Download Molecular and morphological analyses of basal forebrain

Document related concepts

Holonomic brain theory wikipedia , lookup

Limbic system wikipedia , lookup

Development of the nervous system wikipedia , lookup

Feature detection (nervous system) wikipedia , lookup

Nervous system network models wikipedia , lookup

Cognitive neuroscience wikipedia , lookup

Synaptic gating wikipedia , lookup

Nerve growth factor wikipedia , lookup

Signal transduction wikipedia , lookup

Neuromuscular junction wikipedia , lookup

Activity-dependent plasticity wikipedia , lookup

Haemodynamic response wikipedia , lookup

Synaptogenesis wikipedia , lookup

Environmental enrichment wikipedia , lookup

Neuroanatomy wikipedia , lookup

De novo protein synthesis theory of memory formation wikipedia , lookup

Metastability in the brain wikipedia , lookup

Epigenetics in learning and memory wikipedia , lookup

Endocannabinoid system wikipedia , lookup

Neurogenomics wikipedia , lookup

Aging brain wikipedia , lookup

Molecular neuroscience wikipedia , lookup

Channelrhodopsin wikipedia , lookup

Alzheimer's disease wikipedia , lookup

Optogenetics wikipedia , lookup

Neuropsychopharmacology wikipedia , lookup

Clinical neurochemistry wikipedia , lookup

Biochemistry of Alzheimer's disease wikipedia , lookup

Transcript
Boston University
OpenBU
http://open.bu.edu
Theses & Dissertations
Boston University Theses & Dissertations
2013
Molecular and morphological
analyses of basal forebrain
cholinergic neurons in mouse
models of aging and Alzheimer's
disease
Norman, Timothy Alfred Jr.
Boston University
http://hdl.handle.net/2144/12173
Boston University
BOSTON UNIVERSITY
SCHOOL OF MEDICINE
Thesis
MOLECULAR AND MORPHOLOGICAL ANALYSES OF BASAL FOREBRAIN
CHOLINERGIC NEURONS IN MOUSE MODELS OF AGING AND
ALZHEIMER’S DISEASE
By
TIMOTHY ALFRED NORMAN JR.
B.S., Biology, University of North Carolina at Wilmington, 2005
Submitted in partial fulfillment of the
requirements for the degree of
Master of Arts
2013
Approved by
First Reader
_____________________________________________
Jan Krzysztof Blusztajn, Ph.D.
Professor, Pathology and Laboratory Medicine
Second Reader
_____________________________________________
Chris Andry, Ph.D.
Associate Professor, Pathology and Laboratory Medicine
ACKNOWLEDGEMENTS
Many sincere thanks to my PI and mentor, Dr. Jan Blusztajn for the
opportunity to conduct research in his laboratory. Through his excellent
instruction and example, I am confident in my ability to become an independent
scientist and make future discoveries and contributions to the scientific
community. I would also like to acknowledge Dr. Blusztajn and Dr. Chris Andry
for their advice, support, guidance and encouragement throughout my master’s
research and didactic studies.
A special thanks to Dr. Nader Rahimi for inspiring vision and emphasizing
critical thinking and for going the extra mile to challenge his students. These
lessons will undoubtedly aid me in my future endeavors.
Most certainly without Dr. Tarik Haydar and Dr. William Tyler, Joe
Goodliffe and Nadine Aziz, my experiments would not have been possible. They
generously provided technical assistance with confocal imaging, as well as with
the Volocity data analysis software. For this I am extremely grateful.
Last but not least, I would like to sincerely thank my best friend and wife,
Laura Neubauer-Norman. She has proven her love, her loyalty and her patience
in supporting me over these last nine years. Thanks for believing in me and I
enjoy each day that we get to discover life together.
I wish you all the best that life has to offer.
iii
MOLECULAR AND MORPHOLOGICAL ANALYSES OF BASAL FOREBRAIN
CHOLINERGIC NEURONS IN MOUSE MODELS OF AGING AND
ALZHEIMER’S DISEASE
TIMOTHY ALFRED. NORMAN JR.
Boston University School of Medicine, 2013
Major Professor: Jan Krzysztof Blusztajn, PhD., Professor of Pathology
and Laboratory Medicine
ABSTRACT
Basal forebrain cholinergic neurons (BFCNs) of the medial septal nuclei,
the diagonal bands of Broca and the nucleus basalis magnocellularis synthesize
acetylcholine (ACh) and their projections extend to the cerebral cortex,
hippocampus and the amygdala. ACh neurotransmission is essential for
learning, attention, memory, arousal and sleep. BFCNs are dependent on a
regulated neurochemical environment for the induction, development, maturation
and maintenance of their phenotype and viability. However, events that
compromise this neurochemical environment can contribute to BFCN dysfunction
and/or degeneration, decreased ACh levels and disrupted brain function. During
normal aging and Alzheimer’s disease (AD) BFCNs become more vulnerable to
dysfunction due to trophic factor withdrawal, cell signaling impairments and other
cytopathologic changes. AD is characterized by the deposition of Amyloid-beta
(Aβ) plaques and neurofibrillary Tau tangles (NFTs) in the cortex and
hippocampus. These pathological AD hallmarks overlap with cortical and
iv
hippocampal cholinergic dysfunction, implicating both as the drivers of cognitive
and behavioral decline associated with AD. Since, BFCNs are highly vulnerable
to AD pathophysiology, factors that support the BFCN phenotype may have
practical use in preserving neuronal networks and cognitive function.
There is now strong evidence that bone morphogenetic protein-9 (BMP9
also known as growth/differentiating factor 2, GDF2) acts as an induction and
maintenance factor that regulates BFCN differentiation in-vitro and in-vivo. Here
we used transgenic mice that express green fluorescent protein (GFP) in BFCNs
to make observations concerning the BFCN phenotype in aging and AD. We
found qualitative evidence that BFCNs of the 24-month old WT/ChAT-GFP mice
were smaller and more rounded with shorter processes when compared to 6month old mouse BFCNs. We analyzed the effects of intracerebroventricular
infusion of BMP9 on BFCN projection fibers in the APPswe/PS1dE9 mouse
model of AD using laser scanning confocal microscopy. BMP9 not only
increased the density of cholinergic projection fibers in the hippocampus of wild
type and AD model APPswe/PS1dE9 mice, but it also reduced the plaque burden
in the hippocampus of the AD mouse model. These data indicate that BMP9
ameliorated two major pathophysiologic hallmarks of AD, observable in these
transgenic APPswe/PS1dE9 mice. BMP9 reduced Aβ plaque burden in this AD
model, and enhanced the outgrowth and viability of cholinergic fibers within the
hippocampus of both wild-type and APPswe/PS1dE9 mice.
v
Prior studies in our laboratory identified 300 genes differentially expressed
in the BFCNs purified from APPswe/PS1dE9 mice as compared to their wild type
littermates. One of these, the cellular prion protein (PrPc), displayed a 2.8-fold
increase in mRNA expression in APPswe/PS1dE9 mice over wild-type controls.
Our analysis of PrPc expression by quantitative immunohistochemistry revealed a
46% increase in the number of BFCNs expressing PrPc protein medial septum
and vertical and horizontal limbs of the diagonal band of Broca (p=0.0007), but
not in the striatum of the APPswe/PS1dE9 model of AD. Recently, studies have
shown that direct binding of PrPc is required for Aβ neurotoxicity, synaptic
impairment and cognitive dysfunction. Based on our results as well as others, it
is possible that high PrPc expression in the BFCNs could be responsible for at
least part of the cholinergic deficits observed in AD. Studies have also shown
that PrPc is a receptor for Aβ and that this Aβ-PrPc interaction leads to pathologic
tau phosphorylation and toxicity providing evidence that all three major
hypotheses of AD: the Aβ-hypothesis, the tau hypothesis and the cholinergic
hypothesis converge toward PrPc.
vi
PREFACE
The brain is composed of billions of highly organized glial cells and
neurons that receive, integrate, store and export information to interconnected
subpopulations of neurons. It is the command center of the organism that
processes a wide variety of sensorimotor stimuli and higher executive functions
such as cognition, memory, and emotions that govern behavior. Brain regions
that control these functions and the subpopulations of neurons residing therein
can be distinguished by their molecular and morphological characteristics.
These phenotypic traits are established by specific gene expression profiles
which direct the differentiation and maturation of individual neuronal subclasses.
For example, basal forebrain cholinergic neurons can be distinguished by a
specific structural morphology and molecular phenotype. The functional extent of
the cholinergic system can be observed in many diverse locales throughout the
vertebrate organism including the central nervous, cardiovascular, digestive,
respiratory and motor systems. Thus a comprehensive understanding of the
cholinergic network is essential in understanding organismal biology. Here, this
study focuses on the basal forebrain cholinergic neurons and their fibers located
in an area of central nervous system long known to be affected by AD. Initially, I
will synthesize a comprehensive background review on the developmental,
anatomical and molecular phenotype of cholinergic neurons in the basal
forebrain. Results pertaining to the molecular and morphological analyses of
BFCNs as a function of aging and AD-like pathology, were obtained using a
vii
combination of immunohistochemistry and confocal microscopic techniques. If
these initial discoveries are consistent and repeatable, they may contribute to the
development and implementation of a safe and useful intervention for AD.
viii
TABLE OF CONTENTS
Title Page
i
Approval Page
ii
Acknowledgements
iii
Abstract
iv
Preface
vii
List of Figures
xi
List of Abbreviations
xii
Introduction
1
BFCN Development
3
BMP9 Regulates the BFCN Phenotype
7
BFCN Phenotype
12
Nerve Growth Factor and its Receptors
19
Alzheimer’s Disease
26
Aβ Hypothesis of AD
31
Aβ Targeted Therapeutics
32
Aβ Disrupts Multiple Networks in AD
34
Tau Hypothesis of AD
37
Cholinergic Hypothesis of AD
39
Cholinergic Therapeutics
42
Materials and Methods
44
Transgenic mice
44
ix
Tissue Processing
45
Immunofluorescence
46
Confocal Microscopy
48
Image Analysis
48
Results
50
BMP9 Improves AD-like Pathology In-vivo
50
BFCN Gene Expression in APP/CHGFP mice
58
PrPc is Upregulated in BFCNs of APP/CHGFP mice
65
Discussion
70
List of Journal Abbreviations
79
References
80
Curriculum Vita
91
x
LIST OF FIGURES
Figure 1. Factors influencing BFCN development
10
Figure 2. Basal forebrain and cholinergic projection anatomy
11
Figure 3. Cholinergic gene locus
13
Figure 4. BFCN morphological changes in aged WT/CHGFP mice
17
Figure 5. BFCN morphological changes in APP/CHGFP mice
18
Figure 6. p75/NTR and ChATGFP expression in MSN/VDB
21
Figure 7. Quantification of p75/NTR and ChAT colocalization in WT/CHGFP
25
Figure 8. PET scan image of Aβ in AD brain
29
Figure 9. Aβ disrupts multiple networks in the brain
35
Figure 10. Amyloidosis and Cholinergic deficit in APP/CHGFP
41
Figure 11. Abnormal BFCN morphology in APP/CHGFP
42
Figure 12. BMP9 infusion increases HPC cholinergic innervation
52
Figure 13. BMP9 infusion increases CA1 and hDG cholinergic innervation
53
Figure 14. BMP9 may increase cholinergic branching and extention
56
Figure 15. BMP9 infusion improves the cholinergic network in HPC
57
Figure 16. FACS purification of BFCNs
60
Figure 17. Microarray cluster analysis of WT and APP.PS1 BFCNs
63
Figure 18. Differential mRNA expression in WT and APP.PS1 BFCNs
64
Figure 19. Orthoslice analysis of PrPc/CHGFP colocalization method
69
Figure 20. PrPc is upregulated in APP/CHGFP BFCNs of MSN/VDB/HDB
73
Figure 21. PrPc at the center of AD
77
xi
LIST OF ABBREVIATIONS
Aβ
Amyloid-Beta
ACh
Acetylcholine
AD
Alzheimer’s disease
APP
Amyloid Precursor Protein
BACE
Beta-Amyloid Converting Enzyme
BFCN
Basal Forebrain Cholinergic Neurons
BLA
Basolateral Amygdala
BMP9
Bone Morphogenetic Protein 9
BSA
Bovine Serum Albumin
CA
Cornu Ammonis
CGL
Cholinergic Gene Locus
ChAT
Choline Acetyltransferase
ChT
Choline Transporter 1
CNS
Central Nervous System
CTX
Cortex
FGF
Fibroblast Growth Factor
GABA
Gamma-Aminobutyric Acid
hDG
(hilus of) Dentate Gyrus
hESC
(human) Embryonic Stem Cells
DMSO
Dimethyl Sulfoxide
eGFP
enhanced Green Fluorescent Protein
xii
FACS
Flourescence Activated Cell Sorting
HDB
Horizontal Limb of the Diagonal Band of Broca
HPC
Hippocampus
ICV
Intracerebroventricular
LGE
Lateral Ganglionic Eminence
MGE
Medial Ganglionic Eminence
mRNA
(messenger) Ribonucleic Acid
MSN
Medial Septal Nucleus
NBM
Nucleus Basalis of Meynert
NGF
Nerve Growth Factor
NT-3
Neurotrophin 3
OB
Olfactory Bulb
PBS
Phosphate Buffered Saline
PS1
Presenilin 1
pAB
Polyclonal Antibody
POA
Preoptic Area
PrPc
Cellular Prion Protein
PrPsc
Scrapie Prion Protein
RE
Response Element
SHH
Sonic Hedgehog
SR
Stratum Radiatum Moleculare (of the Hippocampus)
STR
Striatum
xiii
VAChT
Vesicular Acetylcholine Transporter
VDB
Vertical Limb of the Diagonal Band of Broca
WT
Wild-Type
xiv
INTRODUCTION
The neuroanatomical development of the basal forebrain cholinergic
nuclei and their projection pathways result from highly orchestrated
morphogenetic, cell-to-cell and autocrine/paracrine signaling mechanisms (Dale
et al., 1997; Sussel et al., 1999). As a result, cholinergic neurons in the basal
forebrain differentiate and project their axon terminals to the hippocampus and
cerebral cortex where they release acetylcholine to modulate neuronal activity.
Thus the development, maturation and maintenance of the cholinergic system is
critical for the processes of attention, learning and memory.
AD leads to dysfunction and degeneration of multiple neuronal subclasses
in specific brain regions by disrupting normally functioning molecular, cellular,
local circuits and neuronal networks, including the basal forebrain cholinergic
network (Palop and Mucke, 2010).
Three AD hypotheses have been described and each additively
contributes to the pathological hallmarks of AD: 1) Aβ deposition, 2)
neurofibrillary tau tangle formation and 3) the cholinergic deficits.
Accumulating evidence implicates the cellular prion protein, PrPc as a
factor that promotes AD pathophysiology and that all three hypotheses of AD
converge at PrPc. We provide evidence that PrPc is upregulated specifically in
BFCNs which may increase their neuronal susceptibility during AD pathological
progression.
1
Since BFCNs are essential for memory and cognitive function, exogenous
application of factors that regulate BFCN development may prove useful in
generating BFCNs from pluripotent stem cells or by promoting their viability and
physiological function in brain regions affected by AD. In support of these ideas,
we provide strong in-vivo evidence that BMP9, a factor that promotes the
cholinergic neuronal phenotype, can ameliorate two pathophysiological hallmarks
of AD by reducing Aβ plaques and by increasing cholinergic density in the
hippocampus.
These data describe an important concept in potential AD therapies by
focusing on factors that influence multiple aspects of AD pathology. Given the
genetic, environmental and pathophysiological complexities of AD it is highly
unlikely that there is one magic bullet to slow, stop or prevent the progressive
nature of the disease. Therapeutic candidates should include those factors with
potential to reduce more than one AD pathophysiological mechanism. Since
PrPc interacts with Aβ, is expressed by BFCNs and may lead to tau
phosphorylation targeting this protein may ultimately improve AD from three
angles. Likewise, by increasing cholinergic innervation and decreasing plaque
load in the hippocampus, BMP9 was shown to improve AD-like pathology in-vivo
and highlights the utility of this developmental regulator of the BFCNs as a
potential AD therapy.
2
BFCN DEVELOPMENT
BFCN origination and development begins with regional specification of
the early telencephalon, proliferation and differentiation is then initiated through
induction, maintenance and maturation signaling mechanisms. These highly
conserved patterns of vertebrate telencephalic development are established by
morphogenetic gradients regulated by growth factors and cytokines which modify
gene transcription to determine body axes, regional boundaries and the
differentiation of forebrain cell types (Shimamura et al., 1995; Rubenstein and
Beachy, 1998). In general, current developmental models are based on the idea
