Burn DJ, Chinnery P, Sherman ease. Mov Disord 2008; 23: 302-305.
S, Vieregge P, Litvan I, Gusella JF, Myers RH [286] Eerola J, Kananen L, Manninen K, Hellstrom O,
and Parsian A. BDNF genetic variants are as- Tienari PJ and Hovatta I. No evidence for short-
sociated with onset age of familial Parkinson er leukocyte telomere length in Parkinsons
disease: GenePD Study. Neurology 2005; 65: disease patients. J Gerontol A Biol Sci Med Sci
1823-1825. 2010; 65: 1181-1184.
[274] Voineskos AN, Lerch JP, Felsky D, Shaikh S, Ra- [287] Hudson G, Faini D, Stutt A, Eccles M, Robinson
jji TK, Miranda D, Lobaugh NJ, Mulsant BH, L, Burn DJ and Chinnery PF. No evidence of
Pollock BG and Kennedy JL. The brain-derived substantia nigra telomere shortening in Par-
neurotrophic factor Val66Met polymorphism kinsons disease. Neurobiol Aging 2011; 32:
and prediction of neural risk for Alzheimer dis- 2107, e2103-2105.
ease. Arch Gen Psychiatry 2011; 68: 198-206. [288] Njajou OT, Blackburn EH, Pawlikowska L,
[275] Chen DY, Bambah-Mukku D, Pollonini G and Mangino M, Damcott CM, Kwok PY, Spector
Alberini CM. Glucocorticoid receptors recruit TD, Newman AB, Harris TB, Cummings SR,
the CaMKII alpha-BDNF-CREB pathways to me- Cawthon RM, Shuldiner AR, Valdes AM and
diate memory consolidation. Nature Neurosci- Hsueh WC. A common variant in the telomer-
ence 2012; 15: 1707-14. ase RNA component is associated with short
[276] Choi JM, Hong JH, Chae MJ, Ngyuen PH, Kang telomere length. PLoS One 2010; 5: e13048.
HS, Ma HI and Kim YJ. Analysis of mutations [289] Shen Q, Zhang Z, Yu L, Cao L, Zhou D, Kan M,
and the association between polymorphisms Li B, Zhang D, He L and Liu Y. Common vari-
in the cerebral dopamine neurotrophic factor ants near TERC are associated with leukocyte

