the master synaptic regulator: activity regulated

112
City University of New York (CUNY) City University of New York (CUNY) CUNY Academic Works CUNY Academic Works Dissertations, Theses, and Capstone Projects CUNY Graduate Center 2-2019 The Master Synaptic Regulator: Activity Regulated Cytoskeleton The Master Synaptic Regulator: Activity Regulated Cytoskeleton Associated Protein, Arc, in Normal Aging and Diseases with Associated Protein, Arc, in Normal Aging and Diseases with Cognitive Impairment Cognitive Impairment Amber Khan The Graduate Center, City University of New York How does access to this work benefit you? Let us know! More information about this work at: https://academicworks.cuny.edu/gc_etds/2988 Discover additional works at: https://academicworks.cuny.edu This work is made publicly available by the City University of New York (CUNY). Contact: [email protected]

Upload: others

Post on 13-Feb-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

City University of New York (CUNY) City University of New York (CUNY)

CUNY Academic Works CUNY Academic Works

Dissertations, Theses, and Capstone Projects CUNY Graduate Center

2-2019

The Master Synaptic Regulator: Activity Regulated Cytoskeleton The Master Synaptic Regulator: Activity Regulated Cytoskeleton

Associated Protein, Arc, in Normal Aging and Diseases with Associated Protein, Arc, in Normal Aging and Diseases with

Cognitive Impairment Cognitive Impairment

Amber Khan The Graduate Center, City University of New York

How does access to this work benefit you? Let us know!

More information about this work at: https://academicworks.cuny.edu/gc_etds/2988

Discover additional works at: https://academicworks.cuny.edu

This work is made publicly available by the City University of New York (CUNY). Contact: [email protected]

The Master Synaptic Regulator: Activity Regulated

Cytoskeleton Associated Protein, Arc, in Normal Aging and

Diseases with Cognitive impairment.

by

Amber Khan

A dissertation submitted to the Graduate Faculty in Biology in partial fulfillment of the

requirements for the degree of Doctor of Philosophy, The City University of New York

2019

ii

© 2018

AMBER KHAN

All Rights Reserved

iii

This manuscript has been read and accepted for the Graduate Faculty in Biology in satisfaction

of the dissertation requirement for the degree of Doctor of Philosophy.

___________________ ______________________________________________

Date Chair of Examining Committee

Dr. Hoau-Yan Wang, CUNY School of Medicine

___________________ ______________________________________________

Date Executive Officer

Dr. Cathy Savage-Dunn

______________________________________________

Dr. Itzhak Mano, CUNY School of Medicine

______________________________________________

Dr. John H. Martin, CUNY School of Medicine

_______________________________________________

Dr. Jonathan Levitt, City College

________________________________________________

Dr. Robert Nagele,

Rowan University School of Osteopathic Medicine

______________________________________________

Supervising Committee

The City University of New York

iv

Abstract:

Master Synaptic Regulator: Activity Regulated Cytoskeleton

Associated Protein, Arc in Normal Aging and Diseases with

Cognitive impairment.

By: Amber Khan

Advisor: Dr. Hoau-Yan Wang

Alzheimer’s disease (AD) is a progressive neurodegenerative disease with complex

underlying pathogenic mechanisms. Epidemiological studies have forecasted that in the next 3

decades, the number of AD cases will rise to epidemic proportions with enormous medical,

emotional and financial burdens impacting individuals affected and society. Among many risk

factors for AD, advancing age is clearly essential and necessary. Revelation of molecular changes

in synaptic activities leading to the prodromal, mild cognitive impairment (MCI) stage may help

illuminate the course of pathogenic progression and its cause-effect relationship with various

targets thereby enabling target-driven disease-modifying therapeutic agents for AD.

Activity-regulated cytoskeleton-associated (Arc) protein is a prominent regulator of

synaptic plasticity and homeostasis that localizes exclusively in postsynaptic regions of the

excitatory systems in the brain. Arc is involved in AMPA receptor endocytosis in LTD and late

phase LTP consolidation at the dendritic fields. NMDA receptor activation increases Arc

expression to facilitate synaptic activities and promote remodeling of dendritic spines. AD

pathologies can be found in brains of cognitively normal elderly individuals. Together with the

fact that synaptic activity is altered during aging and markedly deteriorated in AD, we therefore

v

hypothesize that altered basal and activity-driven Arc expression contribute to deleterious synaptic

changes at old age and in AD. The altered Arc contributes to synaptopathy in AD through its

interactions at the postsynaptic density and dendritic spine. However, the mechanisms responsible

for Arc alteration during normal aging and in AD are currently not clear.

In these studies, we systemically investigate changes in Arc protein levels under basal and

stimulation conditions in hippocampal formation (HF) and prefrontal cortex (PFC) from wild-type

(WT) and 3x transgenic (Tg) AD mice at varying ages. The results derived from WT and 3xTg

AD mice show Arc protein levels increase along with advancing age and in AD under non-

stimulated basal condition. More importantly, Arc expression is increased in response to NMDAR

and insulin receptor stimulation and these stimulation-elicited Arc increases are dramatically

attenuated in AD. We present evidence to show Arc is regulated by phosphorylation on the serine

and tyrosine residues and this post-translational modification process is driven by receptor

stimulation. Arc is also sensitive to oxidative damage as indicated by elevation of nitrated Arc

levels in aged WT and 3 x Tg AD mice. We reveal that Arc is associated with PSD-95/NMDAR

and filamin A (FLNA) signaling pathways. Further, we identify protein kinase C (PKC) and Src

in the PSD-95/NMDAR complexes as well as JAK2 and PAK1 in the FLNA signaling cascade as

the kinases that phosphorylate Arc following activation of the NMDARs and insulin receptor,

respectively. Most importantly, we observed a reduced association of Arc with NMDARs

accompanied by increased Arc linkage to FLNA during normal aging and in AD.

The relevancy of the findings in mouse AD models is affirmed by examining the

postmortem human HF from age-, gender- and postmortem interval-matched sets of cognitively

normal controls, subjects with mild cognitive impairment (MCI) with or without AD pathology

(MCI-AD and MCI-SNAP, respectively) and AD cases. Arc protein levels are higher in MCI-

vi

SNAP and AD under the non-stimulated conditions. Similar to the observation made in mouse

brain, Arc expression is increased by exposure to NMDA/glycine (NMDAR), PNU282987 (α7

nicotinic receptor), insulin (IR) and BDNF (TrkB) in human HF slices from non-demented

controls. The receptor stimulation induced Arc expression is universally and markedly reduced in

MCI-AD and AD as well as in MCI-SNAP although with lesser extent. Oxidative damage to Arc

is evidenced in MCI-AD and AD but not in MCI-SNAP. Arc is predominantly associated with

PSD-95/NMDARs in control and MCI-SNAP cases but linked to FLNA in MCI-AD and AD.