that neuronal populations arise from spatio-temporally regulated gene expression
programs fine-tuned by concentration-dependent patterns or signaling ‘pulses’
within the neurochemical niche. Competent embryonic tissues are organized into
distinct molecular domains to establish transverse and longitudinal borders along
morphogenetic gradients. The establishment of the caudal-rostral, dorsalventral, medial-lateral axes by distinct morphological and molecular boundaries
creates an organized “grid-like” prosomeric model of forebrain development
(Rubenstein et al., 1994). This grid-like patterning of forebrain anatomical
structure is evolutionarily conserved across multiple species of organisms
including lower mammals, i.e., mice to more complex non-human primates and
humans from embryogenesis into adult forebrain development (Wedeen et al.,
2012).
3
The axis of patterning in the rostrobasal telencephalon gives rise to
subpallial proliferative zones of the medial ganglionic eminence (MGE), lateral
ganglionic eminence (LGE) and the preoptic area (POA) which contribute to
nearly all forebrain structures such as the cortex, striatum, hippocampus,
amygdala and septum (Marin et al., 2000; Gelman et al., 2009; Nobrega-Pereira
et al., 2010). Migratory cells from each of these three embryonic proliferative
zones are further classified into septal progenitor domains, SE1-6 (Flames et al.,
2007), which develop into the heterogeneously populated neuronal nuclei located
in the basal forebrain, in which there are several cholinergic nuclei present.
Basal forebrain cholinergic neurogenesis in mice occurs between embryonic
days (E)11-18 along a caudo-rostral gradient, i.e. the more caudolaterally located
nucleus basalis of meynert (NBM) arises prior to the more rostromedial diagonal
bands of Broca (DBB) and the medial septum (MS) (Schwab et al., 1988) (Brady
et al., 1989; Schambra et al., 1989). Interestingly, some of the first studies
identified that the progressive cholinergic degeneration in AD began in the
nucleus basalis of Meynert (NBM) and followed along the same caudorostral
gradient (Doucette and Ball, 1987).
Before the proliferative MGE, LGE, POA areas arise, the early forebrain
developmental pathway is initiated by prechordal mesoderm induction of ventral
midline cells (rostral diencephalon) by cooperative signaling through contactmediated Sonic hedgehog (SHH) and diffusible bone morphogenetic protein 7
(BMP7) gradients. These morphogenic gradients establish the rostro-ventral
4
regionalization of the prospective forebrain around the 10-12 somite stage. At
this time, Nkx2.1 is expressed in progenitor cells of the MGE in the developing
basal telencephalon and anti-BMP7 antibody blocks normal Nkx2.1 expression in
the MGE (Dale et al., 1997).
Nkx2.1 is a homeobox transcription factor whose expression is spatially
restricted to the rostrobasal forebrain during development (Price et al., 1992) and
Nkx2.1 repression in the developing dorsal telencephalon specifies ventral
forebrain development (Sussel et al., 1999). In Nkx2.1 mutant mice, Sussel et al,
observed structural and cytochemical abnormalities in the MGE induced by
disrupted progenitor cell production and migration around E12.5. Of major
importance, by E18.5 the Nkx2.1 mutants displayed a complete loss of TrkApositive cells, an early cholinergic neuronal marker (Sobreviela et al., 1994)
throughout the entire basal forebrain (Sussel et al., 1999). From these studies it
is apparent that normal Nkx2.1 expression within a highly regulated spatiotemporal manner is essential for all cells originating and migrating through the
MGE, including BFCNs. In contrast to other MGE progenitor cells, BFCN Nkx2.1
expression is maintained and can be observed in postmitotic BFCNs into
adulthood (Wei et al., 2012). Other functions ascribed to Nkx2.1 pertain to
neuronal connectivity and migration of cells from the basal forebrain to the
neocortex and striatum; however the role of Nkx2.1 in adult BFCNs has not been
elucidated.
5
Whereas Nkx2.1 expression leads to regional specification of the MGE
and initiates neuronal progenitor subtypes, LIM-homeobox (Lhx) genes are
transcription factors which are expressed in parallel with Nkx2.1 in the
developing basal forebrain (Wanaka et al., 1997). Mice expressing mutant Lhx8
(LIM-homeobox gene Lhx8/L3/Lhx7) also lacked cholinergic neurons in the
telencephalic areas of the striatum, septum, and diagonal bands, but not in the
nucleus accumbens or the magnocellular preoptic nucleus (Zhao et al., 2003).
This demonstrates some heterogeneity in the cholinergic induction pathways and
presents evidence for alternate mechanisms for cholinergic specification.
Importantly, the loss of functional Lhx8 at E13.5 specifically blocked the
formation of BFCNs in these areas without affecting other neuronal types. When
quantified, mutant Lhx8 resulted in ~75% reduction in cholinergic cell counts in
the telencephalon and a significant loss of hippocampal cholinergic inputs.
Subsequent studies by Mori confirmed that Lhx8 plays a pivotal role in the
development and maintenance of BFCNs by targeting the expression of choline
acetyltransferase (ChAT), vesicular acetylcholine transporter (VAChT) and the
75kD/low affinity NGF receptor (p75/NTR), all of which are biochemical markers
of the cholinergic phenotype (Mori et al., 2004).
6
BMP9 REGULATES THE BFCN PHENOTYPE
Bone morphogenetic proteins are known to play a major role in body axis
patterning during embryogenesis. BMP9 is abundantly expressed in the
embryonic basal forebrain and spinal cord at E14 (Lopez-Coviella et al., 2000)
and BMP9 induces Lhx8 expression (Bissonnette et al., 2011). The complete
mechanism from Nkx2.1 to BMP9 induction of Lhx8 remains to be determined.
The temporal expression of BMP9 and its receptor ALK1, have been shown to
overlap with the critical period for BFCN differentiation. E14 primary septal
neurons transiently treated with BMP9 undergo dynamic morphological and
molecular changes in vitro (Lopez-Coviella et al., 2000). Instead of growing
uniform monolayers, BMP9 treated septal cells organized into clusters and
extended long TuJ1-positive projections. These same neurons were also
induced by BMP9 to express ChAT, VAChT and p75/NTR and displayed a 20fold increase in ACh production over PBS treated controls (Lopez-Coviella et al.,
2000). Induction of the BFCN transcriptome and cholinergic phenotype by BMP9
supports the idea that it acts as the differentiating factor responsible for the
induction and maintenance of BFCNs (Lopez-Coviella et al., 2000; LopezCoviella et al., 2005). Moreover, intracerebroventricular (icv) infusion of BMP9 in
mice with a unilateral septohippocampal lesion preserved the ChAT expression
and prevented most of the cholinergic defects due to reduced hippocampal ACh
levels. BMP9 also caused increased expression of nerve growth factor (NGF)
7
and its receptors, p75/NTR and TrkA in the hippocampus ipsilateral to the lesion.
BMP9 activates signaling pathways via serine-threonine kinase BMP receptors
which phosphorylate Smad1/5/8 proteins. BMP9 was identified to be
developmentally regulated in parallel with various types of BMP receptors whose
expression were also tightly regulated between E14 and perinatal time points in
the mouse basal forebrain (Lopez-Coviella et al., 2006). BmpR-Ib and Smad1
proteins were highly expressed during E14-18 and BmpR-II protein levels
increased around postnatal day 1 (P1) which were maintained into adulthood.
Messenger RNA levels for another BMP receptor, Alk1 increased from E17 into
adulthood and ALK1 was proposed as the maintenance receptor for BFCNs after
differentiation. In E14 cultured basal forebrain cells, Smad1/5 phosphorylation
was quickly induced after BMP9 treatment, which also increased their
translocation to the nucleus to complex with Smad4 for up to 1 hr. This
Smad1/5-Smad4 complex leads to activation of specific target genes (Massague
and Wotton, 2000). Once in the nucleus, gene targets of the TGF-β/BMP signal
transduction require several coregulators for transcription initiation. Smadinteracting zinc finger protein, Sizn1, is a coregulator of BFCN differentiation at
the transcription level. Immunoprecipitation experiments showed that Sizn1
directly interacts with Smad1 and recruits transcriptional machinery such as
CBP/p300 to increase expression from the cholinergic gene locus coding for
ChAT and VAChT. Knockdown of Sizn1 in E13.5 primary septal cell cultures
decreased ChAT and VAChT expression after BMP2 treatment (Cho et al.,
8
2008). Not only does this emphasize the ability of Sizn1 to activate the
cholinergic gene locus but it also suggests overlapping signaling mechanisms
between BMP2 and BMP9 to induce cholinergic phenotypic expression. Indeed,
BMP2 treatment of E14 septal cell cultures increased ACh production 15-fold,
whereas BMP9 increased ACh production 20-fold as compared to PBS controls
(Lopez-Coviella et al., 2002). Given that BMP9 promotes Smad1/5 activation
and nuclear translocation with Smad4, along with other undiscovered
comodulators it is reasonable to hypothesize that Sizn1 may also mediate BMP9
induced transcription from the cholinergic gene locus.
These in vitro and in vivo experiments demonstrate that BMP signaling
acts as a differentiation and maintenance pathway for the BFCN phenotype.
Elucidation and confirmation of these developmental pathways has enabled the
in vitro differentiation, maturation, transplantation and functional integration of
BFCNs derived from pluripotent stem cells. After stimulation with fibroblast
growth factor 8 (FGF8) and SHH to produce neuronally committed progenitors
competent to BMP9 treatment, human embryonic stem cells (hESCs) were
differentiated into BFCNs in-vitro, grafted into ex-vivo mouse brain slices and
then confirmed to be electrophysiologically active with voltage-clamp recordings
(Bissonnette et al., 2011). Exogenously applied BMP9 induced the BFCN
phenotype and a dramatic increase in Lhx8 and Gbx1 mRNA transcripts were
observed (Bissonnette et al.). Overexpression of these two transcription factors
was more efficient (94% vs. 85%) than BMP9 at producing BFCNs from hESCs.
9
Progenitor State
BFCN Differentiation
BMP9
E10
Lhx8
Gbx1
Nkx2 .1
?
Maturation
BFCN
E18
Smad1/5
E14
?
NGF
ChAT
VAChT
75/NTR
TrkA
Figure 1 BFCN differentiation from Nkx 2.1 expressing progenitors in the MGE and
LGE at E10, to early BFCN induction and specification at E14. The morphogenic
gradient induces transcription of BFCN cholinergic markers, ChAT, VAChT and the
NGF/neurotrophin receptors at E18 which is maintained into adulthood.
This is possibly due to BMP9 mediated regulatory feedback loops to modulate
its own inductive signaling capacity in conjunction with Lhx8/Gbx1 transcriptional
activation. Furthermore, BMP9 treatment with Lhx8 knockdown by siRNA
abrogated BFCN differentiation, providing further evidence for BMP9
downstream signaling through Lhx8/Gbx1 to activate transcription of the
cholinergic gene locus (Bissonnette et al., 2011). These results confirm and
strengthen the role of BMP9 mediated transcriptional activation as a sufficient
event for BFCN development and advances the idea that BFCN generation from
a patient’s own stem cells may hold therapeutic potential for diseases specifically
affecting this neuronal subclass.
10
CTX
CA1
HPC
CA2
F
CA3
OB
MSN
VDB
DG
BLA
HDB
NBM
A
CTX
CA1
HPC
HPC
CTX
CA2
CA3
F
MSN
BLA
BLA
VDB
HDB
B
HDB
NBM
Figure 2 Sagittal (A, top panel) and coronal (B, lower panel) views with basal
forebrain nuclei labeled in white and projection targets in yellow and red arrows
indicating BFCN projection paths. Modified from Mouse Brain Explorer 2, Allen
Brain Institute.
11
BFCN PHENOTYPE
In 1933, the neurophysiologist and Nobel Laureate, H.H. Dale proposed
the term “cholinergic” for those neural pathways that utilize the neurotransmitter
ACh. These pathways are illustrated in the atlas presented in Figure 2. The
biochemical basis of the cholinergic system requires macromolecules to mediate
ACh synthesis, release, synaptic transmission, degradation and choline
reuptake. In the cytoplasm, choline acetyltransferase (ChAT) forms ACh from
the precursors, choline and acetyl-CoA (Bielarczyk and Szutowicz, 1989) (Jope
and Jenden, 1980) which is essential for survival since transgenic ChAT
knockout mice do not survive past postnatal day 1. After ACh synthesis, BFCNs
utilize vesicular acetylcholine transporter (VAChT) to sequester ACh into quanta
or vesicles to be released from synapses and effect postsynaptic electrochemical
signals. Eliminating ACh release in the hippocampus by genetically reducing
VAChT results in hyperactivity, impaired long-term potentiation and visuospatial
memory (Martyn et al., 2012).
ChAT and VAChT represent a unique “gene within a gene” organization
on human chromosome10q11 establishing the cholinergic gene locus (CGL)
(Eiden, 1998) illustrated in Figure 3. VAChT is located within the first intron of
the ChAT gene, allowing coordinate regulation of this gene locus whose
organization is conserved through C. elegans, Drosophila and H.sapiens.
Coordinate regulation does not necessarily imply coordinate expression. The
12
effects of both CNTF/LIF and retinoic acid receptor-α (RAR-α) stimulation by alltrans-retinoic acid led to proportional increases in ChAT and VAChT mRNA
expression, whereas a 2- and 4-fold increase,respectively was observed in a
septal cell line after cyclic-AMP treatment (Berse and Blusztajn, 1995).
Figure 3 The cholinergic gene locus (CGL) codes for ChAT (its central and
peripheral variants) as well as VAChT from chromosome 10. The CGL consists
of 3 non-coding (R, N, M) and 15-16 coding exons depending on the species.
(*) represent transcriptional start sites for the 5 alternative ChAT variants. The
intronless VAChT gene is located within the first intron, between the first two
non-coding exons of the full-length ChAT reading frame.
13
Differential expression patterns from a common locus could be accomplished by
multiple response elements (RE) in the cholinergic gene locus such as a BMPRE, cAMP-RE, repressor complexes or additional interactions that regulate
transcription through epigenetic modification. REST4, a transcriptional activator,
can de-repress the gene silencing action of the REST/CoREST complex and
activate the cholinergic gene locus in a protein kinase A (PKA) dependent
manner (Hersh and Shimojo, 2003). This may explain the coordinate regulation,
but differential expression in ChAT and VAChT mRNA when the septal cells were
treated with PKA activating cAMP (Berse and Blusztajn, 1995). Epigenetic
modifications may promote regulation of expression from the cholinergic gene
locus as histone deacetylase 9 (HDAC9), a class IIa enzyme that removes an
acetyl group from the histone N-terminal tail and represses the ChAT promoter in
neuronal cultures (Aizawa et al., 2012). Multiple layers of complex regulatory
mechanisms govern ChAT and VAChT expression from the cholinergic gene
locus, to establish the biochemical basis of all properly functioning cholinergic
neurons, however the exact mechanisms for the differential expression patterns
of ChAT and VAChT from a common locus remains elusive.
Postsynaptic molecules such as ionotropic, nicotinic acetylcholine
receptors (nAChR), metabotropic muscarinic acetylcholine receptors (mAChR),
the hydrolytic enzyme, acetylcholinesterase (AChE) control the electrochemical
14
neurotransmission signals provoked by ACh release. Each of these factors is a
salient feature of acetylcholine neurotransmission.
VAChT, a twelve membrane domain transporter protein, is localized in the
nerve terminals where it transports ACh into synaptic vesicles. Presynaptic
excitation of a cholinergic neuron induces calcium-dependent exocytosis of ACh
in the synapse (Li et al., 1995a).
Exocytosis of ACh is mediated by a series of highly-regulated docking,
synaptic membrane-vesicular fusion and lastly, vesicular recycling mechanisms
(Sudhof, 1995). Postsynaptic nAChR and mAChRs, as well as mAChR
autoreceptors located presynaptically can bind ACh (Blusztajn and Berse, 2000)
to initiate temporally-defined patterns of intracellular signaling and gene
expression pathways. AChE, a carboxylesterase capable of hydrolyzing 36,000
molecules of ACh into free acetate and choline per second, effectively terminates