173 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

telomere length in the Chinese Han popula- [297] Chu J, Zhuo JM and Pratico D. Transcriptional
tion. Eur J Hum Genet 2011; 19: 721-723. regulation of beta-secretase-1 by 12/15-lipox-
[290] Kim S, Parks CG, Xu Z, Carswell G, DeRoo LA, ygenase results in enhanced amyloidogenesis
Sandler DP and Taylor JA. Association between and cognitive impairments. Ann Neurol 2012;
genetic variants in DNA and histone methyla- 71: 57-67.
tion and telomere length. PLoS One 2012; 7: [298] Santpere G, Nieto M, Puig B and Ferrer I. Ab-
e40504. normal Sp1 transcription factor expression in
[291] Deelen J, Uh HW, Monajemi R, van Heemst D, Alzheimer disease and tauopathies. Neurosci
Thijssen PE, Bohringer S, van den Akker EB, de Lett 2006; 397: 30-34.
Craen AJ, Rivadeneira F, Uitterlinden AG, West- [299] Karin M, Liu Z and Zandi E. AP-1 function and
endorp RG, Goeman JJ, Slagboom PE, Hou- regulation. Curr Opin Cell Biol 1997; 9: 240-
wing-Duistermaat JJ and Beekman M. Gene 246.
set analysis of GWAS data for human longevity [300] Calon F, Grondin R, Morissette M, Goulet M,
highlights the relevance of the insulin/IGF-1 Blanchet PJ, Di Paolo T and Bedard PJ. Molecu-
signaling and telomere maintenance path- lar basis of levodopa-induced dyskinesias. Ann
ways. Age (Dordr) 2013; 35: 235-249. Neurol 2000; 47: S70-78.
[292] Fossel M. Telomerase and the aging cell: impli- [301] Andersson M, Konradi C and Cenci MA. cAMP
cations for human health. JAMA 1998; 279: response element-binding protein is required
1732-1735. for dopamine-dependent gene expression in
the intact but not the dopamine-denervated
[293] Bertram L, McQueen MB, Mullin K, Blacker D
striatum. J Neurosci 2001; 21: 9930-9943.
and Tanzi RE. Systematic meta-analyses of Al-
[302] Jegga AG, Schneider L, Ouyang X and Zhang J.
zheimer disease genetic association studies:
Systems biology of the autophagy-lysosomal
the AlzGene database. Nat Genet 2007; 39:
pathway. Autophagy 2011; 7: 477-489.
17-23.
[303] Fahrig T, Gerlach I and Horvath E. A synthetic
[294] Lill CM, Roehr JT, McQueen MB, Kavvoura FK,
derivative of the natural product rocaglaol is a
Bagade S, Schjeide BM, Schjeide LM, Meiss-
potent inhibitor of cytokine-mediated signaling
ner E, Zauft U, Allen NC, Liu T, Schilling M, An-
and shows neuroprotective activity in vitro and
derson KJ, Beecham G, Berg D, Biernacka JM, in animal models of Parkinsons disease and
Brice A, DeStefano AL, Do CB, Eriksson N, Fac- traumatic brain injury. Mol Pharmacol 2005;
tor SA, Farrer MJ, Foroud T, Gasser T, Hamza T, 67: 1544-1555.
Hardy JA, Heutink P, Hill-Burns EM, Klein C, [304] Perez-Cadahia B, Drobic B and Davie JR. Acti-
Latourelle JC, Maraganore DM, Martin ER, vation and function of immediate-early genes
Martinez M, Myers RH, Nalls MA, Pankratz N, in the nervous system. Biochem Cell Biol 2011;
Payami H, Satake W, Scott WK, Sharma M, 89: 61-73.
Singleton AB, Stefansson K, Toda T, Tung JY, [305] Gomez Ravetti M, Rosso OA, Berretta R and
Vance J, Wood NW, Zabetian CP; 23andMe Ge- Moscato P. Uncovering molecular biomarkers
netic Epidemiology of Parkinsons Disease that correlate cognitive decline with the chang-
Consortium; International Parkinsons Disease es of hippocampus gene expression profiles in
Genomics Consortium; Parkinsons Disease Alzheimers disease. PLoS One 2010; 5:
GWAS Consortium; Wellcome Trust Case Con- e10153.
trol Consortium 2); Young P, Tanzi RE, Khoury [306] Lu Y, Li T, Qureshi HY, Han D and Paudel HK.
MJ, Zipp F, Lehrach H, Ioannidis JP, Bertram L. Early growth response 1 (Egr-1) regulates
Comprehensive research synopsis and sys- phosphorylation of microtubule-associated
tematic meta-analyses in Parkinsons disease protein tau in mammalian brain. J Biol Chem
genetics: The PDGene database. PLoS Genet 2011; 286: 20569-20581.
2012; 8: e1002548. [307] Lindersson EK, Hojrup P, Gai WP, Locker D,
[295] Maloney B, Ge YW, Petersen RC, Hardy J, Rog- Martin D and Jensen PH. alpha-Synuclein fila-
ers JT, Perez-Tur J and Lahiri DK. Functional ments bind the transcriptional regulator
characterization of three single-nucleotide HMGB-1. Neuroreport 2004; 15: 2735-2739.
polymorphisms present in the human APOE [308] Takata K, Kitamura Y, Kakimura J, Shibagaki K,
promoter sequence: Differential effects in neu- Tsuchiya D, Taniguchi T, Smith MA, Perry G and
ronal cells and on DNA-protein interactions. Shimohama S. Role of high mobility group pro-
Am J Med Genet B Neuropsychiatr Genet tein-1 (HMG1) in amyloid-beta homeostasis.
2010; 153B: 185-201. Biochem Biophys Res Commun 2003; 301:
[296] Citron BA, Dennis JS, Zeitlin RS and Echeverria 699-703.
V. Transcription factor Sp1 dysregulation in Al- [309] Takata K, Takada T, Ito A, Asai M, Tawa M, Saito
zheimers disease. J Neurosci Res 2008; 86: Y, Ashihara E, Tomimoto H, Kitamura Y and Shi-
2499-2504. mohama S. Microglial Amyloid-beta1-40