In summary, the data presented indicate for the first time that phosphorylation of Arc

mediated by kinases in its associated NMDAR and FLNA is a regulatory mechanism for Arc under

physiological conditions. More importantly, defected activity-driven Arc expression is observed

prevalently in diseases with cognitive impairment. The reduced activity-induced Arc expression

can occur with or without elevated Arc protein levels and the altered connections with PSD-

95/NMDAR and FLNA signaling complexes. The data derived from this study indicate that

reduced activity-driven Arc expression and altered Arc connections occur early in the course of

synaptopathy and are integral parts of AD pathologies. Our data suggest that restoring activity-

driven Arc expression may rescue synaptic dysfunction and thereby improve cognitive function in

diseases with cognitive impairments such as Alzheimer’s disease.

vii

Table of Contents:

Abstract: ....................................................................................................................................... iv

Introduction: ................................................................................................................................. 1

a) Cognitive ability varies with age and disease: .................................................................................... 1

I) Specific pathological changes correlate with cognitive decline. .................................................... 2

b) Mild Cognitive Impairment (MCI) is clinically and pathologically a unique, early stage of

dementia. .................................................................................................................................................. 3

c) Alzheimer’s disease is the most common cause of dementia in the global population. .................... 5

I) Alzheimer’s disease is a public health epidemic. ............................................................................ 6

II) Diseases of cognitive impairment are clinically assessed using a diagnostic screening tool. ...... 7

III) Disease pathogenesis is measured and monitored using various detection methods. ................ 8

d) Alzheimer’s Disease has a complex, multifactorial pathogenesis. .................................................. 10

I) Amyloid Beta Plaque Pathology and the Amyloid Hypothesis. .................................................... 11

II) Tau Hyperphosphorylation and the Tau Hypothesis .................................................................. 15

e) Early Neurodegeneration and dysfunction in Alzheimer’s Disease is characterized by synaptic

changes and loss of synapses. ................................................................................................................ 17

f) The post-synaptic density of glutamatergic, excitatory synapses is a major site for long term

potentiation and depression. .................................................................................................................. 19

I) LTP and LTD at glutamatergic excitatory synapses depend on NMDA and AMPA receptor

activity. ................................................................................................................................................ 19

II) Scaffolding Proteins PSD-95 and FLNA function as binding partners and regulate receptors

in key signaling pathways involved in memory formation. .............................................................. 21

III) Immediate Early Genes and their protein products function as a rapid response mechanism to

cellular stimuli. ................................................................................................................................... 23

g) Activity Regulated Cytoskeleton Associated Protein (Arc) ............................................................... 24

I) Synaptic Functions of Arc mediate AMPAR Regulation in LTP/LTD. ....................................... 25

II) Nuclear functions of Arc .............................................................................................................. 28

III) Regulatory mechanisms of Arc: posttranslational modifications ............................................. 29

h) Arc expression and function is altered in disease ............................................................................ 31

I) Cognitive disorders, aging and AD ................................................................................................ 32

viii

Experimental Procedures ........................................................................................................... 35

Human Postmortem Hippocampal formation sections......................................................................... 35

Description of case cohorts .................................................................................................................... 35

3xTg AD Mouse Model Tissue Preservation and Selection ................................................................. 38

a) Ex-Vivo Stimulation ....................................................................................................................... 39

b) Immunoprecipitation and co-immunoprecipitation ...................................................................... 40

c) Western Blot Procedure ................................................................................................................. 41

d) Statistical Evaluation ..................................................................................................................... 43

Results .......................................................................................................................................... 43

1) Isolation of Arc by immunoprecipitation with anti-Arc is complete and specific. ................... 43

2) Assessing Arc Expression .................................................................................................................. 46

2a) Basal and Activity-driven Arc expression is decreased during normal aging and in AD. ........ 46

2b) Assessment of the overall Arc protein levels. .............................................................................. 49

2c) Altered basal and activity-driven Arc expression in Human postmortem HF from well-

matched subjects with Alzheimer’s disease (AD), mild cognitive impairment (MCI) and cognitively

normal control subjects. ..................................................................................................................... 51

3) Arc is modified post-translationally by phosphorylation. ............................................................. 54

3a) Phosphorylation of Arc at serine (pS) and tyrosine (pY) is responsive to receptor stimulation.

............................................................................................................................................................ 58

3b) Arc phosphorylation states are altered during normal aging and in AD pathogenesis. ........... 58

4) Nitration of Arc indicates oxidative stress occurs during AD pathogenesis modifies Arc. ........ 64

6) Regulation of Arc by Phosphorylation is mediated by PKC/Src and JAK2/PAK1 associated

respectively with NMDARs and FLNA ............................................................................................... 73

Conclusions .................................................................................................................................. 77

References .................................................................................................................................... 86

ix

Table of Figures

Figure 1: Time course of Cognitive Decline ....................................................................................1

Figure 2: Comparison of current criteria for mild cognitive impairment (MCI) .............................4

Figure 3: Suggested criteria for the likelihood that MCI is due to AD............................................5

Figure 4: Projected Populations of AD ............................................................................................6

Figure 5: A hypothetical temporal model of Alzheimer's disease biomarkers ................................9

Figure 6: APP processing occurs via either the amyloidogenic or canonical pathway .................12

Figure 7: Tau hyperphosphorylation and Tau pathology. ..............................................................15

Figure 8: The sequence of major pathogenic events leading to AD proposed by the amyloid

cascade hypothesis .........................................................................................................................17

Figure 9: Mechanisms of LTP and LTD in the PSD .....................................................................20

Figure 10: Proposed Molecular Organization of the PSD .............................................................22

Figure 11: Graphic Representation of Arc domain structure ........................................................25

Figure 12: Arc has multiple effects through its actions in the synapse and nucleus .....................27

Figure 13: Sites of Post-translational modifications on Arc ..........................................................29

Figure 14: Anti-Arc immunoprecipitation in the HF and PFC is complete and specific ..............46

Figure 15: Activity dependent Arc expression changes as a function of age and disease .............49

Figure 16: Basal Arc Expression levels in aging and AD .............................................................51

Figure 17: Arc expression is altered in cognitively impaired human HF ......................................53

Figure 18: Arc immunoprecipites contain serine- and tyrosine- but not threonine-phosphorylated