the neurotransmitter signal. The choline transporter 1 (ChT1) is responsible for
the presynaptic, sodium-dependent, high-affinity choline reuptake which recycles
choline for another round of ChAT catalyzed ACh synthesis and VAChT
sequestration (Eiden, 1998; Okuda et al., 2000).
These macromolecules modulate the cholinergic effects to optimize neural
circuits throughout the entire brain. ACh is important for mechanisms of synaptic
plasticity, which is activity-dependent strengthening or weakening of synapses,
thought to be responsible for learning acquisition, memory storage and retrieval,
and long term neural circuit maintenance (Picciotto et al., 2012).
15
Both choline and acetyl-CoA are essential for cellular metabolic pathways
and the production of acetylcholine in BFCNs. Age induced metabolic stress and
this double utilization of acetyl-CoA and choline in cholinergic neurons may
increase BFCN susceptibility to dysfunction and degeneration leading to
cognitive and memory impairments (Perry et al., 1980; Wurtman, 1992)
(Szutowicz et al., 1996).
As choline can be metabolized from phosphatidylcholine, a major
component of the plasma membrane, diseases that result in cholinergic
dysfunction may destabilize the plasma membrane and cytoskeletal dynamics to
cause morphological changes in BFCNs. Our initial observations in 6- and 24month old mice indeed revealed morphological changes with age in BFCNs. We
observed retracted arborizations as cell bodies appeared more rounded and
smaller in size, as illustrated in Figure 4. With AD-like amyloidosis, BFCNs in AD
model mice appear smaller and have tortuous, stunted projections as illustrated
in Figure 5 and representative of the cholinergic defect the APP/CHGFP mice.
These morphological changes in wild type (WT/CHGFP) mice with age and in AD
model (APP/CHGFP) mice are in line with previous reports and highlights the
susceptibility of BFCNs to morphological changes with aging and AD.
16
24 month
6 month
WT/CHGFP
Figure 4 Age-induced morphological changes of BFCNs in the
MSN/VDB of 6 months (top) and 24 months old (bottom) WT/CHGFP
mice (20x objective). Note: Aged BFCNs have shorter processes
and have taken on a smaller, more spherical shape in contrast to the
ellipsoid somas and long projections evident in the 6-month old
BFCNs.
17
APP/CHGFP
8 months
WT/CHGFP
Figure 5 Reduced BFCN cell size and stunted axonal projections in
the MSN of the APP/CHGFP AD mouse model expressing eGFP from
the ChAT promoter (40x, objective lens).
18
NERVE GROWTH FACTOR AND ITS RECEPTORS
Discovery of nerve growth factor (NGF), the first growth factor, by Rita LeviMontalcini in the 1950s transformed modern neuroscience and earned Dr. LeviMontalcini the 1986 Nobel Prize. Initially, NGF was shown to regulate cell
growth, differentiation and survival of noradrenergic neurons. Later it was
learned that NGF’s mechanism of action influenced multiple neuronal cell
populations in the CNS and PNS (Aloe, 2004). High concentrations of NGF in
the terminal cholinergic targets of the cerebral cortex and hippocampus
implicated an NGF-cholinergic association (Johnson et al., 1987). Co-expression
of the NGF receptors, TrkA and p75/NTR with ChAT is a characteristic only
observed in BFCNs and demonstrates a reliance on NGF signaling in the basal
forebrain (Sobreviela et al., 1994).
NGF and its receptors are now known to be important for the development and
maintenance of BFCNs and NGF has been shown by multiple groups to increase
ACh levels (Berse et al., 1999; Auld et al., 2001), ChAT (Gnahn et al., 1983) ,
VAChT (Berse et al., 1999), and TrkA expression (Li et al., 1995b). NGF is also
induced by BMP9 in dissociated E14 primary septal cell cultures (Schnitzler et
al., 2010). These data suggest that both NGF and BMP9 have overlapping
functions to support BFCN development, survival and phenotypic maintenance
throughout neurodevelopment into adulthood. In aged rats (26-30 month old),
impairments to BFCN NGF retrograde transport resulted in a 60% reduction in
19
cholinergic cell size and a 43% decline in TrkA mRNA leading the authors to
propose this as a mechanism for cholinergic hypofunction and degeneration in
aging and AD (Cooper et al., 1994).
Furthermore, triple colocalization of p75/NTR and TrkA NGF receptors
with ChAT is restricted to the cholinergic neurons of the basal forebrain and their
respective projection sites in the CNS (Sobreviela et al., 1994) making these
molecules markers of the BFCN network. Sobreviela (Sobreviela et al., 1994)
performed an extensive quantitative analysis of TrkA and p75/NTR distributions
in the CNS and found virtually all (>95%) TrkA+ neurons in the MS/DBB were
positive for p75/NTR and ChAT with slightly less colocalization of all three BFCN
markers in the NBM (~85%).
TrkA, a type-I receptor tyrosine kinase, has high-affinity for the processed,
mature form of NGF and initiates cell signaling cascades, receptor mediated
endocytosis and retrograde transport of NGF to the BFCN soma for survival and
maturation of BFCNs and maintenance of their target innervation (Fagan et al.,
1997; Moises et al., 2007; Niewiadomska et al., 2011). Conversely, p75/NTR is
a promiscuous neurotrophin receptor that has a lower affinity for NGF and
prefers to bind immature proNGF when acting unilaterally (Sofroniew et al.,
2001). More specifically, loss of NGF signaling in TrkAnull mice produces
significantly smaller and fewer BFCNs (Fagan et al., 1997); whereas deletion of
p75/NTR in mice results in increased BFCN counts and cholinergic fiber
hyperinnervation of hippocampal targets (Yeo et al., 1997). At baseline,
20
p75/NTR is expressed in a 10:1 ratio with TrkA and the vulnerability of BFCNs to
changes in TrkA/NGF signaling may mediate neuronal shrinkage, dysfunction
Figure 6 Co-expression of transgenic ChAT-eGFP (green) and endogenous
p75/NTR (red) in the MSN/VDB provides a positive control validating eGFP
expression in ChAT/p75-positive BFCNs.
Note: Compare the anatomical
features of MS/VDB in this figure to the atlas in Figure 2B.
21
or death, as in aging and AD when TrkA:p75/NTR expression ratios are altered
(Chao and Hempstead, 1995). TrkA downregulation coupled with impaired
retrograde axonal transport in the basal forebrain is one of the early findings in
AD and correlates significantly with lower cognitive performance (Mufson et al.,
1996).
p75/NTR has demonstrated pro-survival capabilities by increasing the
specificity of TrkA, as well as TrkB and TrkC neurotrophin receptors for their
respective growth factors i.e, NGF, BDNF and NT-3 respectively (Kaplan and
Miller, 1997). In the absence of ligand, co-immunoprecipitation experiments
have demonstrated binding between the p75/NTR and TrkA (Huber and Chao,
1995) which required both the intact extracellular and intracellular domains of
both proteins (Bibel et al., 1999). The p75/NTR-TrkA co- receptor increased
NGF responsiveness and neuronal survivability (Chao and Hempstead, 1995).
Much research regarding BFCN phenotypic changes and receptor
expression levels during aging and AD has been carried out, however these
studies have not been entirely consistent. In particular, studies on the role of
p75/NTR in AD have been controversial with research showing both
neuroprotective and neurotoxic effects. p75/NTR has a highly conserved death
domain which in certain contexts are known to induce apoptosis. However, in
cholinergic neurons p75/NTR downregulates cholinergic markers but does not
induce apparent cholinergic cell death (Barrett, 2000). In mice with p75/NTR
functional deletion mutation, Greferath (Greferath et al., 2012) found the number
22
and size of ChAT expressing BFCNs increased but the number of p75/NTRpositive neurons in the basal forebrain was unchanged suggesting a
nonapoptotic, antineurotrophic role for p75/NTR.
Earlier human studies observed a 38% and 43% reductions in p75/NTR
expressing neurons in the basal forebrains of humans with mild cognitive
impairment and mild AD, respectively (Mufson et al., 2002). The latter found that
the reductions of p75/NTR were significantly correlated with cognitive
impairment, as determined by the Mini-Mental State Exam and the Global
Cognitive Test.
These conflicting reports are not surprising, given the complexities
surrounding the physiological role of p75/NTR receptor itself. proNGF-p75/NTR
can cause caspase activation and lead to apoptosis. Not only can p75/NTR
undergo proteolytic cleavage by the Beta-Amyloid Converting Enzyme (BACE)
(Zampieri et al., 2005), but it can also promote cleavage of APP to generate
insoluble Aβ (Costantini et al., 2005). p75/NTR also binds to Aβ in primary
hippocampal cultures (Kuner et al., 1998) and intrahippocampal Aβ injections
were dependent on p75/NTR expression in vivo to produce neurotoxic effects
(Sotthibundhu et al., 2008).
In contrast, there are multiple lines of evidence suggesting that p75/NTR
may mediate anti-Aβ aggregation and pro-survival functions. BACE cleavage of
the p75/NTR extracellular domain (ECD) can confer protection from neurotoxic
amyloid oligomers. Zhang (Zhang et al., 2003) found a dose-dependent
23
upregulation of p75/NTR in primary human neurons exposed to Aβ 1-40 and Aβ1-42.
This correlated with increased neuronal survivability in a PI3K-Akt dependent
mechanism. Transfecting the same cells with a plasmid encoding anti-sensep75/NTR effectively reduced p75/NTR levels and sensitized these neurons to
Aβ1-42 cytotoxicity (Zhang et al., 2003). Another study by Wang et al, showed
that p75/NTR increased Aβ production and deposition in the APPswe/PS1dE9
mouse model of AD, but inhibited Aβ aggregation with its extracellular domain
(ECD). Injection of the p75/NTR ECD into the hippocampus of p75/NTRnullAPPswe/PS1dE9 mice significantly reduced the area fraction of Aβ plaques in
these mice (Wang et al., 2011). Wang et al, also found elevated insoluble Aβ in
sera and less Aβ in the brains of p75/NTR+/+ mice, as compared to p75/NTRnullAPPswe/PS1dE9 mice, suggesting that p75/NTR may prevent Aβ aggregation
and increase Aβ clearance.
p75/NTR expression is consistently increased by BMP9 in-vitro and in-vivo
(Lopez-Coviella et al., 2000; Lopez-Coviella et al., 2005; Schnitzler et al., 2008b).
Since BMP9 is important for BFCN development, maturation and maintenance, it
would be worthwhile to determine whether BMP9 modulates the enzymatic
proteolysis of p75/NTR ECD toward the more advantageous, neuroprotective
pathway during AD pathology.
24
Figure 7 Colocalization of transgenic ChAT-eGFP and p75/NTR expression
in the VDB. Costes Colocalization Coefficient (M1): 0.999; Costes
Colocalization Coefficient (M2): 0.957.
These colocalization coefficients
calculated by Volocity software for the representative image support the
findings of (Sobreviela et al., 1994). Note the morphology and the
overlapping densities of the p75/NTR and ChAT fiber networks.
25
ALZHEIMER’S DISEASE
AD is a complex, neurodegenerative disease that robs humans of their
most valuable, personal possession: their mind. AD is diagnosed by the
presence of two histopathologic hallmarks: Aβ plaques and neurofibrillary tau
tangles. Major brain regions affected by AD are the neocortex, the medial
temporal lobe and adjacent limbic structures including the hippocampus and
amygdala, as well as the cerebellum. Atrophy and dysfunction in these regions
which are responsible for learning, attention, memory, insight, judgment,
orientation, emotion and other higher executive abilities lead to global loss of
cognitive ability. Included in this global cognitive impairment are memory loss,
dementia, psychosis, depression, aggression/agitation and other behavioral
detriments.
AD is a disorder with a strong genetic component and is divided into
familial and sporadic forms.. Approximately 25% of AD is the familial form and
the rest fall under sporadic. Late-onset AD (LOAD) is the most common variety
of sporadic AD and typically affects those 65 years and older. Sporadic AD is
thought to arise due to age-induced exposures and accumulation of
environmental risk factors, however very little is known about the etiology of
sporadic AD (Bird, 1993). Recently, genome wide association studies have
found a strong genetic component leading to increased risk of early-onset,
familial AD as well as late-onset (sporadic AD) (Guerreiro et al., 2013).
26
The familial form of AD is much less common and is associated with early
onset AD (EOAD) in people less than 65 years old. Familial AD displays an
autosomal dominant pattern of inheritance. The genetic variants in the genes
coding for APP, PSEN1 and PSEN2 are deterministically associated with earlyonset AD. Late onset-AD is associated with specific genetic variants in the
genes coding for TREM2, MTHFD1L, BCHE, CR1, CLU, BIN1, PICALM, MS4A,
CD2AP, EPHA1, ABCA7 and CD33. However, aging and inheritance of the
apolipoprotein ApoE4 allele have the largest effect of AD risk (Naj et al., 2011;
Guerreiro et al., 2013).
27
AD GENETIC
VARIANT
TYPE OF AD
DISEASE IMPLICATION or
GENE FUNCTION
APP, PSEN1/PSEN2
Deterministic, Early-onset
AD
Increased production of toxic Aβ
intermediates and plaque formation
APOE4
Late-onset AD
Defective lipid and amyloid
clearance
TREM2
Late-onset AD
Defective microglial phagocytosis
EPHA1
Late-onset AD
Axon guidance and ADAM10
metalloprotease substrate
recognition (OMIM: 179610)
CD2AP
Late-onset AD
Vesicle formation and cytoskeletal
polarization
(OMIM: 604421)
CR1
Late-onset AD
Complement receptor and activator
of innate immune response (OMIM:
120620)
CLU
Late-onset AD
Clearance of cellular debris and
apoptosis (OMIM: 185430)
BIN1
Late-onset AD
Tumor suppressor that interacts
with phospholipases
MTHFD1L
Late-onset AD
Conversion of methionine from
homocysteine
BCHE
Potential association with
Early and Late-onset AD
Hydrolysis of ACh and correlates to
plaque burden.
TABLE 1 Selected genetic variants identified as risk factors for early-onset
and late onset AD. Gene function listed instead of pathologic contribution
since most genetic variants are only associated with AD risk and have not
yet been causally linked to AD pathophysiology.
28
Diagnosing Alzheimer’s disease has been challenging because the cognitive
dysfunction, memory loss and behavioral changes occur many years after the
onset of disease progression (Niedowicz et al., 2012). Until recently, a
confirmatory diagnosis of AD was only available after postmortem autopsy
analysis for Aβ plaques and neurofibrillary tangles. Advances in PET imaging
and biomarker discovery may enable early diagnosis and the initiation of
preventative or symptomatic treatments of for humans with preclinical AD.
Figure 8 Positron Emission Tomography (PET) scan images of a cognitively
normal brain compared to an AD brain.
Aβ plaques are radiolabeled with
Pittsburgh compound B (PiB) which is now used to help diagnose AD before
symptoms are evident.
Higher PiB concentration (red and yellow regions)
indicates increased presence of Aβ plaques. Image from Alzheimer’s Disease
Education and Referral center, National institute of Aging.
29
Research into the genetic components of AD has allowed the generation
of transgenic mice to study certain parameters of the disease pathophysiology invivo. Although not a complete recapitulation of human AD pathology, these mice
can be engineered as classical knockouts, transgenics or knock-ins to test
certain AD genes and gene-gene interactions. Most AD mouse models focus on
APP and Aβ plaque formation, however double-, triple-, on up to 5x- transgenic
mice have been produced to study narrow or broad aspects of AD
pathophysiology. These mice exhibit reactive gliosis and inflammation,
dystrophic neurites and cognitive deficits which are also observed in human AD,
but the mice lack significant brain atrophy (Mineur et al., 2005; Oakley et al.,
2006). Our studies involve the APPswe/PS1dE9 mouse model of AD engineered
to express mutated human forms of APP and PSEN1 which are known to be
deterministic of AD in humans (see Materials and Methods for more info on
APPswe/PS1dE9 mice). Previous studies have found these mice display
progressive amyloidosis with a significant cholinergic deficit (Figures 5, 10, 11
and 15) and cognitive impairments (Savonenko et al., 2005) (Gimbel et al.,
2010).
30
Aβ HYPOTHESIS OF AD
AD is a progressive disease characterized by memory loss and cognitive