174 Am J Neurodegener Dis 2013;2(3):145-175


Pathways to neurodegeneration in AD and PD

Phagocytosis Dysfunction Is Caused by High- [319] Jaeger S and Pietrzik CU. Functional role of li-
Mobility Group Box Protein-1: Implications for poprotein receptors in Alzheimers disease.
the Pathological Progression of Alzheimers Curr Alzheimer Res 2008; 5: 15-25.
Disease. Int J Alzheimers Dis 2012; 2012: [320] Waldron E, Heilig C, Schweitzer A, Nadella N,
685739. Jaeger S, Martin AM, Weggen S, Brix K and Pi-
[310] Scaffidi P, Misteli T and Bianchi ME. Release of etrzik CU. LRP1 modulates APP trafficking
chromatin protein HMGB1 by necrotic cells along early compartments of the secretory
triggers inflammation. Nature 2002; 418: 191- pathway. Neurobiol Dis 2008; 31: 188-197.
195. [321] Liu Q, Trotter J, Zhang J, Peters MM, Cheng H,
[311] Gao HM, Zhou H, Zhang F, Wilson BC, Kam W Bao J, Han X, Weeber EJ and Bu G. Neuronal
and Hong JS. HMGB1 acts on microglia Mac1 LRP1 knockout in adult mice leads to impaired
to mediate chronic neuroinflammation that brain lipid metabolism and progressive, age-
drives progressive neurodegeneration. J Neu- dependent synapse loss and neurodegenera-
rosci 2011; 31: 1081-1092. tion. J Neurosci 2010; 30: 17068-17078.
[312] Clifford DB, Fagan AM, Holtzman DM, Morris [322] Sloan CD, Shen L, West JD, Wishart HA, Flash-
JC, Teshome M, Shah AR and Kauwe JS. CSF man LA, Rabin LA, Santulli RB, Guerin SJ,
biomarkers of Alzheimer disease in HIV-associ- Rhodes CH, Tsongalis GJ, McAllister TW, Ahles
ated neurologic disease. Neurology 2009; 73: TA, Lee SL, Moore JH and Saykin AJ. Genetic
1982-1987. pathway-based hierarchical clustering analysis
[313] Giunta B, Hou H, Zhu Y, Rrapo E, Tian J, Ta- of older adults with cognitive complaints and
kashi M, Commins D, Singer E, He J, Fernan- amnestic mild cognitive impairment using clin-
dez F and Tan J. HIV-1 Tat contributes to Al- ical and neuroimaging phenotypes. Am J Med
zheimers disease-like pathology in PSAPP Genet B Neuropsychiatr Genet 2010; 153B:
mice. Int J Clin Exp Pathol 2009; 2: 433-443. 1060-1069.
[314] Hickman SE, Allison EK and El Khoury J. Mi- [323] Manolio TA, Collins FS, Cox NJ, Goldstein DB,
croglial dysfunction and defective beta-amy- Hindorff LA, Hunter DJ, McCarthy MI, Ramos
loid clearance pathways in aging Alzheimers EM, Cardon LR, Chakravarti A, Cho JH, Guttm-
disease mice. J Neurosci 2008; 28: 8354- acher AE, Kong A, Kruglyak L, Mardis E, Rotimi
8360. CN, Slatkin M, Valle D, Whittemore AS, Boehn-
[315] Tarassishin L, Loudig O, Bauman A, Shafit-Za- ke M, Clark AG, Eichler EE, Gibson G, Haines
gardo B, Suh HS and Lee SC. Interferon regula- JL, Mackay TF, McCarroll SA and Visscher PM.
tory factor 3 inhibits astrocyte inflammatory Finding the missing heritability of complex dis-
gene expression through suppression of the eases. Nature 2009; 461: 747-753.
proinflammatory miR-155 and miR-155*. Glia [324] Zuk O, Hechter E, Sunyaev SR and Lander ES.
2011; 59: 1911-1922. The mystery of missing heritability: Genetic in-
[316] Chakrabarty P, Ceballos-Diaz C, Lin WL, Bec- teractions create phantom heritability. Proc
card A, Jansen-West K, McFarland NR, Janus Natl Acad Sci U S A 2012; 109: 1193-1198.
C, Dickson D, Das P and Golde TE. Interferon- [325] Peregrin-Alvarez JM, Sanford C and Parkinson
gamma induces progressive nigrostriatal de- J. The conservation and evolutionary modular-
generation and basal ganglia calcification. Nat ity of metabolism. Genome Biol 2009; 10:
Neurosci 2011; 14: 694-696. R63.
[317] Soares HD, Potter WZ, Pickering E, Kuhn M, Im- [326] Ayalew M, Le-Niculescu H, Levey DF, Jain N,
mermann FW, Shera DM, Ferm M, Dean RA, Changala B, Patel SD, Winiger E, Breier A,
Simon AJ, Swenson F, Siuciak JA, Kaplow J, Th- Shekhar A, Amdur R, Koller D, Nurnberger JI,
ambisetty M, Zagouras P, Koroshetz WJ, Wan Corvin A, Geyer M, Tsuang MT, Salomon D,
HI, Trojanowski JQ, Shaw LM; Biomarkers Con- Schork NJ, Fanous AH, ODonovan MC and
sortium Alzheimers Disease Plasma Pro- Niculescu AB. Convergent functional genomics
teomics Project. Plasma biomarkers associat- of schizophrenia: from comprehensive under-
ed with the apolipoprotein E genotype and standing to genetic risk prediction. Mol Psy-
Alzheimer disease. Arch Neurol 2012; 69: chiatry 2012; 17: 887-905.
1310-1317. [327] Zhong H, Yang X, Kaplan LM, Molony C and
[318] Barcia C, Ros CM, Annese V, Gomez A, Ros- Schadt EE. Integrating pathway analysis and
Bernal F, Aguado-Yera D, Martinez-Pagan ME, genetics of gene expression for genome-wide
de Pablos V, Fernandez-Villalba E and Herrero association studies. Am J Hum Genet 2010;
MT. IFN-gamma signaling, with the synergistic 86: 581-591.
contribution of TNF-alpha, mediates cell spe- [328] Ala-Korpela M, Kangas AJ and Inouye M.
cific microglial and astroglial activation in ex- Genome-wide association studies and sys-
perimental models of Parkinsons disease. tems biology: together at last. Trends Genet
Cell Death Dis 2011; 2: e142. 2011; 27: 493-498.

175 Am J Neurodegener Dis 2013;2(3):145-175

You might also like