Arc in the HF and PFC...................................................................................................................56

x

Figure 19: Immunoprecipitation with antibodies against the indicated phosphoepitopes in the HF

and PFC from WT mice .................................................................................................................57

Figure 20: Basal and activity dependent Arc phosphorylation expression are altered in HF of

aged WT and 3xTg AD mice .........................................................................................................60

Figure 21: Basal and activity dependent Arc phosphorylation expression are altered in PFC of

aged WT and 3xTg AD mice. ........................................................................................................61

Figure 22: Human HF shows disease dependent changes in Arc phosphorylation. ......................62

Figure 23: a-c: Arc does not show phosphorylation on the Threonine epitope .............................63

Figure 24: Arc is modified by nitration in the HF and PFC of WT and 3xTg AD animals ..........65

Figure 25: Increased nitrated Arc in the postmortem human HF from patients with cognitive

impairments....................................................................................................................................66

Figure 26: Altered Arc linkages with signaling complexes in aged and 3xTg AD mice ..............70

Figure 27: Altered Arc linkages with signaling complexes in aged and 3xTg AD mice in HF ....71

Figure 28: Altered Arc linkages with signaling complexes in aged and 3xTg AD mice in PFC ..72

Figure 29: Identification of critical kinases responsible for pS-Arc and pY-Arc in HF................75

Figure 30: Revelation of the kinases that mediate Arc phosphorylation in PFC ...........................76

Figure 31: Proposed mechanism of Arc function in physiological and pathogenic conditions .....84

1

Introduction:

a) Cognitive ability varies with age and disease:

Basic cognitive functions are broadly defined as processes to attain knowledge of, and

interact with, the environment. These abilities are a dynamic skill set that include memory (explicit

versus implicit), intelligence (crystallized versus fluid), and executive function, and are some of

the primary functions that decline with age.1 These abilities can also be influenced by other factors

such as genetics and lifestyle choices which also affect the brain throughout an individual’s

lifetime. When comparing the effects of neurodegenerative disease on cognitive abilities, a greater

functional deficit is apparent compared to the healthy age-matched population (figure 1).1-3

Figure 1: Time course of Cognitive Decline. Progression from normal aging to Alzheimer’s disease or other

types of dementia. Cognitive function diminishes with advancing age. The onset and progression of dementia

correlates with the severity of the loss of function progressing.

Source: http://www.mind.uci.edu/dementia/mild-cognitive-impairment/

2

I) Specific pathological changes correlate with cognitive decline.

Several pathological changes have been connected to age-related cognitive decline.

Region-specific changes in grey matter volume begin after the age of twenty and continue to

progress in an age-dependent manner1. Such changes have been attributed to several potential

causes, including subtle changes in the synaptic structure of the brain especially in the

hippocampus and prefrontal cortex. As studies in mice have shown, animals undergo age-

dependent changes in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA-

R) and N-methyl-D-aspartate receptor (NMDA-R) density and assembly’s in the postsynaptic

density (PSD)4. In Sprague-Dawley rats, the abundance of NMDA-Rs is lower in animals at 25

months of age and older compared to very young rats (< 3 months)5,6. Such changes in

compositions and/or density of these crucial synaptic receptors can conceivably negatively affect

the neuronal function due to altered signaling downstream of NMDA-Rs and AMPA-Rs.

Significantly, these receptor changes also compromise synaptic transmission and ultimately,

diminish long term potentiation induction and maintenance5-7. Such changes contribute to overall

grey matter volume loss through decreased synaptic density and reduction in neuron size.

Changes in volume and function of white matter in various brain regions has also been

reported in non-demented, aged subjects and patients with cognitive impairment8. Significant age-

related decreases in white matter volume was noted in regions important for their communication

with the hippocampus.8,10-11 In support, anterior changes in white matter, and loss of integrity in

white matter tracts of the corpus callosum have been reported using diffuse tensor imaging (DTI).12

The pathological changes and consequent functional impairments seen in aging are accelerated in

diseases of cognitive impairment.

3

b) Mild Cognitive Impairment (MCI) is clinically and pathologically a unique,

early stage of dementia.

Mild cognitive impairment (MCI) is a clinical classification that was originally identified

and described by Petersen and colleagues in 1999.12 It marks an important phase of disease

progression that is generally considered a transitional state between normal cognitive function and

dementia.12,13 Individuals with MCI have been shown to convert to Alzheimer’s disease at a rate

of 5-20% per year. By comparison, the incidence rate of Alzheimer’s disease in the general

population of age-matched individuals is approximately 1-2%13. One of the original goals of

identifying this prodromal stage of cognitive impairment was to establish a window for early

therapeutic intervention in dementia patients that were specific to Alzheimer’s disease dementia13-

15. In the nearly two decades since its original conception, the definition of MCI has evolved and

expanded to include unique subtypes, thereby complicating the clinical and pathological definition

of MCI14. Importantly, these subtypes include heterogeneous patient population each with distinct

pathogenic pathways and the potential to progress to a variety of dementias.

MCI is classified into distinct subtypes based on a patient’s clinical presentation (figure 2).

A subject will be classified as either amnestic MCI (aMCI) or non-amnestic MCI (naMCI) based

on neuropsychological tests of cognitive function13,15,16. If an individual exhibits memory deficits,

they will be given a diagnosis of aMCI. If a subject shows reduced cognitive function in another

domain, (e.g. language, executive function) but does not test poorly in memory, they are classified

as naMCI 14. Individuals can be classified as having impairments in a single cognitive domain or

multiple domains.

4

In 2011, the National Institute on Aging-Alzheimer’s Association (NIA-AA) proposed

criteria to subdivide individuals into two distinct categories based on pathology using imaging and

fluid biomarkers: MCI-AD (MCI caused by Alzheimer’s disease) and MCI-sNAP (MCI suspected

non-Alzheimer disease pathology)17. Although MCI-sNAP subjects exhibit neurodegeneration

similar to AD subjects, they do not exhibit elevated levels of Aβ deposition as present in AD

brains16. In contrast, MCI-AD subjects have accelerated neuronal injury, degeneration, and

biomarker profiles that more closely parallel the levels seen in AD16 (figure 3).

Figure 2: Comparison of current criteria for mild cognitive impairment (MCI).

Source: Modified from Petersen RC, Caracciolo B, Brayne C, Gauthier S, Jelic V, Fratiglioni L. Mild

cognitive impairment: a concept in evolution. Journal of internal medicine. 275(3):214-228.

doi:10.1111/joim.12190. (2014)

5

Importantly, both amnestic and non-amnestic patients can be classified into either MCI-AD or

MCI-sNAP groups; this indicates that a clinical diagnosis does not necessarily predict the

pathology underlying the disease. Further, although the rates of conversion to AD are higher in

MCI-AD patients, both MCI-sNAP and MCI-AD individuals can progress to AD. This

underscores the idea that AD has a complex pathogenic mechanism.

c) Alzheimer’s disease is the most common cause of dementia in the global

population.