impairment exacerbated by Aβ-induced synaptic failure (Selkoe, 2002). There is
now a large body of evidence implicating Aβ as the molecular pathogenic culprit
of AD, which has culminated into the Aβ hypothesis. Aβ monomers, oligomers,
fibrils and plaques generated from insoluble fragments of the amyloid precursor
protein (APP) cleavage have been extensively investigated and it is widely
accepted that Aβ deposition disrupts molecular, cellular and neuronal network
dynamics. The Aβ hypothesis provides a mechanistic explanation for progressive
AD and according to Walsh (Walsh and Selkoe, 2004) is supported by the
following observations: 1) progressively increased Aβ peptide deposition around
neuritic and dystrophic neurites in the cortex and hippocampus, 2) synthetic Aβ
peptides display in vitro and in vivo toxicity, 3) inherited APP mutations results in
higher production of amyloidogenic Aβ , 4) transgenic animals with APP and
presenilin PSEN1/PSEN2 mutations generate greater levels of the more toxic
Aβ42 peptide, resulting in more severe, rapid onset AD-like pathology, and 5)
strong correlations have been identified between Aβ oligomers, cognitive deficits
and synaptic retraction.
31
Aβ TARGETED THERAPEUTICS
The Aβ-hypothesis has opened the possibility of immunotherapies for
inhibiting amyloidosis in the brains of AD patients. This was first attempted in
humans in a clinical trial of intravenously infused bapineuzumab which reduced
Aβ burden and tau aggregation, but it failed to demonstrate any improvements in
cognition or activities of daily living (Lobello et al., 2012).
It is difficult to interpret these results due to the late initiation of treatment
in the disease process as plaque accumulation and synaptic degeneration is
known to occur decades before symptoms of cognitive dysfunction appear. To
address this issue, the very first AD prevention trial was approved by the NIH and
will systematically test Genentech’s anti-Aβ antibody crenezumab in a presymptomatic Columbian cohort (n=300) harboring the PSEN1 (E280A) mutation.
A smaller group of pre-symptomatic American participants with other presenilin
mutations from the DIAN cohort will also receive crenezumab (Selkoe, 2012).
The outcome of this preventative study will likely represent the largest and most
authoritative challenge to the Aβ-hypothesis.
One reason Aβ has garnered so much attention is because the molecular
underpinnings of APP catabolism and generation of neurotoxic Aβ is the most
understood process in AD pathophysiology. In addition, understanding the
pathogenic pathways and molecular regulation of Aβ generation from APP
32
provides multiple targets for prevention and/or treatment. APP is sequentially
cleaved by BACE and gamma-secretase to generate Aβ40 and Aβ42 fragments.
Pharmacological approaches developed by Eli Lilly, using the gamma-secretase
inhibitor semagacestat (LY450139) to prevent Aβ generation showed significant
dose-dependent reductions of Aβ40 in human CNS over 12 hours (Bateman et al.,
2009), however Phase III clinical trials were abruptly ended as results indicated
worse cognitive performance compared to placebo. Further complications
included gastrointestinal irritability and increased risks of skin cancer due to
interference of signaling by Notch – a known substrate for gamma-secretase
(Imbimbo and Giardina, 2011). Elan Pharmaceuticals has since developed and
tested ELN475516, a gamma secretase inhibitor that specifically inhibits
cleavage of APP and reduces Aβ40 without interfering with Notch signaling in
mice (Basi et al., 2010). The safety or effectiveness of ELN475516 remains to
be tested in humans.
Inhibiting Aβ generation is more appealing than blocking fibrillary plaque
formation because the smaller Aβ oligomers are thought to be more toxic to
synapses (Lue et al., 1999) and Aβ oligomers are a better determinant of
neurodegeneration and cognitive decline than Aβ plaques (McLean et al., 1999).
By decreasing the number of plaques without increasing the clearance rate of Aβ
oligomers, a potential therapy may increase the number of free oligomers (or
dimers and trimers). This might actually induce more harm than good since Aβ
33
oligomers are more hydrophobic and are known to interfere with CNS physiology
at multiple levels.
Aβ DISRUPTS MULTIPLE NETWORKS IN AD
Olzscha et al. (Olzscha et al., 2011) have proposed that amyloid-like
aggregates induce a “fatal network collapse” by affecting key cellular processes
such as chromatin remodeling, transcription, vesicular transport, signaling and
cytoskeletal dynamics. In addition to synaptic retraction and degeneration,
multiple groups have shown that Aβ interferes with AMPA-R, NMDA-R, nACh,
mACh and mGlu5 receptors at synapses. This would disrupt intracellular
functions as well as local circuit connections and ultimately larger neuronal
networks (Palop and Mucke, 2010). As mentioned earlier, Aβ oligomers have
been shown to bind p75/NTR, nAChRs, mAChRs and PrPc all of which are
expressed pre- or post-synaptically as a part of the cholinergic network. This
suggests that the Aβ-hypothesis and cholinergic hypothesis of AD may not be
mutually exclusive.
34
Figure 9 Aβ disrupts multiple functional networks at the molecular,
synaptic, local circuits and neuronal network levels.
Additionally, Aβ has been found to bind, sequester and negatively affect
intracellular transport by inducing tau hyperphosphorylation via α7-nAChRs,
mitochondrial release of cytochrome C to activate apoptotic cascades, and
initiating the stress-induced unfolded protein response in the ER. Aβ possesses
nanomolar affinity for the PrPc protein and this interaction was essential to
mediate Aβ-induced toxicity and cognitive impairment (Lauren et al., 2009).
However, less specific interactions with other proteins may be propagated
through the disordered, unstructured domains allowing Aβ to evolve an aberrant
35
interactome interfering with essential function and inducing toxicity (Bolognesi et
al., 2010). Aβ contains a hydrophilic, extracellular domain and a hydrophobic,
intramembrane domain which allows Aβ monomers to act as nucleating centers
to create expanded oligomers. These hydrophilic-hydrophobic regions establish
structural disorder within Aβ to induce aggregation of individual monomers into βsheet rich oligomers, fibrils and plaques. These pathogenic Aβ units then
interact and sequester numerous proteins with essential functions disrupting a
host of physiological functions (Olzscha et al., 2011). This amyloid induced fatal
network collapse in molecular and cellular networks can be extrapolated to larger
neuronal networks that require interconnected brain regions for appropriate
social, emotional and behavioral integration. As these interconnections suffer
synaptic failure and neurodegeneration, network efficiencies in the brain become
compromised then cognitive and behavioral impairments ensue.
36
TAU HYPOTHESIS OF AD
Tau (also known as, MAPT) is a member of microtubule associated
protein (MAP) family. Tau provides structural integrity and stability to
microtubules. In the CNS, microtubules influence neuronal morphology, dendritic
scaffolding and intracellular transport of nutrients, neurotransmitters and
organelles such as mitochondria and rough endoplasmic reticulum. These
processes are essential for cytoskeletal maintenance, synaptic plasticity and cell
viability (Savelieff et al., 2013). Various mechanisms resulting in tau
hyperphosphorylation induces its dissociation from microtubules yielding “seeds”
of intracellular tau monomers to form oligomers, granules and neurofibrillary
tangles (NFTs) (Takashima, 2010). Phosphorylated tau is also resistant to both
proteolytic cleavage by CAPN2 a calcium-activated neutral protease, as well as
polyubiquitin mediated proteosomal degradation (Iqbal et al., 2009). These
conditions lead to increased intracellular concentrations of the pathologic tau
intermediates which form the NFTs.
NFTs can be observed in the frontal and temporal lobes of the neocortex,
the entorhinal cortex as well as limbic structures of the AD brain and are
correlative of synaptic and neuronal loss (Takashima, 2010) (Braak and Braak,
1991). Evidence for the tau hypothesis of AD include: 1) NFTs, in the absence
of amyloid plaques, are found in brains afflicted with other dementia-related
pathologies (Ward et al., 2012), 2) Neurodegeneration correlates with phospho-
37
tau/NFT levels but not amyloid plaques (Iqbal et al., 2009) (de Souza et al.,
2012), 3) Normal, aged individuals may contain Aβ plaque burdens as high as
AD patients, but lack tau-positive dystrophic neurites and neuritic plaques
(Dickson et al., 1992), 4) overexpression of the P301S human tau mutation in
transgenic mice induces cortical, hippocampal and amydalar NFT formation,
inflammatory gliosis and neurodegeneration (Yoshiyama et al., 2007).
The extent of NFTs in the brain strongly correlate with neurodegeneration
and cognitive impairment in AD (Braak and Braak, 1991). Using NFT
immunostaining, Braak and Braak, identified six different stages of AD pathology
as defined by the amount of NFT involvement in postmortem human brain
samples. Braak and Braak (BB) stages I/II include NFTs confined to the
transentorhinal area, BB stages III/IV include NFTs in the hippocampus and other
limbic regions and BB stages V/VI refer to extensive NFTs throughout neocortical
regions.
Tau can be phosphorylated on 38 serine/threonine residues by multiple
kinases including glycogen synthase kinase-3 (GSK-3), cyclin-dependent kinase5 (cdk5), protein kinase A (PKA), the extracellular regulated kinases (ERK1/2),
stress-activated protein kinases (SAPKs) casein kinase 1 (CK-1) and FYNkinase. Interestingly, a single nucleotide polymorphism (rs7768046) in the FYNkinase and (rs913275) in the PPP2R4 phosphatase are associated with
increased total-tau and phosphorylated tau in cerebrospinal fluid from AD
patients (Bekris et al., 2012).
38
Whereas multiple kinases facilitate tau hyperphosphorylation, a relatively
few number of phosphoprotein phosphatases are able to balance this pathologic
mechanism. Only PP-1, PP-2A and PP-2B have been shown to dephosphorylate
and inhibit tau’s self-assembly to preserve microtubule stability (Wang et al.,
1996). Significantly decreased phosphatase activity has been measured in AD
brains, suggestive of a dephosphorylation defect that contributes to the
phosphor-tau mediated pathophysiology of AD (Gong et al., 1993; Guadagna et
al., 2012). These studies may lead to tau-specific phosphatase activators or
kinase inhibitors to reduce tau hyperphosphorylation and preserve the neuronal
cytoskeleton, intracellular transport or possibly even cognitive function.
CHOLINERGIC HYPOTHESIS OF AD
The cholinergic hypothesis of AD proposed by Bartus (Bartus et al., 1982)
is supported by a vast body of research pertaining to: 1) alterations in choline
uptake, the rate-limiting step for ACh synthesis (Slotkin et al., 1990), 2) reduced
ChAT activity and ACh release, 3) reduced (α7)-nAChR expression (Hernandez
et al., 2010), 4) dysfunctional or reduced M1/M3-mAChR expression (Terry and
Buccafusco, 2003), and 5) impaired cholinergic axonal transport of NGF (Mufson
et al., 1996). Dystrophic cholinergic neurites, neuritic plaques and reduced
hippocampal and cortical cholinergic innervation as well as reduced BFCN
counts and synaptic degeneration have all been observed in human and
39
transgenic mouse models of AD (Mufson et al., 2003; Perez et al., 2007). One of
the earliest observed features discovered in postmortem AD brains is the
pronounced cholinergic degeneration which is correlated to severity of memory
loss and cognitive decline. After Davies (Davies and Maloney, 1976) reported a
selective loss of cholinergic neurons in AD patients, it was postulated that AD
may be a selective neurotransmitter disease affecting acetylcholine signaling.
Subsequent losses of cholinoceptive pyramidal neurons and even greater
synaptic degeneration in the cortex as measured by synaptophysin staining are
the strongest correlates of dementia and memory loss (Terry et al., 1991; Francis
et al., 1999).
Further support for the cholinergic hypothesis comes from studies
demonstrating various cognitive dysfunction and learning impairments in mice,
rats, monkeys and humans with basal forebrain lesions and pharmacological
cholinergic blockade (Terry and Buccafusco, 2003). Blocking cholinergic
signaling with the mAChR antagonists, scopolamine, atropine or glycopyrrolate
produce amnestic effects with age-related sensitivity whereas memory
improvements have been observed with specific nAChR and mAChR agonists
(Ray et al., 1992; Terry and Buccafusco, 2003).
40
Figure 10 Laser scanning confocal micrograph of Aβ42 immunostaining (red)
in the hippocampus (left) and cerebral cortex (right) in APP/CHGFP mice.
Note: dystrophic, tortuous neurites adjacent to the plaques and the weak,
punctate network of cholinergic fibers (green), (40x objective lens).
41
A
Figure 11
B
C
Abnormal cholinergic morphologies in the vertical limb of the
diagonal band (A), striatum (B) and cerebral cortex (C) of 8 month old
APP/CHGFP mice, (20x objective lens).
CHOLINERGIC THERAPEUTICS
The only approved therapeutic drugs for AD block AChE to inhibit ACh
hydrolysis thereby potentiating ACh neurotransmission at the synapse. The first
generation AChE inhibitor, tacrine showed significant improvement in cognition
and quality of life as measured by Alzheimer’s Disease Assessment Scalecognitive subscale (ADAS-cog) scores and Clinician Interview Based Impression
of Change plus caregiver input (CIBIC+) tests. However, given that ACh has
widespread modulatory effects in the gastrointestinal and cardiovascular
systems, multiple side effects involving gastrointestinal discomfort, nausea,
vomiting, cardiovascular complications and hepatotoxicity reduced long-term
42
patient compliance and the effectiveness of tacrine (Terry and Buccafusco,
2003).
Second generation AChE inhibitors such as donepezil have reduced side
effects while demonstrating similar improvements in cognitive function as well as
delaying reductions in activities of daily living (ADLs) by up to a year (Rogers et
al., 1998). Early to mild AD patients receiving donepezil demonstrated improved
quality of life, reduced amount of direct supervision and delayed
institutionalization. However not every AD patient responded equally to
donepezil treatment and none of the AChE inhibitors improve cognitive
performance to normal levels (Mesulam, 2004).
The cholinergic hypothesis merely explains the insidious loss of attention
and working memory during aging and AD (Schliebs and Arendt, 2011).
Potentiating cholinergic neurotransmission to improve cognition simply treats the
symptoms of AD and not the underlying cause, making it difficult, if not
impossible to pharmacologically counterbalance the cognitive assault of the
causative agent with AChE inhibitors or AChR agonists. The causative agent for
AD has remained elusive, however various cellular and molecular biological
approaches have yielded tremendous gains in the understanding of AD
pathophysiology. Multiple studies indicate that BFCN dysfunction and/or
degeneration contributes to the memory deficits in advanced aging and AD
(Mufson et al., 2008; Grothe et al., 2012; Haense et al., 2012).
43
Therefore, interventions that inhibit Aβ-deposition or tau-phosphorylation
while protecting BFCN viability may preserve ACh neurotransmission thus
preserving memory function and improving cognition.
MATERIALS AND METHODS
Transgenic Mice
The following strains were used:
1) AD model mice B6C3-Tg(APPswe,PSEN1dE9)85Dbo/Mmjax and
2) transgenic mice expressing the enhanced green fluorescent protein in
cholinergic cells: B6.Cg-Tg(RP23-268L19-EGFP)2Mik/J.
Briefly, the AD model mice express two separate transgenes that inserted into a
single locus, both under control of the prion promoter. The first transgene is the
murine APP protein harboring the human Aβ sequence with Swedish mutations,
K595N/M596L (APPswe), associated with the familial form of AD. The second
transgenic insert harbors the human presenilin 1 (PS1) protein with a deleted
ninth axon (PS1dE9), which was discovered in individuals with early-onset
familial AD. In order to study the cholinergic phenotypic changes during AD-like
progression in the APPswe/PS1dE9 model, these mice were crossed with
transgenic C57BL/6J mice expressing GFP under the control of the Chat
promoter. The triple transgenic offspring of APPswe/PS1dE9 (APP.PS1) x
CHAT-GFP (CHGFP) will be referred to throughout the paper as APP/CHGFP.
All mice were kept on a 12 hour light-dark cycle and given food and water ad
44
libitum. This study used 6- and 8-month-old AD/CHGFP and WT/CHGFP mice
for the microarray study and PrPc immunofluorescence, respectively. Genotypes
were confirmed with PCR primers specific for human APP. APP/PS1null
homozygous ChAT-GFP+/+ transgenic mice expressing GFP from the ChAT
promoter, were used as littermate controls for the AD study (WT/ChGFP).
Tthese WT/CHGFP mice were used separately for analysis of BFCNs in the 6month old to 24-month old aging profile.
Tissue Processing
Mice were euthanized with CO2 and decapitated. Brains were placed in an icecold metal brain mold containing 1 mm slots. The medial septum along with the
vertical and horizontal diagonal bands of Broca was isolated by cutting a 2 mm
section rostral to the optic chiasm. The remaining frontal cortex and olfactory
bulbs, rostral to the septal slice, were bisected down the midline and then
dissected to isolate each region. The remaining caudal tissue, was bisected
down the midline. The left olfactory bulb, left frontal cortex, left hippocampus
was placed into small eppendorf tubes on dry ice and then stored at -70°C for
biochemical analysis. The right olfactory bulb, right frontal cortex and right hemibrain were drop fixed in 4% paraformaldehyde, 75 mM lysine, 10 mM sodium
periodate; pH 7.4 solution for 24 hours at 4 °C and subsequently cryoprotected
in a 10% glycerol/2%DMSO solution in 0.1M phosphate buffered saline (PBS, pH
7.4) for 24 hours, afterward the tissues were placed into 20% glycerol/2%DMSO
45
in 0.1M PBS solution and stored at 4°C. The 2 mm forebrain slice containing the
basal forebrain and the right hemi-brain containing the hippocampus, were
serially sectioned in the coronal plane on a freeze sliding microtome at 40 μm.
Free floating sections were then stored in 0.02% sodium azide solution in 0.1M
PBS at 4°C.
Immunofluorescence
For Aβ42 immunofluorescence, free floating sections were removed from 0.02%
sodium azide/PBS storage solution and rinsed with PBS. First, the sections
were incubated for 3 hr in a blocking buffer solution consisting of 10% normal
donkey serum and 0.3% Triton X-100 in PBS. The sections were then incubated
overnight with an antibody buffer solution of 1% BSA, 0.3% Triton X-100 in PBS
supplemented with recombinant affinity-purified, primary rabbit anti-Aβ42
monoclonal antibody (Life Technologies, 1:2500; cat.# 700254). The next day,
sections were rinsed with PBS, blocked in the aforementioned blocking buffer for
3 hrs then incubated in the dark for 6 hrs in the antibody buffer solution
supplemented with secondary Alexa Fluor-594 donkey anti-rabbit IgG or donkey
anti-goat IgG antibody (Life Technologies,1:1000). After the final PBS rinse, the
sections were allowed to dry on SuperfrostPlus slides (Fisher) then coverslipped
with VectaShield and allowed to dry at room temperature in the dark, afterwards
the slides were stored at -20 °C. Each PBS rinse step consisted of 3-10 min
washes and all rinses and incubations were performed at room temperature on a
46
rotating shaker. No Aβ42 fluorescence was observed in wild-type mice or in
APP/PS1dE9 mice incubated with secondary antibody alone.
For PrPc and p75/NTR immunostaining, tissue sections were washed in PBS (pH
7.4), incubated in blocking buffer solution consisting of 1%BSA and 0.3% Triton
X-100 in PBS for two hours to reduce nonspecific binding. Goat anti-mouse PrPc
primary antibody (1:750, Abcam) or rabbit anti-mouse p75/NTR primary antibody
(Millipore, 1:500) were added to an antibody buffer solution of 10% appropriate
serum, 0.3% Triton X-100, 0.02% sodium azide in 0.1 M PBS and the sections
were incubated overnight at room temperature on a rotating shaker. Sections
were rinsed in PBS and blocked in the aforementioned blocking buffer for 2-3
hours. Sections were then incubated for 6 hr in the dark on a shaker at room
temperature, in the aforementioned antibody buffer solution containing secondary
donkey anti-rabbit IgG (H+L) or donkey anti-goat IgG (H+L) conjugated to
AlexaFluor-594 (1:1000, Molecular Probes). After 3x10 min PBS rinses, the
sections were incubated with DAPI (1:1000, Invitrogen) in 0.1M PBS for 10-15
minutes on a shaker in the dark then washed for 15 minutes in PBS. The
sections were placed on subbed SuperFrostPlus slides (Fisher) and allowed to
dry. Sections were then mounted with ProLong Gold (Invitrogen) or VectaShield
(Vector Labs) mounting media and coverslipped. Slides were allowed to harden
overnight at 4°C and then sealed with nail polish.
47
Confocal Microscopy
Imaging was performed on an LSM710 NLO laser scanning confocal microscope
(Zeiss) using Argon/Krypton and Helium/Neon lasers set to 488nm and 594nm,
respectively. Z-stack images of the entire MS, VDB and HDB were scanned.
Imaging parameters such as pinhole size, detectors, and laser intensity were
optimized and held constant in all experiments. Minor heterogeneities from
section to section were adjusted using gain and digital offset settings. To
minimize crosstalk and bleed-through during imaging, each fluorescent signal
was imaged in sequential scanning mode with an EC-Plan NeoFluar 40x/1.30 Oil
DIC M27 objective (Zeiss).
Image Analysis
PrPc Quantification
Three coronal sections from 8 female mice (WT; n=4, APP/CHGFP; n=4)
spanning the basal forebrain were selected for semiquantitative analyses of PrPc
expression in ChAT+ cell bodies. Images were coded and counted in a blinded
fashion. In Volocity 3D image analysis software (Perkin Elmer), the orthoslice
tool was used to determine the total number of BFCNs expression PrPc in the XZ and Y-Z dimension as shown in Figure 19. Statistical analysis performed with
SYSTAT by one-way ANOVA. Data are presented as mean ± SEM.
48
Hippocampal Cholinergic Fiber Quantification after BMP9 Treatment
All fluorescence imaging of cholinergic fibers and amyloid plaques was
performed on an LSM710 laser scanning confocal microscope (Zeiss) with an
EC-Plan NeoFluar 40x/1.30 Oil DIC M27 oil immersion objective at 1.2x optical
zoom for final magnification of 48x. Laser settings were optimized and held
constant as follows: laser intensity, 2% for Alexa-Fluor 488 and 594;
aperture/pinhole, 1.23AU; averaging speed, 4; scan speed, 6; image resolution,
512 x 512. For every mouse (n=29) belonging to one of four groups (APP/BMP,
n=7; APP/PBS, n=7; WT/BMP, n=8; WT/PBS, n=7) two coronal hippocampal
sections (1 anterior, 1 posterior) were imaged. A total of 263, 10μm z-stacks
taken at 1μm intervals were scanned in the hilus of the DG or the stratum
radiatum layer of CA1, CA2, and CA3. Images were coded by an individual
blinded to the experimental parameters and then transferred to Volocity® 3D
analysis software (PerkinElmer).
An empirically optimized batch protocol
function was established to: 1) find objects of interest using threshold intensities
between 20-255, and 2) exclude objects less than 0.3μm3 and greater than
100μm3.
The batch protocol was then automatically applied to all images and
the total object volume (in μm3) per image/per individual was then exported to an
Excel spreadsheet. After a human operator “data-validation” step to confirm
Volocity® object recognition [see (Piltti et al., 2011)], a total of 8 images (of 263)
required manual correction. Object volumes were accepted as an index of
cholinergic fibers present in the hippocampus. Statistical analysis of differences
49
in cholinergic fiber volumes between genotype- and BMP9 treatment groups was
performed with SigmaPlot software by two-way ANOVA. Data are expressed as
mean ± SEM.
RESULTS
The aim of these experiments was to test the utility of confocal microscopy
to image cholinergic neurons and cholinergic nerve fibers in transgenic mice
engineered to express enhanced green fluorescent protein in cholinergic cells.
These techniques were applied to studies of aged mice (Figure 4) as well as
mice engineered to develop the pathological changes associated with AD for the
purpose of understanding the molecular and morphological changes incurred by
the basal forebrain cholinergic system during these two processes. In order to
study the effects of BMP9 infusion on AD-like pathology, we studied animals at
10-months of age because they display significant amyloid deposition, reduced
ChAT activity and BFCN impairments exemplified by neuritic plaques and
dystrophic neurites in the cholinergic projection targets of the hippocampus and
cerebral cortex (Perez et al., 2007).
BMP9 INFUSION IMPROVES AD-LIKE PATHOLOGY IN VIVO
We imaged GFP-positive cholinergic fibers in the hippocampus using laser
scanning confocal microscopy and quantified z-stacks in the hippocampal areas
of the stratum radiatum moleculare of CA1, CA2, CA3 fields and in the hilus of
50
the dentate gyrus. This analysis revealed that BMP9 infusion significantly
increased cholinergic fiber densities in WT/CHGFP and APP/CHGFP mice for
the combined regions of interest through the hippocampus (Figure 12) and
separately within the CA1-stratum radiatum moleculare and hilus of the dentate
gyrus (Figures 13A & B). BMP9 infusion dramatically increased ChAT protein in
APP/PS1 mice above normal WT levels in the HPC as detected by ELISA and
also induced modest but significant increases of hippocampal NGF and NT-3
protein expression levels (Burke et. al, manuscript in preparation). These data
provide further evidence that BMP9 supports a neurochemical niche sufficient for
BFCN trophic support and may represent a therapy for AD patients by increasing
cholinergic neurotransmission to potentially maintain memory or cognitive
performance.
The observed increase in hippocampal cholinergic density could be
attributed to increased axonal branching of existing cholinergic neurons. One
interesting observation is that BMP9 increased NT-3 levels which has been
shown to increase synaptic connections during development by promoting
cholinergic branching and axonal extention to the hippocampus and cerebral
cortex (Robertson et al., 2006). A qualitative analysis of BMP9 vs. PBS infused
WT/CHGFP mice support the notion of increased cholinergic extentions and
possibly branching mediated by neurotrophin signaling possibly via NT-3 (Figure
14).
51
CA1
CA2
Cholinergic fiber volume, µm3
CA3
12000
PBS
BMP9
10000
8000
6000
4000
2000
0
Figure 12
hDG
WT/CHGFP
APP.PS1/CHGFP
Quantification of total hippocampal cholinergic fiber densities
within the stratum radiatum moleculare (SR) region of CA1, CA2, CA3 and in
the hilus of the dentate (hDG) gyrus among the four groups, expressed as
mean ± SEM.
52
Cholinergic fibers avoided Aβ42 plaques and in PBS-infused APP/CHGFP mice
neuritic plaques and dystrophic neurites were prominent as previously described
by (Perez et al., 2007). APP/CHGFP mice that received BMP9 infusion
displayed fewer dystrophic neurites adjacent to Aβ42 plaques, as depicted in
Figure 15 and there was an enhancement in the cholinergic fiber network in
these animals. Visualizaton of Aβ42 immunofluorescence at higher
magnifications revealed numerous, diffuse plaques as well as large plaques in
the hippocampus of PBS-infused mice while BMP9 infused mice contained
mostly large, dense plaques. As Aβ42 is more hydrophobic than Aβ40, it is
postulated that its oligomers are the most toxic Aβ species (Selkoe, 1997).
Figure 13
(Next two pages)
Confocal images from each of the four
experimental groups and a scatter plot (n=27) of the cholinergic densities in
the hilus of the dentate gyrus (A) and CA1/stratum radiatum (B) regions of
the hippocampus. This analysis revealed that BMP9 effectively increased
cholinergic innervation to the hippocampus of WT/CHGFP and APP/CHGFP
mice. Red bars are the mean for each group; scale bar, 50 μm.
53
Mean DG Fiber Counts
13A.
Mean Fiber Density in DG (μm3)
25000
20000
Y Data
15000
10000
5000
0
Wt/Pbs
Wt/Bmp
App/Pbs
App/Bmp
X Data
Group vs FIber Counts/Objects
BMP9
APP/PS1
WT
PBS
54
Mean CA1 Counts
Mean Fiber Density in CA1 (um3)
13B.
18000
16000
14000
Y Data
12000
10000
8000
6000
4000
2000
0
Wt/Pbs
Wt/Bmp
App/Pbs
App/Bmp
X Data
Group vs FIber Counts/Objects
BMP9
APP/PS1
WT
PBS
55
BMP9
WT/CHGFP
PBS
Figure 14 Cholinergic neurites in the granular layer of CA1
appeared longer in WT/CHGFP mice infused with BMP9 versus
PBS (40x).
56
PBS
Ab42
GFP
Ab42
GFP
BMP9
Figure 15 Confocal images of immunofluorescent staining of Aβ42 (red) in the
hippocampus also showing disrupted cholinergic network (green).
PBS-
infused (controls) contained weak, inconsistent fibers and more dystrophic
neurites (white arrows) whereas BMP9 infusion ameliorated these cholinergic
deficits in APP/CHGFP animals.
57
The apparent BMP9 mediated reductions in the number of smaller, diffuse
plaques may effectively reduce the total plaque surface area and minimize
disruptive Aβ42 interactions with proteins of essential function. The mechanism
by which BMP9 reduces Aβ42 plaque burden is a prospect for further research to
determine whether BMP9 reduces plaque deposition or increases plaque
clearance. The ability of BMP9 to induce exuberant cholinergic growth in the
hippocampus of an AD-like model compels future studies to determine if the
improved cholinergic network improves cognition in the APPswe/PS1dE9 mice.
This could be achieved using well characterized cognitive-behavioral assays, e.g.
the Barnes maze to test spatial memory capabilities of the mouse to locate a
dark escape box for safety, or the Morris water maze which also measures
visuospatial memory as a function of latent time to locate a platform and escape
from the stress of swimming.
These data demonstrate that administration of BMP9 ameliorates two
overt pathophysiological components of AD by reducing amyloid plaque burden
and by increasing the ChAT-positive cholinergic network in the hippocampus.
BFCN GENE EXPRESSION IN APP/CHGFP MICE
Of approximately 30,000 genes in the human genome, different cell types
express only an individual subset of these genes which permit their specification
and function. Cells that are selectively affected by disease, are known to
58
undergo homeostatic or pathologic changes in gene expression, a paradigm that
may provide diagnostic value or biomarker identification and/or medical targets of
therapy.
The molecular analysis of specific cell types in the brain is hampered by
the brain’s heterogeneity of densely packed neuronal subclasses and glial cells.
Furthermore, a given neuronal population within a specific brain region likely
represents a minority subset of cells within that region. Despite this, whole cell
populations dissociated from a resected area of the brain have enabled a variety
of in vitro studies of cell-function and cell-cell interactions but a thorough
transcriptomic analysis of a specific cell type remains challenging with this
methodology. Importantly, fluorescence-activated cell sorting (FACS) has been
used to identify subsets of cells in mixed populations. This technique has
provided major advances in cancer and immunology research but is underutilized
in studying the CNS. Originally, a method for the generation of healthy, purified
BFCN cultures by FACS using a fluorescent antibody against p75/NTR produced
cultures enriched in the cholinergic markers p75/NTR, ChAT and ChT (Schnitzler
et al., 2008a). This method has been simplified with the use of transgenic mice
expressing GFP from the ChAT promoter and can be a powerful tool for purifying
BFCNs to study their gene expression profiles in normal, aging and diseased
states.
59
Figure 16 FACS purified BFCNs from WT/ChAT-eGFP. Note: 3.7% of
cells from E18 mice septae express ChAT-eGFP and these cells have
enriched mRNA expression levels for BFCN markers; Chat, p75, Cht.
Loading control: Actb; beta-actin. Adapted from: A.C. Schnitzler et. al,
J. Neurosci. (2010).
60
To study the gene expression profile of BFCNs in the APPswe/PS1dE9, a mouse
model with AD-like pathology, the septal region was harvested from 6 month old
WT/CHGFP and APP/CHGFP mice expressing GFP from the ChAT promoter.
BFCNs constitute approximately 3-5% of the entire cell population in the septum
as indicated in Figure 16 and previous experiments.