Dementia is a syndrome that affects memory, language and other cognitive skills severely

enough to impair an individual’s everyday functioning. This is a result of damage to neurons in

specific regions of the brain. Alzheimer’s disease is the leading cause of dementia, but it is not

the only one20. Several other types of dementia are commonly seen in elderly patients, including:

Figure 3: Suggested criteria for the likelihood that MCI is due to AD. The presence of Aβ42 can be detected

with PET and CSF analysis. The presence of neuronal injury can be detected using MRI. Low levels of Aβ42 and

elevated tau in the CSF with tau/ Aβ42 ratio ≥ 0.39 indicates progression to AD, as does a low ratio of Aβ42 to tau

in the CSF

Source: Modified from Petersen, R. C. Mild cognitive impairment. New England Journal of Medicine, 364(23),

2227-2234. (2011).

6

vascular dementia, Lewy body dementia, and Fronto-temporal dementia among others19. Many

individuals who are diagnosed with one form of dementia may have pathology and symptoms of

a secondary form, resulting in wide varieties of mixed dementia20-22. This mixed pathology

complicates research models as well as the successful diagnosis and treatment of patients.

Regardless of this complication, the overwhelming data indicates that a majority of dementia

patients are diagnosed with AD as the primary form of dementia20. Further, this number is

projected to increase in the coming decades.

I) Alzheimer’s disease is a public health epidemic.

Increasingly larger percentages of the global population are living longer than previous

generations21. The total number of people over 60 years old is estimated to become 22% of the

world’s population by 2050 with the number of people afflicted with some form of dementia

doubling every 20 years21,23 (figure 4).

Figure 4: Projected Populations of AD. The projected number of people age 65 years and older (total

and by age group) in the U.S. population with Alzheimer's disease, 2010 to 2050.

Source: Alzheimer's Association. 2016 Alzheimer's disease facts and figures. Alzheimer's &

Dementia, 12(4), 459-509. (2016).

7

Due to this increase in the cognitively impaired patient population, the incidence rate of

AD is expected to increase proportionally. It is estimated that by the year 2050, 13.8 million

individuals in the United States of America alone will have AD dementia24. The estimated cost

of care for AD patients is expected to approach $221 billion nationally21. These conservative

financial estimates underscore the urgent and significant need for early and reliable diagnosis,

effective treatment and management of the economic burden as the AD patient population

continues to grow.

II) Diseases of cognitive impairment are clinically assessed using a diagnostic

screening tool.

The Folstein Mini Mental State Exam (MMSE) is a widely used neuropsychological

assessment tool in the Western hemisphere. It is used by physicians and other clinical

professionals to evaluate changes in an individual’s cognitive function over time25. The DSM

recommends that physicians administer this test to patients who are 65 years and older and

indicate/exhibit a possibility of dementia26. The MMSE is a paper based 11 item exam with a

maximum possible score of 30. Subjects answer questions on different topics ranging from

comprehension, memory recall and attention. A score in the range of 24-30 is considered normal

while a lower score indicates some degree of cognitive impairment27. One of the major limitations

of this screening tool is it exhibits a ceiling effect. That is, individuals who fall into the pre-

dementia phase of cognitive decline will not be effectively diagnosed as at risk using this exam

because they will fall into the upper “normal” range27.

The Montreal Cognitive Assessment (MoCA), another frequently used assessment tool

that addresses the MMSE’s issue of poor sensitivity, especially in the earlier stages of cognitive

8

impairment29. Similar in format to the MMSE, the MCoA is an 11 item, 30 point test. However,

it includes more challenging questions that focus on higher level functioning such as visuospatial

processing, language and executive functions.29 This more thorough interrogation of executive

function may be crucial in discerning MCI from healthy cognitive function as well as from normal

cognitive aging.27-30 However, in some cases, MCI patients do not exhibit clinical symptoms.

Further, a varying degree of subjectivity can make clinical diagnoses difficult. Therefore, other

methods are used to measure pathological changes for detection and diagnosis of diseases.

III) Disease pathogenesis is measured and monitored using various detection

methods.

MCI was designated as a unique phase of cognitive impairment by Petersen and colleagues

1999, with the intention of providing an opportunity for early therapeutic intervention12. Indeed,

changes in specific pathological traits associated with AD begin during progressive mild cognitive

impairment (MCI) or earlier. To monitor disease progression as well as determine disease

prognosis, researchers and clinicians use a combination of different neuroimaging and fluid

biomarker methods (figure 5). In addition, neuropsychological testing to assess clinical symptoms

is conducted to correlate clinical and pathological symptoms in subjects. Neuroimaging

techniques, which are less invasive than CSF biomarkers, include structural MRI, functional MRI,

and PET imaging using the (11)C-labelled Pittsburgh Compound-B to determine the levels of

amyloid deposits. The most commonly used fluid biomarkers are cerebrospinal fluid (CSF) levels

of Aβ42, total tau (t-tau) and phosphorylated tau (p-tau)31.

9

AD causes region-specific cortical thinning and atrophy, specifically, in the hippocampus

and cortex32. To measure these changes, structural MRI is used to assess volumetric changes and

loss in a progressive, longitudinal manner. Similarly, functional changes are known to occur as a

result of impaired synaptic function. This results in decreased metabolic activity which is assessed

using functional MRI32. Amyloid beta deposition in the brain is assessed using PET imaging31.

The Pittsburgh Compound used in PET imaging to detect levels of Aβ deposition has been used in

the largest number of patients to date31,32.

Figure 5: A hypothetical temporal model of Alzheimer's disease biomarkers. The threshold for the first detection

of biomarkers associated with pathophysiological changes is indicated by the black horizontal line. The area below

the detection threshold is the zone in which abnormal changes lie. In this model, tau pathology can precede Aβ

deposition in time, but only in the early stages of the disease. Aβ deposition occurs independently and rises above the

biomarker detection threshold (purple and red arrows). This accelerates tauopathy, and CSF tau then rises above the

detection threshold (light blue arrow). Later, changes in PET and MRI (dark blue arrow) rise above the detection

threshold. Finally, cognitive impairment becomes evident (green arrow), with a wide range of cognitive responses that

depend on the individual's risk profile (light green‐filled area). Note that while CSF Aβ42 alteration is plotted as a

biomarker (purple), this represents a decrease in CSF Aβ42 levels and is a surrogate for an increase in parenchymal

Aβ42 and changes in other Aβ peptides in the brain tissue. Aβ, amyloid β‐protein; FDG, fluorodeoxyglucose; MCI,

mild cognitive impairment.