FACS purified BFCN mRNA was extracted using the guanidiniumthiocyanate-phenol/chloroform method, after a cRNA amplification step and a
spiked standard curve was used to normalize RNA frequencies. Next, mRNA
reaction mixtures from WT/CHGFP and APP/CHGFP BFCNs obtained by FACS
were enriched in mRNA for the cholinergic markers p75/NTR, ChAT and ChT
and then subjected to Affymetrix Mouse Genome 430 2.0. These arrays were
stained with Streptavidin R-phyocoerythrin using the GeneChip Fluidics Station
400 and scanned with a Hewlett-Packard GeneArray Scanner. The data
collected was then analyzed by Microarray Suite 4.0 software.
This analysis identified some 300 differentially expressed genes in
WT/CHGFP and APP/CHGFP mice BFCNs which clearly formed separate
clusters as shown in Figure 17. Some of these genes as shown in Figure 18
have been previously implicated in the pathophysiology of AD and amyloidosis.
In particular one of these genes is LIMK1.
61
LIMK1 mRNA levels were reduced 50% below WT expression levels. LIMK1 is
cytoplasmic protein kinase regulating cytoskeletal structure and function and is
important during neurodevelopment by promoting axonal extention. It is very
interesting that LIMK1 has been shown to directly interact with the BMP type II
receptors to deregulate actin dynamics (Foletta et al., 2003). LIMK1 also
interacts with neuregulins which are known to be important for the development
of hippocampal-dependent visuospatial cognition (Wang et al., 1998).
Furthermore, inhibition of Aβ-induced LIMK1 phosphorylation of Cofilin with a
specific peptide competitor abrogated hippocampal neuronal dystrophy and
degeneration (Heredia et al., 2006).
In light of these data, our observation that LIMK1 mRNA is reduced in
BFCNs of AD-like mice may validate the use of this model to advance the study
of human AD, since as altered levels of phospho-LIMK1 have been reported in
human brain regions affected by AD (Heredia et al., 2006).
62
Figure 17 Gene expression analysis with
Affymetrix microarrays in FACS-purified
BFCN from 6 month old wild type (WT) and
APP.PS1
male
mice.
Graphical
representation of cluster analysis of 300
differentially-expressed genes is shown.
Note that the WT and APP.PS1 BFCN
genes
organize
clusters.
63
into
clearly
separate
mRNA levels, % of wild type
175
150
WT
APP.PS1
125
100
75
50
25
0
Limk1 Map2 Capn2
Il33
Notch3
FIGURE 18 Candidate genes with differential mRNA expression in BFCN purified
from 6-month old APP.PS1 male mice as compared with the WT littermates.
APP/PS1 mRNA expression levels in the graph above were normalized to WT
expression levels derived from Figure 17.
64
PrPc IS UPREGULATED IN BFCNs OF APP/CHGFP MICE
The mRNA coding for the cellular prion protein (PrPc) was observed to be
upregulated in BFCNs of APPswe/PS1dE9 mice. PrPc is a glycosyl
phosphatidylinositol (GPI) anchored-cell surface protein which localizes to lipid
rafts in the extracellular leaflet of the plasma membrane. It is expressed in the
peripheral tissues as well as in neuronal cell bodies and at post-synaptic
densities. In its misfolded form, PrPc is converted to amyloidogenic PrPSc and is
responsible for the fatal neurodegenerative disorders classified as transmissible
spongiform encephalopathies, including Creutzfeldt-Jakob disease, kuru and
Gersmann-Straussler-Scheinker syndrome (Prusiner and DeArmond, 1990)).
Also, a D178N mutation and homozygosity for methionine at PrPc codon 129 are
known to cause fatal familial insomnia (Montagna et al., 2003). Interestingly, a
single nucleotide polymorphism that causes a M129V mutation in PrP c has also
been associated with late-onset Alzheimer’s disease (Gunther and Strittmatter,
2010).
Much more is known about the pathogenic actions of PrPc however new
research is beginning to shed light onto the physiological roles of this protein.
PrPc functions as a copper-dependent NMDA-R ion channel regulator (You et al.,
2011), as a cell adhesion molecule by interacting with integrins, NCAM and
laminin, as reviewed in (Biasini et al., 2012) and as a. It has been argued that Aβ
oligomers are dependent on PrPc expression to mediate LTP inhibition and
65
synaptic dysfunction in pyramidal neurons of the hippocampus (Gimbel et al.,
2010), whereas the physiological and pathological function of PrPc expression
within BFCNs is largely unknown. Research has demonstrated two possible
ways that Aβ/PrPc interactions can cause excitotoxicity in neurons. First, Aβ
treatment of dissociated GABAergic and cholinergic forebrain neurons caused
reductions in calcium-activated potassium conductance in a PrPc dependent
fashion (Alier et al., 2011). This scenario could effectively reduce the refractory
period of membrane repolarization, extend action potentials and excitability and
facilitate BFCN excitotoxicity. Secondly, coimmunoprecipation of PrP c with
NR2D subunits of NMDA receptors suggests a direct interaction with this
glutamate activated non-selective cation channel. Hippocampal neurons treated
with copper chelators, Aβ42 or PrPc inactivation resulted in nondesensitized
NMDA-induced currents and neurotoxicity (You et al., 2011). This suggests that
PrPc may normally modulate ion channels concentrated at synapses whereas Aβ
binding to PrPc interferes with this essential regulatory function preventing
excitotoxicity. Interestingly, PrPc has been shown to undergo α- and γ-secretase
cleavage, similar to APP (Checler, 2012) and provides further evidence for
common mechanisms in neurodegenerative diseases.
Previous members in our lab enabled the microarray analysis of BFCN
mRNA (Schnitzler et al., 2008a). This analysis implicated PrPc as a gene
displaying a dysregulated expression profile in the APPswe/PS1dE9 mouse
66
model of AD. BFCNs from 6-month old APP/CHGFP mice exhibited a 2.8-fold
increase in PrPc mRNA levels as compared to WT/CHGFP BFCNs (p = 0.02).
In light of this, we sought to determine the percentage of CHATGFPpositive BFCNs expressing PrPc protein using semi-quantitative
immunofluorescence and laser scanning confocal microscopy. After
immunofluorescence and imaging, the Volocity software orthoslice tool was used
to quantify the total number of of cholinergic neurons in the basal forebrain and in
the striatum that coexpressed PrPc as illustrated in Figure 19.
The striatum (STR) is a forebrain region associated with planning,
anticipating and modulating pathways that control movement and procedural
memory. The striatum is comprised of medium spiny neurons (96%), Dieter’
neurons (2%), parvalbumin-, calretinin-, somatostatin-positive GABAergic
interneurons as well as cholinergic interneurons (1%). Cholinergic interneurons in
the striatum did not demonstrate any major difference in PrPc protein expression
among APP/CHGFP as compared to WT/CHGFP mice. At older ages, the
APPswe/PS1dE9 mice develop overt cholinergic deficits and Aβ-plaque
deposition the striatum (Perez et al., 2007). Consistent with these reports, we
did not see any amyloidosis in the striatum of these 8 month old mice, however
signs of cellular blebbing and dystrophic cholinergic neurons in the striatum were
observed at this time point. This result suggests that cholinergic degeneration in
the striatum may precede Aβ deposition which points to trophic withdrawal,
67
excitotoxicity or other uncharacterized mechanisms as the cause of cholinergic
dysfunction.
Consistent with other studies in mice and in humans, immunofluorescence
for Aβ plaques did not reveal any staining in the MSN, VDB and HDB. However,
the percentage of BFCNs expressing PrPc in these regions revealed a 46%
increase in PrPc protein expression as shown in Figure 20. The functional
significance of increased PrPc expression in APPswe/PS1dE9 BFCNs remains to
be determined although it provides fertile ground to study the role of PrP c as a
possible pathogenic factor mediating cholinergic dysfunction in AD.
68
Figure 19
An orthoslice screenshot from the quantitative analysis
procedure used to determine PrPc and ChATGFP overlap. Sections
through the basal forebrain cholinergic nuclei and striatum were
immunostained for PrPc (red). 3D Z-stack images were imported to a
Volocity library and analyzed in the X, Y, and Z planes using the
orthoslice tool.
This analysis enabled counting of total number of
neurons positive for both ChATGFP and PrPc in both the MS/VDB/HDB
and the STR areas (40x objective).
69
DISCUSSION
These results support our underlying hypotheses that as a result of aging
and AD,that BFCNs acquire altered morphological features, and BFCNs in AD
display a differential gene-protein expression profile. Secondly, we hypothesized
that BMP9, an induction factor sufficient for BFCN differentiation in-vitro and invivo, would support BFCN maintenance would promote BFCN growth and
survivability, in a model of AD characterized by heavy Aβ-amyloidosis and a
pronounced cholinergic deficit.
Our data reveal that a 7-day BMP9 infusion induced an exuberant
improvement in the cholinergic network in the hippocampus of WT/CHGFP and
APP/CHGFP mice. We detected a quantitative increase in cholinergic fiber
density within the DG (hilus) and the stratum radiatum layers of CA1, CA2 and
CA3 with BMP9 infusion. Consistent with previous reports, BMP9 also increased
expression of the NGF receptors, p75/NTR and TrkA (data not shown; Burke et.
al, manuscript in preparation). BMP9 induced the BFCN phenotype and an
optimized trophic environment for BFCNs by increasing levels of NGF and NT-3
(Burke et. al, manuscript in preparation) which possibly contributed to the robust
upregulation of cholinergic innervation in the hippocampus of APP/CHGFP mice.
Since ACh neurotransmission acts as a neuromodulator, the regrowth and
replacement of the exact cholinergic synapses may not be necessary to enhance
memory and cognition (Bissonnette et al., 2011).
70
BMP9 infusion quantitatively reduced Aβ plaque levels in the anterior and
posterior hippocampal sections of APP/CHGFP mice (Burke et. al, manuscript in
preparation). BMP9 also reduced the number of dystrophic cholinergic neurites
surrounding Aβ plaques in the hippocampus and shifted plaque
immunofluorescent staining patterns from scattered-diffuse to dense-compact
plaque morphologies. These data demonstrate that brains affected by AD-like
amyloidosis may be primed for BMP9 signaling since a short 7-day infusion
resulted in a dramatic increase in cholinergic fiber density and a reduction of Aβ
in APP/CHGFP mice. The data from this AD-like mouse model suggest that
BMP9 may possess some therapeutic value to counteract the two major
pathophysiological hallmarks of AD, Aβ -amyloidosis and the cholinergic deficit.
With aging and AD, BFCNs undergo morphological changes.
Qualitatively, we observed that BFCNs in 24-month old WT/CHGFP mice
appeared to have retracted arborizations and their cell somas were more
rounded in shape and smaller in size. These alterations may be due to ageinduced disruptions of specific cytoskeleton networks leading to reduced stability,
cell shrinkage and axonal retraction. Not surprisingly, APP/CHGFP BFCNs were
also observed to display smaller soma sizes and shorter projections as
compared to healthy WT/CHGFP mice. Based on the microarray data for
APP/CHGFP BFCNs, proteins associated with cytoskeletal architecture appear
to be involved in the AD-like pathophysiology affecting the form and function of
71
BFCNs. MAP2 mRNA was reduced by 50% and CAPN2 levels were increased
by more than 75% in BFCNs from APP/CHGFP mice.
Calpain 2 (CAPN2) is a calcium-activated neutral protease located to lipid
rafts where it regulates cytoskeleton turnover by cleaving actin, microtubuleassociated proteins, spectrins and neurofilaments (Adamec et al., 2002). CAPN2
has been associated in a variety of neurodegenerative disorders including AD.
Using confocal immunofluorescence, the active form of CAPN2 was shown to
colocalize with 50-75% of hyperphosphorylated, neurofibrillary tau tangles
(NFTs) in AD brain tissue (Adamec et al., 2002). These studies as well as our
microarray gene expression analysis of APPswe/PS1dE9 BFCNs implicate the
proteolytic mechanism of CAPN2 as a potential driver of cytoskeletal disruptions
of cholinergic morphology during AD-like pathophysiology.
MAP2 is another cytoskeletal protein which crosslinks and stabilizes
microtubules to microtubules and microtubules to intermediate filaments.
Dendritic localized MAP2 and axonal localized Tau (MAPT) are in the same
microtubule-associated protein (MAP) family and hyperphosphorylation of these
proteins leads to microtubule destabilization, aggregation and NFT formation.
The APPswe/PS1dE9 AD-like model do not produce NFTs whereas NFTs are a
hallmark feature observed in post-mortem human AD brain tissue.
72
PrPc
APP/CHGFP
WT/CHGFP
ChAT-GFP
73
Merge
Figure 20
Representative images and quantification of PrPc expression
(red) in BFCNs (green). APP/CHGFP (APP.PS1) basal forebrain cholinergic
neurons reveal a significant increase in the percentage of BFCNs expressing
PrPc as compared to WT/CHGFP BFCNs (p=0.007). White arrows in upper
panel indicate PrPc+ and CHATGFP+ cells. Note that APP/CHGFP BFCNs
(left, bottom panel) consistently appear smaller than WT BFCNs (left,top
panel).
74
Based on PrPc mRNA levels from the microarray analysis, we compiled z-stacks
of BFCNs expressing GFP and used a specific immunofluorescent staining to
detect PrPc protein expression in BFCNs. When compared to WT/CHGFP
control littermates, the percentage of BFCNs in APP/CHGFP BFCNs expressing
PrPc increased by 46% (Figure 20).
It should be noted that the engineered APPswe and PS1dE9 transgenes
are under the transcriptional control of an upstream (Prnp) prion promoter in
order to drive high transgene expression in the brain and highest expression is
observed in the cortex and hippocampus (Perez et al., 2007) however our
studies focused on the basal ganglia cholinergic nuclei in the medial septum and
the diagonal bands of Broca. It is highly unlikely that the integration of the
exogenous Prnp promoter would increase the transcriptional activity of the
endogenous promoter leading to increased PrPc mRNA or protein expression in
basal forebrain cholinergic neurons. Additionally, transcription preinitiation
complexes formed by TFIIA, -IIB, -IID, -IIE, -IIF, -IIH and RNA polymerase II at
transcription start sites specify basal level gene expression and are found to be
in general abundance within the nucleus (Thomas and Chiang, 2006). However
specific coactivators and cofactors which fine-tune gene-specific promoter
activity may be in shorter supply. In this scenario, introduction of the exogenous,
transgenic Prnp promoter could reduce the endogenous Prnp promoter activity
and PrPc levels by roughly half. Furthermore, there was no difference between
PrPc expression in striatal cholinergic interneurons of WT and APPswe/PS1dE9
75
mice, indicating that PrPc up-regulation demonstrated some cell-type specificity.
These data lead us to reason that the measured increase of PrPc expression in
BFCNs is not due to the effects of the exogenous Prnp promoter but is a byproduct of the underlying pathophysiology that this AD model phenotypically
expresses.
The upregulation of PrPc is interesting due to the fact that Aβ-PrPc was
shown to inhibit synaptic plasticity in the hippocampus (Lauren et al., 2009). Aβ
binding to PrPc may interfere with its physiological functions at the synapse, such
as NMDA-R regulation and cell adhesion or it may activate aberrant signaling
cascades. In support of this idea, two independent groups have proposed a
pathologic mechanism for Aβ oligomer signaling via PrPc leading to activated
FYN kinase (Larson et al., 2012; Um et al., 2012). Um et al, (Um et al., 2012)
determined that oligomeric Aβ-PrPc-FYN kinase signaling reduced synaptic
levels of NMDA-R/NR2B subunits, induced dendritic spine retraction and
neuronal impairment. Larson et al, (Larson et al., 2012) used size exclusion
chromatography to show that Aβ dimers from human AD brain extracts bind PrPc,
leading to phosphorylation and activation of FYN kinase which resulted in tau
phosphorylation and toxicity in vivo.
76
Cholinergic Hypothesis
PrP
c
Aβ-Hypothesis
Tau Hypothesis
Figure 21 PrPc at the center of the AD Triad.
This proposed
model shows how PrPc contributes to all three AD hypotheses.
This model is based on results from multiple labs demonstrating
the role of PrPc in various in-vitro and in-vivo experiments,
however the contribution of PrPc to human AD is still uncertain.
77
We present the first examination of PrPc expression levels in BFCNs of
this AD-like mouse model. This increase in PrPc mRNA expression in 6-month
old and PrPc protein levels in 8-month old APPswe/PS1dE9 mice suggests that
PrPc upregulation may potentially be an early event during AD-like pathogenesis
in this model.
The relationship of PrPc to all three major AD hypotheses is especially
intriguing. The convergence of three pathophysiological mechanisms observed
in AD onto PrPc implicates it as a target for AD treatment through the use of a
small peptidomimetic competitor of Aβ-PrPc interactions which would be able to
cross the blood-brain barrier. Finally, PrPc is known to undergo proteolytic
cleavage by ADAM17/TACE to generate an N1 terminal fragment that mediates
neuroprotection (Checler, 2012). Fluharty et al, have also demonstrated that this
PrPc-N1 fragment binds Aβ to prevent neurotoxicity (Fluharty et al., 2013). Since
PrPc was elevated in the APPswe/PS1dE9 model of AD the next studies should
investigate PrPc levels in basal forebrain cholinergic neurons of human AD brain
samples to determine if this protein is relevant to human disease. Since the
upregulation of PrPc may also lead to increased N1 shedding, this
neuroprotective fragment of PrPc could potentially be a biomarker of AD.
78
LIST OF JOURNAL ABBREVIATIONS
ACS Chem Biol
ACS Chemical Biology
Alzheimers Res Ther
Alzheimers Research and Therapy
Ann Neurol
Annals of Neurology
Annu Rev Neurosci
Annual Review of Neuroscience
Arch Intern Med
Archives of Internal Medicine
Behav Brain Res
Behavioural Brain Research
Biochem J
Biochemical Journal
Biol Psychiatry
Biological Psychiatry
Brain Res
Brain Research
Brain Res Dev Brain Res
Brain Research Developmental Brain Research
Curr Opin Cell Biol
Current Opinions in Cell Biology
Curr Opin Neurobiol
Current Opinion in Neurobiology
Curr Top Med Chem
Current Topics in Medicinal Chemistry
Dev Neurosci
Developmental Neuroscience
Embo J
The EMBO Journal
Eur J Neurosci
European Journal of Neuroscience
Expert Rev Neurother
Expert Review of Neurotherapeutics
J Biol Chem
Journal of Biological Chemistry
J Cell Biol
Journal of Cell Biology
J Chem Neuroanat
Journal of Chemical Neuroanatomy
J Comp Neurol
Journal of Comparative Neurology
79
J Geriatr Psychiatry Neurol
Journal of Geriatric Psychiatry and Neurology
J Neurochem
Journal of Neurochemistry
J Neurosci
Journal of Neuroscience
J Neurosci Res
Journal of Neuroscience Research
J Pharmacol Exp Ther
Journal of Pharmacology and Experimental
Therapeutics
J Physiol Paris
Journal of Physiology - Paris
Life Sci
Life Science
Mol Cell Biol
Molecular and Cellular Biology
Mol Neurobiol
Molecular Neurobiology
Nat Neurosci
Nature Neuroscience
Nat Protoc
Nature Protocols
Neurobiol Aging
Neurobiology of Aging
Neurobiol Dis
Neurobiology of Disease
Neurosci Lett
Neuroscience Letters
Physiol Genomics
Physiological Genomics
Proc Natl Acad Sci U S A
Proceedings of the National Academies of
Sciences, United States of America
Stem Cell Res
Stem Cell Research
Trends Cell Biol
Trends in Cell Biology
Trends Neurosci
Trends in Neuroscience
80
REFERENCES
Adamec E, Mohan P, Vonsattel JP, Nixon RA (2002) Calpain activation in
neurodegenerative diseases: confocal immunofluorescence study with
antibodies specifically recognizing the active form of calpain 2. Acta
Neuropathol 104:92-104.
Aizawa S, Teramoto K, Yamamuro Y (2012) Histone deacetylase 9 as a negative
regulator for choline acetyltransferase gene in NG108-15 neuronal cells.
Neuroscience 205:63-72.
Alier K, Ma L, Yang J, Westaway D, Jhamandas JH (2011) Abeta inhibition of
ionic conductance in mouse basal forebrain neurons is dependent upon
the cellular prion protein PrPC. J Neurosci 31:16292-16297.
Aloe L (2004) Rita Levi-Montalcini: the discovery of nerve growth factor and
modern neurobiology. Trends Cell Biol 14:395-399.
Auld DS, Mennicken F, Quirion R (2001) Nerve growth factor rapidly induces
prolonged acetylcholine release from cultured basal forebrain neurons:
differentiation between neuromodulatory and neurotrophic influences. J
Neurosci 21:3375-3382.
Barrett GL (2000) The p75 neurotrophin receptor and neuronal apoptosis. Prog
Neurobiol 61:205-229.
Bartus RT, Dean RL, 3rd, Beer B, Lippa AS (1982) The cholinergic hypothesis of
geriatric memory dysfunction. Science 217:408-414.
Basi GS, Hemphill S, Brigham EF, Liao A, Aubele DL, Baker J, Barbour R, Bova
M, Chen XH, Dappen MS, Eichenbaum T, Goldbach E, Hawkinson J,
Lawler-Herbold R, Hu K, Hui T, Jagodzinski JJ, Keim PS, Kholodenko D,
Latimer LH, Lee M, Marugg J, Mattson MN, McCauley S, Miller JL, Motter
R, Mutter L, Neitzel ML, Ni H, Nguyen L, Quinn K, Ruslim L, Semko CM,
Shapiro P, Smith J, Soriano F, Szoke B, Tanaka K, Tang P, Tucker JA, Ye
XM, Yu M, Wu J, Xu YZ, Garofalo AW, Sauer JM, Konradi AW, Ness D,
Shopp G, Pleiss MA, Freedman SB, Schenk D (2010) Amyloid precursor
81
protein selective gamma-secretase inhibitors for treatment of Alzheimer's
disease. Alzheimers Res Ther 2:36.
Bateman RJ, Siemers ER, Mawuenyega KG, Wen G, Browning KR, Sigurdson
WC, Yarasheski KE, Friedrich SW, Demattos RB, May PC, Paul SM,
Holtzman DM (2009) A gamma-secretase inhibitor decreases amyloidbeta production in the central nervous system. Ann Neurol 66:48-54.
Bekris LM, Millard S, Lutz F, Li G, Galasko DR, Farlow MR, Quinn JF, Kaye JA,
Leverenz JB, Tsuang DW, Yu CE, Peskind ER (2012) Tau
phosphorylation pathway genes and cerebrospinal fluid tau levels in
Alzheimer's disease. Am J Med Genet B Neuropsychiatr Genet 159B:874883.
Berse B, Blusztajn JK (1995) Coordinated up-regulation of choline
acetyltransferase and vesicular acetylcholine transporter gene expression
by the retinoic acid receptor alpha, cAMP, and leukemia inhibitory
factor/ciliary neurotrophic factor signaling pathways in a murine septal cell
line. J Biol Chem 270:22101-22104.
Berse B, Lopez-Coviella I, Blusztajn JK (1999) Activation of TrkA by nerve
growth factor upregulates expression of the cholinergic gene locus but
attenuates the response to ciliary neurotrophic growth factor. Biochem J
342 ( Pt 2):301-308.
Biasini E, Turnbaugh JA, Unterberger U, Harris DA (2012) Prion protein at the
crossroads of physiology and disease. Trends Neurosci 35:92-103.
Bibel M, Hoppe E, Barde YA (1999) Biochemical and functional interactions
between the neurotrophin receptors trk and p75NTR. Embo J 18:616-622.
Bielarczyk H, Szutowicz A (1989) Evidence for the regulatory function of
synaptoplasmic acetyl-CoA in acetylcholine synthesis in nerve endings.
Biochem J 262:377-380.
Bird TD (1993) Alzheimer Disease Overview.
82
Bissonnette CJ, Lyass L, Bhattacharyya BJ, Belmadani A, Miller RJ, Kessler JA
(2011) The controlled generation of functional basal forebrain cholinergic
neurons from human embryonic stem cells. Stem Cells 29:802-811.
Blusztajn JK, Berse B (2000) The cholinergic neuronal phenotype in Alzheimer's
disease. Metabolic Brain Disease 15:45-64.
Bolognesi B, Kumita JR, Barros TP, Esbjorner EK, Luheshi LM, Crowther DC,
Wilson MR, Dobson CM, Favrin G, Yerbury JJ (2010) ANS binding reveals
common features of cytotoxic amyloid species. ACS Chem Biol 5:735-740.
Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-related
changes. Acta Neuropathologica 82:239-259.
Brady DR, Phelps PE, Vaughn JE (1989) Neurogenesis of basal forebrain
cholinergic neurons in rat. Brain Res Dev Brain Res 47:81-92.
Chao MV, Hempstead BL (1995) p75 and Trk: a two-receptor system. Trends
Neurosci 18:321-326.
Checler F (2012) Two-steps control of cellular prion physiology by the
extracellular regulated kinase-1 (ERK1). Prion 6:23-25.
Cho G, Lim Y, Zand D, Golden JA (2008) Sizn1 is a novel protein that functions
as a transcriptional coactivator of bone morphogenic protein signaling. Mol
Cell Biol 28:1565-1572.
Cooper JD, Lindholm D, Sofroniew MV (1994) Reduced transport of [125I]nerve
growth factor by cholinergic neurons and down-regulated TrkA expression
in the medial septum of aged rats. Neuroscience 62:625-629.
Costantini C, Weindruch R, Della Valle G, Puglielli L (2005) A TrkA-to-p75NTR
molecular switch activates amyloid beta-peptide generation during aging.
Biochem J 391:59-67.
83
Dale JK, Vesque C, Lints TJ, Sampath TK, Furley A, Dodd J, Placzek M (1997)
Cooperation of BMP7 and SHH in the induction of forebrain ventral midline
cells by prechordal mesoderm. Cell 90:257-269.
Davies P, Maloney AJ (1976) Selective loss of central cholinergic neurons in
Alzheimer's disease. Lancet 2:1403.
de Souza LC, Chupin M, Lamari F, Jardel C, Leclercq D, Colliot O, Lehericy S,
Dubois B, Sarazin M (2012) CSF tau markers are correlated with
hippocampal volume in Alzheimer's disease. Neurobiol Aging 33:12531257.
Dickson DW, Crystal HA, Mattiace LA, Masur DM, Blau AD, Davies P, Yen SH,
Aronson MK (1992) Identification of normal and pathological aging in
prospectively studied nondemented elderly humans. Neurobiol Aging
13:179-189.
Doucette R, Ball MJ (1987) Left-right symmetry of neuronal cell counts in the
nucleus basalis of Meynert of control and of Alzheimer-diseased brains.
Brain Res 422:357-360.
Eiden LE (1998) The cholinergic gene locus. J Neurochem 70:2227-2240.
Fagan AM, Garber M, Barbacid M, Silos-Santiago I, Holtzman DM (1997) A role
for TrkA during maturation of striatal and basal forebrain cholinergic
neurons in vivo. J Neurosci 17:7644-7654.
Flames N, Pla R, Gelman DM, Rubenstein JL, Puelles L, Marin O (2007)
Delineation of multiple subpallial progenitor domains by the combinatorial
expression of transcriptional codes. J Neurosci 27:9682-9695.
Fluharty BR, Biasini E, Stravalaci M, Sclip A, Diomede L, Balducci C, La Vitola P,
Messa M, Colombo L, Forloni G, Borsello T, Gobbi M, Harris DA (2013)
An N-terminal Fragment of the Prion Protein Binds to Amyloid-beta
Oligomers and Inhibits Their Neurotoxicity in Vivo. J Biol Chem 288:78577866.
84
Foletta VC, Lim MA, Soosairajah J, Kelly AP, Stanley EG, Shannon M, He W,
Das S, Massague J, Bernard O (2003) Direct signaling by the BMP type II
receptor via the cytoskeletal regulator LIMK1. J Cell Biol 162:1089-1098.
Francis PT, Palmer AM, Snape M, Wilcock GK (1999) The cholinergic hypothesis
of Alzheimer's disease: a review of progress. Journal of Neurology,
Neurosurgery & Psychiatry:137-147.
Gelman DM, Martini FJ, Nobrega-Pereira S, Pierani A, Kessaris N, Marin O
(2009) The embryonic preoptic area is a novel source of cortical
GABAergic interneurons. J Neurosci 29:9380-9389.
Gimbel DA, Nygaard HB, Coffey EE, Gunther EC, Lauren J, Gimbel ZA,
Strittmatter SM (2010) Memory impairment in transgenic Alzheimer mice
requires cellular prion protein. Journal of Neuroscience 30:6367-6374.
Gnahn H, Hefti F, Heumann R, Schwab ME, Thoenen H (1983) NGF-mediated
increase of choline acetyltransferase (ChAT) in the neonatal rat forebrain:
evidence for a physiological role of NGF in the brain? Brain Res 285:4552.
Gong CX, Singh TJ, Grundke-Iqbal I, Iqbal K (1993) Phosphoprotein
phosphatase activities in Alzheimer disease brain. J Neurochem 61:921927.
Greferath U, Trieu J, Barrett GL (2012) The p75 neurotrophin receptor has
nonapoptotic antineurotrophic actions in the basal forebrain. J Neurosci
Res 90:278-287.
Grothe M, Heinsen H, Teipel SJ (2012) Atrophy of the cholinergic Basal forebrain
over the adult age range and in early stages of Alzheimer's disease. Biol
Psychiatry 71:805-813.
Guadagna S, Esiri MM, Williams RJ, Francis PT (2012) Tau phosphorylation in
human brain: relationship to behavioral disturbance in dementia. Neurobiol
Aging 33:2798-2806.
85
Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E,
Cruchaga C, Sassi C, Kauwe JS, Younkin S, Hazrati L, Collinge J, Pocock
J, Lashley T, Williams J, Lambert JC, Amouyel P, Goate A, Rademakers
R, Morgan K, Powell J, St George-Hyslop P, Singleton A, Hardy J (2013)
TREM2 variants in Alzheimer's disease. N Engl J Med 368:117-127.
Gunther EC, Strittmatter SM (2010) Beta-amyloid oligomers and cellular prion
protein in Alzheimer's disease. J Mol Med (Berl) 88:331-338.
Haense C, Kalbe E, Herholz K, Hohmann C, Neumaier B, Krais R, Heiss WD
(2012) Cholinergic system function and cognition in mild cognitive
impairment. Neurobiol Aging 33:867-877.
Heredia L, Helguera P, de Olmos S, Kedikian G, Sola Vigo F, LaFerla F,
Staufenbiel M, de Olmos J, Busciglio J, Caceres A, Lorenzo A (2006)
Phosphorylation of actin-depolymerizing factor/cofilin by LIM-kinase
mediates amyloid beta-induced degeneration: a potential mechanism of
neuronal dystrophy in Alzheimer's disease. J Neurosci 26:6533-6542.
Hernandez CM, Kayed R, Zheng H, Sweatt JD, Dineley KT (2010) Loss of
alpha7 nicotinic receptors enhances beta-amyloid oligomer accumulation,
exacerbating early-stage cognitive decline and septohippocampal
pathology in a mouse model of Alzheimer's disease. J Neurosci 30:24422453.
Hersh LB, Shimojo M (2003) Regulation of cholinergic gene expression by the
neuron restrictive silencer factor/repressor element-1 silencing
transcription factor. Life Sci 72:2021-2028.
Huber LJ, Chao MV (1995) A potential interaction of p75 and trkA NGF receptors
revealed by affinity crosslinking and immunoprecipitation. J Neurosci Res
40:557-563.
Imbimbo BP, Giardina GA (2011) gamma-secretase inhibitors and modulators for
the treatment of Alzheimer's disease: disappointments and hopes. Curr
Top Med Chem 11:1555-1570.
86
Iqbal K, Liu F, Gong CX, Alonso Adel C, Grundke-Iqbal I (2009) Mechanisms of
tau-induced neurodegeneration. Acta Neuropathol 118:53-69.
Johnson EM, Jr., Taniuchi M, Clark HB, Springer JE, Koh S, Tayrien MW, Loy R
(1987) Demonstration of the retrograde transport of nerve growth factor
receptor in the peripheral and central nervous system. J Neurosci 7:923929.
Jope RS, Jenden DJ (1980) The utilization of choline and acetyl coenzyme A for
the synthesis of acetylcholine. J Neurochem 35:318-325.
Kaplan DR, Miller FD (1997) Signal transduction by the neurotrophin receptors.
Curr Opin Cell Biol 9:213-221.
Kuner P, Schubenel R, Hertel C (1998) Beta-amyloid binds to p75NTR and
activates NFkappaB in human neuroblastoma cells. J Neurosci Res
54:798-804.
Larson M, Sherman MA, Amar F, Nuvolone M, Schneider JA, Bennett DA,
Aguzzi A, Lesne SE (2012) The complex PrP(c)-Fyn couples human
oligomeric Abeta with pathological tau changes in Alzheimer's disease. J
Neurosci 32:16857-16871a.
Lauren J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM (2009) Cellular
prion protein mediates impairment of synaptic plasticity by amyloid-beta
oligomers. 457:1128-1132.
Li C, Ullrich B, Zhang JZ, Anderson RG, Brose N, Sudhof TC (1995a) Ca(2+)dependent and -independent activities of neural and non-neural
synaptotagmins. Nature 375:594-599.
Li Y, Holtzman DM, Kromer LF, Kaplan DR, Chua-Couzens J, Clary DO, Knusel
B, Mobley WC (1995b) Regulation of TrkA and ChAT expression in
developing rat basal forebrain: evidence that both exogenous and
endogenous NGF regulate differentiation of cholinergic neurons. J
Neurosci 15:2888-2905.
87
Lobello K, Ryan JM, Liu E, Rippon G, Black R (2012) Targeting Beta amyloid: a
clinical review of immunotherapeutic approaches in Alzheimer's disease.
Int J Alzheimers Dis 2012:628070.
Lopez-Coviella I, Berse B, Thies RS, Blusztajn JK (2002) Upregulation of
acetylcholine synthesis by bone morphogenetic protein 9 in a murine
septal cell line. J Physiol Paris 96:53-59.
Lopez-Coviella I, Berse B, Krauss R, Thies RS, Blusztajn JK (2000) Induction
and maintenance of the neuronal cholinergic phenotype in the central
nervous system by BMP-9. Science 289:313-316.
Lopez-Coviella I, Mellott TM, Kovacheva VP, Berse B, Slack BE, Zemelko V,
Schnitzler A, Blusztajn JK (2006) Developmental pattern of expression of
BMP receptors and Smads and activation of Smad1 and Smad5 by BMP9
in mouse basal forebrain. Brain Res 1088:49-56.
Lopez-Coviella I, Follettie MT, Mellott TJ, Kovacheva VP, Slack BE, Diesl V,
Berse B, Thies RS, Blusztajn JK (2005) Bone morphogenetic protein 9
induces the transcriptome of basal forebrain cholinergic neurons. Proc
Natl Acad Sci U S A 102:6984-6989.
Lue LF, Kuo YM, Roher AE, Brachova L, Shen Y, Sue L, Beach T, Kurth JH,
Rydel RE, Rogers J (1999) Soluble amyloid beta peptide concentration as
a predictor of synaptic change in Alzheimer's disease. Am J Pathol
155:853-862.
Marin O, Anderson SA, Rubenstein JL (2000) Origin and molecular specification