Source: Selkoe, D. J. and Hardy, J. EMBO Mol Med. 8, 595-608 (2016)

10

Since CSF is in direct contact with neurons that house and eventually expel the hallmark

pathological features of AD, the levels of Aβ and tau (p/total) in CSF functions as a barometer of

neuronal damage and neuronal AD pathology. Several studies have shown that CSF Aβ42 levels

in patients with AD were reduced compared to age-matched cognitively normal controls31-33. This

is due to amyloid peptide aggregation and plaque formation in the brain, resulting in a decreased

Aβ42availability in the CSF. Hence, there is an inverse relationship between CSF Aβ42 in AD and

disease progression. Total tau levels in CSF have been shown to be three times higher in AD

patients compared to age-matched controls. However, because there are many neurodegenerative

diseases with tauopathies, this measure of AD disease progression is nonspecific33. In contrast,

phosphorylated-tau (p-tau) protein is an extremely specific biomarker of AD. CSF p-tau levels in

non-AD diseases, including tauopathies, are normal compared to AD.

d) Alzheimer’s Disease has a complex, multifactorial pathogenesis.

AD, an extremely complex chronic neurodegenerative disease that is pathologically

characterized by progressive neuronal death, accumulation of Aβ42-rich amyloid plaques and p-

Tau containing neurofibrillary tangles 29. While it has been consistently shown that AD patients

have markedly deteriorated cognitive functioning beyond their age, there is no clear relationship

between the progression of pathological changes and clinical symptoms. The conclusive AD

diagnosis can only be made with pathological markers in the postmortem condition that supports

clinical symptoms36. The etiology and pathogenesis can vary between subjects due to a variety of

factors and can be attributed to changes in a several different signaling pathways that have been

11

shown to alter the course of disease progression, which further complicates attempts to

characterize the disease.

Advancing age is an essential and necessary risk factor for AD37. One well characterized

risk factor that occurs in aging and neurodegeneration is oxidative stress. Due to an imbalance in

redox states, reactive oxygen or nitration species accumulate along with increasing age to hasten

AD pathogenic progression38-40. Another important signaling pathway that is related to AD

disease progression is insulin signaling. Insulin receptors are widely distributed throughout the

brain41. Increasing age is reported to be associated with a decrease in central insulin receptor

signaling (i.e. IR, IRS-1, Akt) termed insulin resistance42. Comparing to age-matched non-

demented subjects, insulin resistance in the brain is more severe 42. The dysfunctional insulin and

insulin-like growth factor (IGF-1) signaling pathways are integral components of the complex

AD pathogenic mechanisms. Dysfunction of the APP processing pathway, which results in

amyloid beta plaque generation, is central and critical in AD pathogenesis. This pathology, in

addition to the dysfunctions in the insulin signaling pathway, increased oxidative stress, and

immune dysfunction collectively contribute to disease progression.

I) Amyloid Beta Plaque Pathology and the Amyloid Hypothesis.

The amyloid hypothesis proposes that a dyshomeostasis in Amyloid-beta (Aβ) protein is

a key pathogenic event in AD43. This imbalance is caused by a combination of increased Aβ

production and/or a failure in Aβ clearance mechanisms.

12

Amyloid beta peptides are created through the cleavage of the amyloid precursor protein

(APP) in a sequential series of enzymatic cleavages. APP is a member of an evolutionarily

conserved protein family that includes APP, APLP1 and APLP244. It is sequentially cleaved in

either the canonical pathway or the amyloidogenic pathway to generate P3 or Aβ peptides (figure

Figure 6: APP processing occurs via either the Amyloidogenic or canonical pathway. APP initially undergoes

cleavage at either α - or β-secretase sites to release sAPPα or sAPPβ, respectively. This allows γ-secretase to

cleave the membrane-embedded fragments . Proteolysis by γ-secretase promotes the extracellular release of the

amyloidogenic Aβ fragments () in the toxic pathway whereas proteolysis by γ-secretase generates an N-terminally

truncated and non-toxic 3 kDa P3 peptide in the non-toxic pathway.

Source: Fraering, P. C. Structural and Functional Determinants of γ-secretase, an intramembrane protease

implicated in Alzheimer's disease. Current genomics, 8(8), 531-549. (2007).

13

6). In the non-toxic, canonical pathway, α-secretase makes an initial cleavage which generates

secreted APP α (sAPPα), a large extracellular fragment that is released into the extracellular

medium, and the C-terminal fragment (CTF) that remains in the membrane. The α-CTF is cleaved

subsequently by the γ-secretase complex, a group of enzymes which includes presenilin 1 or 2

(PS1 and PS2) together with other enzymes. Significantly, the cleavage site of APP by α-secretase

lies within the Aβ sequence and thus prohibits Aβ peptide production. In contrast, cleavage of

APP by β-secretase mediates the first step in the amyloidogenic pathway. This causes the release

of a large sAPPβ fragment into the extracellular medium and a longer β-CTF that remains in the

membrane. The β-CTF fragment is then cleaved by γ-secretase. This successive cleavage of β-

CTF leads to the generation of Aβ peptides with 38-43 amino acids that are secreted into

extracellular space43.

Most Aβ peptides are 40 residues in length (Aβ40), however, a small percentage contain

42 residues (Aβ42). Increased extracellular accumulation of toxic Aβ species, particularly Aβ42,

promotes the formation of Aβ oligomers. As the amount of both monomeric and oligomeric forms

of Aβ overwhelm the brains capacity to remove and degrade the toxic Aβ species, they form

extracellular plaques. Ultimately, through activation of various different cellular mechanisms

including inflammatory cascades, the neurons will become damaged and die.

Since its original proposal several decades ago, this amyloid hypothesis has been met with

great skepticism due to its inability to fully explain the pathology seen in AD and failure to

reconcile discrepancies seen between cognitive impairment and degree of Aβ deposits since

cognitive deficits correlate better with soluble Aβ levels45-48. An alternative amyloid pathogenic

theory that indicates soluble Aβ monomers, dimers or small oligomers bind the α7 nicotinic

acetylcholine receptors (α7nAChRs) with high affinity to elicit toxic signaling. The Aβ-

14

α7nAChR complexes are then internalized and accumulated, resulting in impaired transport and

ultimately lysis of the affected neurons to form plaques49-55. Although it remains incomplete, the

amyloid theory provides a broad framework to guide drug development and to gain insights into

the complex mechanisms of AD. Critically, the amyloid hypothesis is able to explain the fact that

all AD patients experience progressive Aβ plaque buildup in brain areas that are relevant to

memory and cognition, and that mutations of APP accelerate Aβ production56. Experimental

studies have shown that increased Aβ deposition may be the critical initiating step in the AD

pathogenesis cascade57-59.