of striatal interneurons. J Neurosci 20:6063-6076.
Martyn AC, De Jaeger X, Magalhaes AC, Kesarwani R, Goncalves DF, Raulic S,
Guzman MS, Jackson MF, Izquierdo I, Macdonald JF, Prado MA, Prado
VF (2012) Elimination of the vesicular acetylcholine transporter in the
forebrain causes hyperactivity and deficits in spatial memory and longterm potentiation. Proc Natl Acad Sci U S A 109:17651-17656.
Massague J, Wotton D (2000) Transcriptional control by the TGF-beta/Smad
signaling system. Embo J 19:1745-1754.
88
McLean CA, Cherny RA, Fraser FW, Fuller SJ, Smith MJ, Beyreuther K, Bush AI,
Masters CL (1999) Soluble pool of Abeta amyloid as a determinant of
severity of neurodegeneration in Alzheimer's disease. Ann Neurol 46:860866.
Mesulam M (2004) The cholinergic Lesion of Alzheimer's Disease: Pivotal Factor
or Side Show? Learning and Memory:43-49.
Mineur YS, McLoughlin D, Crusio WE, Sluyter F (2005) Genetic mouse models
of Alzheimer's disease. Neural Plast 12:299-310.
Moises T, Dreier A, Flohr S, Esser M, Brauers E, Reiss K, Merken D, Weis J,
Kruttgen A (2007) Tracking TrkA's trafficking: NGF receptor trafficking
controls NGF receptor signaling. Mol Neurobiol 35:151-159.
Montagna P, Gambetti P, Cortelli P, Lugaresi E (2003) Familial and sporadic fatal
insomnia. Lancet Neurol 2:167-176.
Mori T, Yuxing Z, Takaki H, Takeuchi M, Iseki K, Hagino S, Kitanaka J,
Takemura M, Misawa H, Ikawa M, Okabe M, Wanaka A (2004) The LIM
homeobox gene, L3/Lhx8, is necessary for proper development of basal
forebrain cholinergic neurons. Eur J Neurosci 19:3129-3141.
Mufson EJ, Li JM, Sobreviela T, Kordower JH (1996) Decreased trkA gene
expression within basal forebrain neurons in Alzheimer's disease.
Neuroreport 8:25-29.
Mufson EJ, Ginsberg SD, Ikonomovic MD, DeKosky ST (2003) Human
cholinergic basal forebrain: chemoanatomy and neurologic dysfunction. J
Chem Neuroanat 26:233-242.
Mufson EJ, Counts SE, Perez SE, Ginsberg SD (2008) Cholinergic system
during the progression of Alzheimer's disease: therapeutic implications.
Expert Rev Neurother 8:1703-1718.
Mufson EJ, Ma SY, Dills J, Cochran EJ, Leurgans S, Wuu J, Bennett DA, Jaffar
S, Gilmor ML, Levey AI, Kordower JH (2002) Loss of basal forebrain
89
P75(NTR) immunoreactivity in subjects with mild cognitive impairment and
Alzheimer's disease. J Comp Neurol 443:136-153.
Naj AC, Jun G, Beecham GW, Wang LS, Vardarajan BN, Buros J, Gallins PJ,
Buxbaum JD, Jarvik GP, Crane PK, Larson EB, Bird TD, Boeve BF, GraffRadford NR, De Jager PL, Evans D, Schneider JA, Carrasquillo MM,
Ertekin-Taner N, Younkin SG, Cruchaga C, Kauwe JS, Nowotny P,
Kramer P, Hardy J, Huentelman MJ, Myers AJ, Barmada MM, Demirci FY,
Baldwin CT, Green RC, Rogaeva E, St George-Hyslop P, Arnold SE,
Barber R, Beach T, Bigio EH, Bowen JD, Boxer A, Burke JR, Cairns NJ,
Carlson CS, Carney RM, Carroll SL, Chui HC, Clark DG, Corneveaux J,
Cotman CW, Cummings JL, DeCarli C, DeKosky ST, Diaz-Arrastia R, Dick
M, Dickson DW, Ellis WG, Faber KM, Fallon KB, Farlow MR, Ferris S,
Frosch MP, Galasko DR, Ganguli M, Gearing M, Geschwind DH, Ghetti B,
Gilbert JR, Gilman S, Giordani B, Glass JD, Growdon JH, Hamilton RL,
Harrell LE, Head E, Honig LS, Hulette CM, Hyman BT, Jicha GA, Jin LW,
Johnson N, Karlawish J, Karydas A, Kaye JA, Kim R, Koo EH, Kowall NW,
Lah JJ, Levey AI, Lieberman AP, Lopez OL, Mack WJ, Marson DC,
Martiniuk F, Mash DC, Masliah E, McCormick WC, McCurry SM, McDavid
AN, McKee AC, Mesulam M, Miller BL, et al. (2011) Common variants at
MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with lateonset Alzheimer's disease. Nat Genet 43:436-441.
Niedowicz DM, Beckett TL, Matveev S, Weidner AM, Baig I, Kryscio RJ,
Mendiondo MS, LeVine H, 3rd, Keller JN, Murphy MP (2012) Pittsburgh
compound B and the postmortem diagnosis of Alzheimer disease. Ann
Neurol 72:564-570.
Niewiadomska G, Mietelska-Porowska A, Mazurkiewicz M (2011) The cholinergic
system, nerve growth factor and the cytoskeleton. Behav Brain Res
221:515-526.
Nobrega-Pereira S, Gelman D, Bartolini G, Pla R, Pierani A, Marin O (2010)
Origin and molecular specification of globus pallidus neurons. J Neurosci
30:2824-2834.
Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-Bongaarts A,
Ohno M, Disterhoft J, Van Eldik L, Berry R, Vassar R (2006) Intraneuronal
beta-amyloid aggregates, neurodegeneration, and neuron loss in
90
transgenic mice with five familial Alzheimer's disease mutations: potential
factors in amyloid plaque formation. J Neurosci 26:10129-10140.
Okuda T, Haga T, Kanai Y, Endou H, Ishihara T, Katsura I (2000) Identification
and characterization of the high-affinity choline transporter. Nat Neurosci
3:120-125.
Olzscha H, Schermann SM, Woerner AC, Pinkert S, Hecht MH, Tartaglia GG,
Vendruscolo M, Hayer-Hartl M, Hartl FU, Vabulas RM (2011) Amyloid-like
aggregates sequester numerous metastable proteins with essential
cellular functions. Cell 144:67-78.
Palop JJ, Mucke L (2010) Synaptic depression and aberrant excitatory network
activity in Alzheimer's disease: two faces of the same coin?
Neuromolecular Med 12:48-55.
Perez SE, Dar S, Ikonomovic MD, DeKosky ST, Mufson EJ (2007) Cholinergic
forebrain degeneration in the APPswe/PS1DeltaE9 transgenic mouse.
Neurobiol Dis 28:3-15.
Perry EK, Perry RH, Tomlinson BE, Blessed G, Gibson PH (1980) Coenzyme Aacetylating enzymes in Alzheimer's disease: possible cholinergic
'compartment' of pyruvate dehydrogenase. Neurosci Lett 18:105-110.
Picciotto MR, Higley MJ, Mineur YS (2012) Acetylcholine as a neuromodulator:
cholinergic signaling shapes nervous system function and behavior.
Neuron 76:116-129.
Piltti KM, Haus DL, Do E, Perez H, Anderson AJ, Cummings BJ (2011)
Computer-aided 2D and 3D quantification of human stem cell fate from in
vitro samples using Volocity high performance image analysis software.
Stem Cell Res 7:256-263.
Price M, Lazzaro D, Pohl T, Mattei MG, Ruther U, Olivo JC, Duboule D, Di Lauro
R (1992) Regional expression of the homeobox gene Nkx-2.2 in the
developing mammalian forebrain. Neuron 8:241-255.
91
Prusiner SB, DeArmond SJ (1990) Prion disease of the central nervous system.
Monographs in Pathology:86-122.
Ray PG, Meador KJ, Loring DW, Zamrini EW, Yang XH, Buccafusco JJ (1992)
Central anticholinergic hypersensitivity in aging. J Geriatr Psychiatry
Neurol 5:72-77.
Robertson RT, Baratta J, Yu J, Guthrie KM (2006) A role for neurotrophin-3 in
targeting developing cholinergic axon projections to cerebral cortex.
Neuroscience 143:523-539.
Rogers SL, Doody RS, Mohs RC, Friedhoff LT (1998) Donepezil improves
cognition and global function in Alzheimer disease: a 15-week, doubleblind, placebo-controlled study. Donepezil Study Group. Arch Intern Med
158:1021-1031.
Rubenstein JL, Beachy PA (1998) Patterning of the embryonic forebrain. Curr
Opin Neurobiol 8:18-26.
Rubenstein JL, Martinez S, Shimamura K, Puelles L (1994) The embryonic
vertebrate forebrain: the prosomeric model. Science 266:578-580.
Savelieff MG, Lee S, Liu Y, Lim MH (2013) Untangling Amyloid-beta, Tau, and
Metals in Alzheimer's Disease. ACS Chem Biol.
Savonenko A, Xu GM, Melnikova T, Morton JL, Gonzales V, Wong MP, Price DL,
Tang F, Markowska AL, Borchelt DR (2005) Episodic-like memory deficits
in the APPswe/PS1dE9 mouse model of Alzheimer's disease:
relationships to beta-amyloid deposition and neurotransmitter
abnormalities. Neurobiol Dis 18:602-617.
Schambra UB, Sulik KK, Petrusz P, Lauder JM (1989) Ontogeny of cholinergic
neurons in the mouse forebrain. J Comp Neurol 288:101-122.
Schliebs R, Arendt T (2011) The cholinergic system in aging and neuronal
degeneration. Behav Brain Res 221:555-563.
92
Schnitzler AC, Lopez-Coviella I, Blusztajn JK (2008a) Purification and culture of
nerve growth factor receptor (p75)-expressing basal forebrain cholinergic
neurons. Nat Protoc 3:34-40.
Schnitzler AC, Lopez-Coviella I, Blusztajn JK (2008b) Differential modulation of
nerve growth factor receptor (p75) and cholinergic gene expression in
purified p75-expressing and non-expressing basal forebrain neurons by
BMP9. Brain Res 1246:19-28.
Schnitzler AC, Mellott TJ, Lopez-Coviella I, Tallini YN, Kotlikoff MI, Follettie MT,
Blusztajn JK (2010) BMP9 (bone morphogenetic protein 9) induces NGF
as an autocrine/paracrine cholinergic trophic factor in developing basal
forebrain neurons. J Neurosci 30:8221-8228.
Schwab C, Bruckner G, Mares V, Biesold D (1988) A combined method of
acetylcholinesterase histochemistry and [3H]thymidine autoradiography:
application to neurogenesis of the rat basal forebrain cholinergic system.
Neurosci Lett 90:69-74.
Selkoe DJ (1997) Alzheimer's disease: genotypes, phenotypes, and treatments.
Science 275:630-631.
Selkoe DJ (2002) Alzheimer's disease is a synaptic failure. Science 298:789-791.
Selkoe DJ (2012) Preventing Alzheimer's disease. Science 337:1488-1492.
Shimamura K, Hartigan DJ, Martinez S, Puelles L, Rubenstein JL (1995)
Longitudinal organization of the anterior neural plate and neural tube.
Development 121:3923-3933.
Slotkin TA, Seidler FJ, Crain BJ, Bell JM, Bissette G, Nemeroff CB (1990)
Regulatory changes in presynaptic cholinergic function assessed in rapid
autopsy material from patients with Alzheimer disease: implications for
etiology and therapy. Proc Natl Acad Sci U S A 87:2452-2455.
Sobreviela T, Clary DO, Reichardt LF, Brandabur MM, Kordower JH, Mufson EJ
(1994) TrkA-immunoreactive profiles in the central nervous system:
93
colocalization with neurons containing p75 nerve growth factor receptor,
choline acetyltransferase, and serotonin. J Comp Neurol 350:587-611.
Sofroniew MV, Howe CL, Mobley WC (2001) Nerve growth factor signaling,
neuroprotection, and neural repair. Annu Rev Neurosci 24:1217-1281.
Sotthibundhu A, Sykes AM, Fox B, Underwood CK, Thangnipon W, Coulson EJ
(2008) Beta-amyloid(1-42) induces neuronal death through the p75
neurotrophin receptor. J Neurosci 28:3941-3946.
Sudhof TC (1995) The synaptic vesicle cycle: a cascade of protein-protein
interactions. Nature 375:645-653.
Sussel L, Marin O, Kimura S, Rubenstein JL (1999) Loss of Nkx2.1 homeobox
gene function results in a ventral to dorsal molecular respecification within
the basal telencephalon: evidence for a transformation of the pallidum into
the striatum. Development 126:3359-3370.
Szutowicz A, Tomaszewicz M, Bielarczyk H (1996) Disturbances of acetyl-CoA,
energy and acetylcholine metabolism in some encephalopathies. Acta
Neurobiol Exp (Wars) 56:323-339.
Takashima A (2010) Tau aggregation is a therapeutic target for Alzheimer's
disease. Curr Alzheimer Res 7:665-669.
Terry AV, Jr., Buccafusco JJ (2003) The cholinergic hypothesis of age and
Alzheimer's disease-related cognitive deficits: recent challenges and their
implications for novel drug development. J Pharmacol Exp Ther 306:821827.
Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, Hansen LA,
Katzman R (1991) Physical basis of cognitive alterations in Alzheimer's
disease: synapse loss is the major correlate of cognitive impairment. Ann
Neurol 30:572-580.
Thomas MC, Chiang CM (2006) The general transcription machinery and general
cofactors. Crit Rev Biochem Mol Biol 41:105-178.
94
Um JW, Nygaard HB, Heiss JK, Kostylev MA, Stagi M, Vortmeyer A, Wisniewski
T, Gunther EC, Strittmatter SM (2012) Alzheimer amyloid-beta oligomer
bound to postsynaptic prion protein activates Fyn to impair neurons. Nat
Neurosci 15:1227-1235.
Walsh DM, Selkoe DJ (2004) Deciphering the molecular basis of memory failure
in Alzheimer's disease. Neuron 44:181-193.
Wanaka A, Matsumoto K, Kashihara Y, Furuyama T, Tanaka T, Mori T, Tanno Y,
Yokoya S, Kitanaka J, Takemura M, Tohyama M (1997) LIMhomeodomain gene family in neural development. Dev Neurosci 19:97100.
Wang JY, Frenzel KE, Wen D, Falls DL (1998) Transmembrane neuregulins
interact with LIM kinase 1, a cytoplasmic protein kinase implicated in
development of visuospatial cognition. J Biol Chem 273:20525-20534.
Wang JZ, Grundke-Iqbal I, Iqbal K (1996) Restoration of biological activity of
Alzheimer abnormally phosphorylated tau by dephosphorylation with
protein phosphatase-2A, -2B and -1. Brain Res Mol Brain Res 38:200-208.
Wang YJ, Wang X, Lu JJ, Li QX, Gao CY, Liu XH, Sun Y, Yang M, Lim Y, Evin
G, Zhong JH, Masters C, Zhou XF (2011) p75NTR regulates Abeta
deposition by increasing Abeta production but inhibiting Abeta aggregation
with its extracellular domain. J Neurosci 31:2292-2304.
Ward SM, Himmelstein DS, Lancia JK, Binder LI (2012) Tau oligomers and tau
toxicity in neurodegenerative disease. Biochem Soc Trans 40:667-671.
Wedeen VJ, Rosene DL, Wang R, Dai G, Mortazavi F, Hagmann P, Kaas JH,
Tseng WY (2012) The geometric structure of the brain fiber pathways.
Science 335:1628-1634.
Wei B, Huang Z, He S, Sun C, You Y, Liu F, Yang Z (2012) The onion skin-like
organization of the septum arises from multiple embryonic origins to form
multiple adult neuronal fates. Neuroscience 222:110-123.
95
Wurtman RJ (1992) Choline metabolism as a basis for the selective vulnerability
of cholinergic neurons. Trends Neurosci 15:117-122.
Yeo TT, Chua-Couzens J, Butcher LL, Bredesen DE, Cooper JD, Valletta JS,
Mobley WC, Longo FM (1997) Absence of p75NTR causes increased
basal forebrain cholinergic neuron size, choline acetyltransferase activity,
and target innervation. J Neurosci 17:7594-7605.
Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, Maeda J,
Suhara T, Trojanowski JQ, Lee VM (2007) Synapse loss and microglial
activation precede tangles in a P301S tauopathy mouse model. Neuron
53:337-351.
You H, Tsutsui S, Hameed S, Kannanayakal TJ, Zamponi GW (2011) A-beta
neurotoxicity depends on interactions between copper ions, prion protein,
and N-methyl-D-aspartate receptors. PNAS-Proceedings of the National
Academy of Sciences of the United States of America 109:1737-1742.
Zampieri N, Xu CF, Neubert TA, Chao MV (2005) Cleavage of p75 neurotrophin
receptor by alpha-secretase and gamma-secretase requires specific
receptor domains. J Biol Chem 280:14563-14571.
Zhang Y, Hong Y, Bounhar Y, Blacker M, Roucou X, Tounekti O, Vereker E,
Bowers WJ, Federoff HJ, Goodyer CG, LeBlanc A (2003) p75
neurotrophin receptor protects primary cultures of human neurons against
extracellular amyloid beta peptide cytotoxicity. J Neurosci 23:7385-7394.
Zhao Y, Marin O, Hermesz E, Powell A, Flames N, Palkovits M, Rubenstein JL,
Westphal H (2003) The LIM-homeobox gene Lhx8 is required for the
development of many cholinergic neurons in the mouse forebrain. Proc
Natl Acad Sci U S A 100:9005-9010.
96
CURRICULUM VITAE
Timothy Alfred Norman Jr.
Year of Birth: 1983
[email protected]
EDUCATION
University of North Carolina at Wilmington, Wilmington, NC
Bachelor of Science in Biology – May 2005
Boston University School of Medicine, Boston, MA
Master of Arts – May 2013
Department of Pathology and Laboratory Medicine
EXPERIENCE
BOSTON UNIVERSITY
SCHOOL OF MEDICINE
9/2011-5/2013
Graduate Student
Project 1: Analysis of differential protein expression in neurons
using immunohistochemistry and confocal microscopy.
Discovered the cellular prion protein was upregulated in
cholinergic neurons of the APPswe/PS1dE9 mouse model of AD.
Project 2: Confocal imaging, immunofluorescence and
quantification of cholinergic innervation in the hippocampus of
APPswe/PS1dE9 mice after BMP9 infusion.
Teaching Assistant, BT432: Basic Mechanisms of Disease
Crafted and delivered powerpoint lectures for cardiovascular
th
diseases and traumatic brain injury from Robbins and Cotran 8
edition with Dr. Chris Andry
MEDCO HEALTH
SOLUTIONS
11/2007-4/2011
Lead Pharmacy Technician
Communicated assignments and departmental production goals
to pharmacists and technicians. Ensured product safety and
accuracy while processing 750K prescriptions per week.
Performed new hire training and minor supervisory and
administrative functions including schedule preparation and goal
targets.
UNIVERSITY OF UTAH
MEDICAL CENTER
3/2006-8/2006
Psychiatric Technician
Assessed patient mental status and activities of daily living.
Application of crisis prevention and intervention techniques to
ensure patient and staff safety. Obtained vital signs,
documented patient behavior and assisted with group therapy.
PUBLICATIONS
Rebecca M. Burke, Timothy A. Norman, Tarik F. Haydar, Jan
Krzystof Bluszajn, Tiffany J. Mellot. BMP9 ameliorates the
amyloidosis and the cholinergic defect in the APPswe/PS1dE9
mouse model of Alzheimer’s disease. (manuscript in preparation)
97