15

II) Tau Hyperphosphorylation and the Tau Hypothesis

One of the other key pathogenic mechanisms in AD is tau hyperphosphorylation. Tau

belongs to the family of microtubule-associated proteins60. There are six isoforms of tau proteins

primarily found within neurons in the adult brain, created through alternative splicing60. The

primary function of tau is to monitor the assembly and stabilization of microtubules. Several

neurodegenerative diseases, collectively termed tauopathies, are linked to tau pathology. Among

Figure 7: Tau hyperphosphorylation and Tau pathology. In AD pathology, tau phosphorylation events occur

in a sequential series of steps.

Source: Johnson, G. V., & Stoothoff, W. H. (2004). Tau phosphorylation in neuronal cell function and

dysfunction. Journal of cell science, 117(24), 5721-5729.

16

them are frontotemporal dementia (FTD), Pick’s disease, progressive supranuclear palsy (PSP),

traumatic brain injury (TBI) and AD. Specifically, in these conditions, tau pathology is due to

dysregulated tau phosphorylation resulting in hyperphosphorylated tau61.

Hyperphosphorylated tau loses affinity for microtubules and dissociates from them. The

increased pool of free hyperphosphorylated tau after dissociation from microtubules is likely an

important first step to its aggregation in AD. Untethered from microtubules, hyperphosphorylated

tau twists together to form paired helical filaments (PHFs) that are found abundantly in

neurofibrillary tangles (figure 7). In a toxic gain of function, hyperphosphorylated tau also

actively disrupts microtubules and inhibits their assembly and even sequesters functional tau and

other microtubule-associated proteins. Hyperphosphorylation also changes tau’s localization

from axon-predominant to include dendrites, neuronal cell bodies and presynaptic areas, leading

to wide spread synaptic, dendritic and axonal dysfunction. In cultured cells, abnormal tau is

shown to convert normal tau into abnormal tau. This conversion mechanism has been

hypothesized to be a mechanism by which tau aggregates in particular neuron populations

propagate to other brain areas62-64. This spread of tau is correlated with disease progression

according to Braak staging, and is considered a later development in AD pathogenic disease

progression relative to Aβ plaque deposition36.

17

e) Early Neurodegeneration and dysfunction in Alzheimer’s Disease is

characterized by synaptic changes and loss of synapses. One of the earliest pathological manifestations of AD, is a dysfunction and loss of synapses

(figure 8). This change at the synapses, which has been investigated for several years and is

recognized as a significant transformation in the progression toward disease, begins decades before

Figure 8: The sequence of major pathogenic events leading to AD proposed by the amyloid

cascade hypothesis. Highlighted in red is a critical early event: compromise in synaptic function due to

Aβ oligomers, which may directly injure the synapses.

Source: Modified from Selkoe, D. J. and Hardy, J. EMBO Mol Med. 2016;8:595-608

18

the presentation of clinical symptoms of AD and continues throughout the course of the disease.

It has been well studied and established that dendritic spine morphology is modifiable and dictated

by functional changes occurring at the dendritic spines65. That is, the spine morphology change

according to the coordinated responses of the scaffold proteins, receptors and channels located at

these synapses to environmental signals. In the absence of any neurodegenerative disease, brain

changes in older adults, are largely due to decreases in synaptic density, rather than frank cell

death66,67. However, age-related synaptic loss is accelerated in brains of MCI and AD. In a

quantitative study measuring the density of synapses in the frontal and temporal cortices using

biopsied tissue, a significant decrease (25-36%) in the abundance of synapses was found in patients

who had been given a diagnosis of AD in the past 2-4 years. Additionally, there is an estimated of

15-35% synapse loss per cortical neuron in individuals with AD68. In another quantitative

assessment study using postmortem hippocampus from AD subjects, there was a 44% synapse

loss. In comparison, the age matched MCI group showed a statistically non-significant 13%

synapse loss. These were synaptic losses compared to the age matched NCI (no cognitive

impairment) group, which experienced no significant synapse loss67. Importantly, these

quantitative studies show that synapse loss correlates better with cognitive deficits as indicated by

MMSE scores than the overall density of plaques and tangles68,69. Together, these studies indicate

that synaptic destruction occurs early in the AD pathogenesis and contributes to functional deficits.

Thus, unraveling the underlying mechanisms for synaptic destruction can lead to more effective

treatments for AD.

19

f) The post-synaptic density of glutamatergic, excitatory synapses is a major site

for long term potentiation and depression.

The postsynaptic density (PSD) is a thick, multilayer compartment. It is a region of critical

protein-protein interactions that regulate LTP, LTD, and downstream signal transduction in the

postsynaptic cells. Altered PSD structure and molecular compositions are implicated in numerous

neurological diseases including AD. Synaptic plasticity is a broad term that describes long lasting,

activity-related strengthening or weakening of synaptic transmission. Such modifications in

synaptic strength enable consolidation and encoding of memories through coordinated changes in

various cellular mechanisms and signaling pathways that result in remodeling of dendritic spines

of pyramidal neurons in key brain regions such as cerebral cortex. These include the well-

established phenomena of long term potentiation (LTP) and long term depression (LTD).

I) LTP and LTD at glutamatergic excitatory synapses depend on NMDA and

AMPA receptor activity.

LTP was first demonstrated in 197370, and has been extensively studied since. The NMDA-

R dependent model of LTP is considered to be widespread in the CNS71, although other types of

LTP do exist (e.g. mossy fiber LTP)72. There are two phases of LTP: early phase (induction) and

late phase (consolidation). Early phase LTP (E-LTP) is initiated by an influx of Ca2+ ions through

NMDA receptors.73 Increased [Ca2+] leads to activation of Calcium/Calmodulin Kinase II

(CamKII). CamKII contacts the actin cytoskeleton through an unknown number of mediators and

modulators and results in changes in dendritic spine morphology (figure 9a and 9b).74-76

Concurrently, AMPA receptors are activated by kinase-dependent phosphorylation. LTP results in

an increase in surface level expression of AMPA-R receptors.77 The consolidation phase of LTP

is dependent on gene transcription for the synthesis of new proteins in order to maintain synaptic

strength. The duration of this phase can range from 60 minutes to days or weeks. Specifically, the

20

activation of transcription factor CREB, along with various signaling proteins and kinases have

been implicated in this late phase.71,75

Long term depression (LTD), similar to LTP, is widely expressed in the CNS.71 Like LTP,

induction of LTD can be dependent on a postsynaptic influx of [Ca2+] through NMDA-R.

Alternatively, in mGluR mediated LTD, the induction and changes in expression are due to surface

mGlu-receptors. Although these two different forms of LTD have different mechanisms, the

outcome of both is the same: a decrease in the amount of AMPA-R receptors expressed at the

postsynaptic membrane75,78. Several studies have shown that this dynamic fluctuation in surface

Figure 9: Mechanisms of LTP and LTD in the PSD.

(a) Weak activity of the presynaptic neuron leads to a modest depolarization and Ca2+ influx through NMDA receptors.

This preferentially activates phosphatases that dephosphorylate AMPA receptors, thus promoting receptor endocytosis.

Strong activity (which is paired with strong depolarization) triggers LTP in part via activation of CaMKII, receptor

phosphorylation, and exocytosis

(b) Structural changes associated with LTP and LTD. (1) Synaptic strength correlates with spine volume and the area

of the postsynaptic density (orange). Note that the PSD in potentiated synapses is often perforated. (B) LTP can also

lead to the appearance of new spines.

Source: Modified from Lüscher, C., & Malenka, R. C. (2012). NMDA receptor-dependent long-term potentiation and

long-term depression (LTP/LTD). Cold Spring Harbor perspectives in biology, a005710.

a)

b)

21

expression of AMPA-R is a result of receptor endocytosis mediated through various signaling

cascades. Several postsynaptic proteins have been implicated in this process and more molecules

remain to be determined. After induction by low-frequency stimulation, the increased [Ca2+] in the

postsynaptic cell activates calcineurin and protein phosphatase I, as well as various kinases (e.g.

CamKII, PKC) to ultimately decrease AMPA-R levels in the postsynaptic membrane75,77,78.

Importantly, this endocytic process of LTD, as well as the accompanying dendritic shrinkage

through cytoskeletal remodeling (figure 9), involve several postsynaptic proteins, including Arc,

implicated as important players in these processes of memory formation and encoding74,75,78.

II) Scaffolding Proteins PSD-95 and FLNA function as binding partners and

regulate receptors in key signaling pathways involved in memory formation.

The postsynaptic density (PSD) of glutamatergic, excitatory neurons is the site of a variety

of important protein-protein interactions. The PSD is multilayered, with an intentional

organization of molecules based on their functional role(s)79 (figure 10). Within this laminar

structure, each layer has different dynamic compositions. Proteins are organized to allow for

efficient downstream transduction of signals received from the presynaptic cell. The membrane,

or surface layer, contains transmembrane proteins such as NMDA-R and AMPA-R receptors.

NMDA-R activity at the PSD results in changes in AMPA-R concentrations seen in LTP and LTD.

In the cytoplasmic layer of the PSD, there are a host of proteins that interact with the cytoskeleton

to mediate dynamic changes in dendritic spine structure80. Among the most critical and abundant

proteins located throughout the PSD are scaffolding proteins. This specific protein class regulates

a host of signaling pathways. Specifically, they organize and stabilize various proteins and

receptors, such as NMDA-R and AMPA-R, into complexes in the PSD.

22

One critical scaffolding protein is PSD-95. Substantial data shows that PSD-95 is one of

the critical proteins involved in AMPA-R dependent changes at the PSD in response to synaptic

activity. Overexpression of PSD-95 selectively increased AMPA-R mediated EPSCs in

hippocampal neurons without detectable change in NMDAR-mediated EPSCs81-83. Conversely, in

a PSD-95 knockdown study using RNA interference (RNAi), there was a selective reduction in

AMPA-R mediated EPSCs84,85. These studies show that PSD-95 regulates the synaptic

transmission of AMPA-R mediated EPSCs. Although NMDAR-mediated EPSC is not influenced

by genetic manipulation of the PSD-95, PSD-95 is associated with and recruited to NMDARs upon

receptor activation86, 52-54. The functional importance of the NMDAR-PSD-95 linkage is

demonstrated in the conditions in which excitotoxicity has been elicited such as in stroke as

blockade of PSD-95 reduces infarct size87.

Figure 10: Proposed Molecular Organization of the PSD.

The synapse membrane is enriched with both AMPA and NMDA receptors, but mGluRs are

distributed in the perisynaptic membrane. The PSD is composed of multiple layers, with the

stable molecules localized in the surface layer (dark blue). The cytoplasmic layer (light green)

is enriched with dynamic proteins interacting with F-actin. CaMKII holoenzymes are also

enriched on the cytoplasmic face of the PSD.

23

Filamin A (FLNA), also known as actin binding protein-a, is another scaffolding protein

that is abundant in the PSD. FLNA was shown to be essential in cytoskeleton assembly and

rearrangement. The function of FLNA can be modulated by phosphorylation mediated by

serine/threonine kinases such as Pak1 found in the cytoskeletal compartment of the PSD88.

Dysfunction or altered conformation of FLNA is a prominent pathology of AD89. FLNA is

recruited to α7nAChR and toll-like receptor 4 (TLR4) upon soluble Aβ binding to α7nAChRs and

TLR4 complex. Thus Aβ interaction with α7nAChRs promotes Aβ toxic signaling toactivate

downstream kinases which hyperphosphorylate tau and ultimately the formation of the second type

of hallmark pathology seen in AD: neurofibrillary tangles. By activation TLR4, Aβ triggers

release of inflammatory cytokines to promote neuroinflammation. These critical proteins and

many others, including several immediate early gene proteins also located in the PSD, are crucial

in the synaptic functions that regulate neuron activity in memory formation.

III) Immediate Early Genes and their protein products function as a rapid

response mechanism to cellular stimuli.

Immediate early genes (IEGs), a category of genes expressed in response to stimuli, are

implicated in LTP and LTD because of their rapid and transient responses to synaptic activation88.

Various types of stimuli can provoke upregulation of IEGs. Increased neuronal activity via sensory

stimuli, pharmacologically induced seizure activity, and behavioral tasks such as the MWM all

induce IEG expression in neurons in a specific brain regions89-92-80.

The response of IEGs to neuronal activity can be seen in the PSD. Specifically, IEG protein

products (e.g. Arc and homer1a, H1a) are critical in the PSD signal response to neuronal stimuli.

Using a fluorescence in situ hybridization (FISH) experiment strategy, Arc and H1a RNA

24

transcription was induced in rat hippocampal and cortical neurons when rats were exploring a

novel environment. This result supports the idea that Arc and H1a in the PSD may influence long

term changes in synaptic plasticity93

g) Activity Regulated Cytoskeleton Associated Protein (Arc)

First discovered in 1995, Arc/Arg3.1 (activity regulated cytoskeleton associated) is a

vertebrate specific gene that is a member of the IEG family94. It produces Arc (Activity regulated

cytoskeleton associated), a single 55kDa protein that functions in the postsynaptic density and

nuclear regions of the neuron95. Arc’s mRNA is localized to dendrites in response to synaptic

activity from the nucleus and is subsequently locally translated in response to synaptic activity.

Significantly, studies have shown that Arc is expressed almost exclusively in excitatory,

glutamatergic neurons in the hippocampus and neocortex96. In support, visual cortex of the Arc

knockout mice (Arc-/-), was not affected by both sensory stimuli, or the absence thereof97,98. These

experiments indicate that Arc plays a critical role in experience-dependent synaptic plasticity in

the cortex. Due to this important, central function, a great effort has been dedicated to analyzing

Arc’s structure with the goal of improving our knowledge regarding Arc’s mechanisms of

action99,100. Arc, however, is more challenging to understand at the molecular level because it is a

single copy gene that does not have any known genetic homologues98.

One of the challenges to understanding the regulatory mechanisms involved in Arc’s

unique functions is the limited information available regarding Arc’s structure and binding sites.

In 2015, Zhang and colleagues demonstrated the first partial crystallization of Arc, however, full

length Arc has never been crystallized100. Beyond this crystallization study, mass spectrometry

and spectroscopy analyses indicated that Arc is likely to contain two lobar regions with

predominantly α-helical content that are linked by a disordered, flexible region99,100. Through

25

various research studies, several cytosolic, dendritic and nuclear proteins have been found to

interact with Arc via dedicated binding sequences in either the Arc C-lobe or N-lobe (figure 11).

Since the mechanisms through which Arc influences neurons have not been fully elucidated, Arc-

protein interactions, their dedicated binding sites and the functions of these interactions continue

to be investigated.

I) Synaptic Functions of Arc mediate AMPAR Regulation in LTP/LTD.

Arc protein plays a central role in both long-term potentiation (LTP) and long-term

depression (LTD) by acting in response to activity through interactions with several proteins in

the postsynaptic density94. Specifically, LTP increases Arc expression whereas LTD leads to

decreased local Arc translation and expression90. This is also supported by earlier studies that

show late phase LTP or the consolidation phase of LTP requires sustained Arc expression. Arc’s

role in LTP and LTD has been investigated in numerous studies, yet many questions still remain.

Figure 11: Graphic Representation of Arc domain structure. Shown are selected regions and known

binding sites with function. NRD, nuclear retention domain; NES, nuclear export signal; NLS nuclear

localization signal. Scale unit − amino acids.

Source: Modified from Nikolaienko, O., Patil, S., Eriksen, M. S., & Bramham, C. R. (2017,

September). Arc protein: a flexible hub for synaptic plasticity and cognition. In Seminars in cell &

developmental biology. Academic Press.

26

Experiments have shown that while Arc likely plays a role in early-phase LTP, it is critical

for late phase LTP99-101. When Arc antisense oligodeoxynucleotides (ODNs) were infused into

the hippocampi of rats before LTP induction, LTP consolidation was lost, but early-phase LTP

remained intact99. In another study by Messaoudi et al (2007), rats were infused with Arc

antisense ODNs 2h after HFS. The rats had a profound and permanent loss of late-phase LTP, but

not early-phase LTP112. These experiments indicate that Arc is essential and necessary for the

consolidation phase of LTP.

LTP has been shown to induce dendritic spine remodeling, a morphological change that is

linked to learning and memory formation114,115. Thus, Arc not only regulates synapse strength, but

may also play a role in sculpting dendritic spine morphology during LTP although the underlying

mechanisms remain to be fully elucidated. Through mechanisms and interactions that are not fully

understood, Arc is involved in cytoskeletal dynamics at the PSD. One protein that interacts with

Arc is cofilin, a postsynaptic protein that binds and severs filamentous actin (F-actin), thus

promoting the formation of actin monomers116,117. Cofilin is negatively regulated by

phosphorylation which is induced by synaptic activity (e.g. HFS)115. During late phase LTP in the

hippocampus of live rats, cofilin is phosphorylated during LTP and blocking cofilin

phosphorylation partially reduced late phase LTP, and, 115. In this same study, Arc AS infusion 2h

after LTP induction resulted in cofilin dephosphorylation and subsequent activation22. These

results indicate that Arc is necessary for stabilization of F-actin, and cofilin phosphorylation during

LTP. Other signaling cascades involving Arc and its role in LTP induced cytoskeletal changes

continue to be investigated.

27

In addition to its various, critical functions in LTP, Arc is also necessary for LTD. It has

been well established that AMPARs at the synapse are internalized via clathrin-coated vesicles

following LTD indiction118-120. In 2006, Chowdhury et al. showed that Arc mediated this AMPAR

trafficking via its interactions with the accessory proteins endophilin and dynamin118. Specifically,

Arc affected synaptic activity by interacting with endophilin 1 and dynamin 2 to promote AMPAR

endocytosis (figure 12). Arc mutants with altered binding sites for either endophilin or dynamin,

significantly reduces surface GluR1 levels118. This indicates that Association of Arc with both

endophilin 1 and dynamin 2 is a necessary regulator of the endocytosis of AMPARs during LTD.

Figure 12: Arc has multiple effects through its actions in the synapse and nucleus.

Interactions with various cellular proteins mediate Arc’s functions in the synapse and nucleus to affect

LTP and LTD. Arc is shown to form a complex with Endophilin and Dynamin to regulate AMPAR

endocytosis in LTD. It has also been shown to interact with βSpectrinIV and PML-NB in the nucleus,

although the exact function of this interaction is unclear. LTP; long term potentiation. LTD; long term

depression. PML-NB; promyelocytic leukemia nuclear bodies.

Source: Korb, E., & Finkbeiner, S. Arc in synaptic plasticity: from gene to behavior. Trends in

neurosciences, 34(11), 591-598. (2011).

28

Arc expression is induced when group 1 mGluRs are activated. This leads to endocytosis

of the AMPARs in a process termed mGluR-LTD. Overexpression of the Arc protein in neurons

causes a reduction in surface expression of certain types of GluA subunits and both mGluR- and

NMDAR-mediated LTD121. Exactly how Arc regulates the endocytic machinery is unclear, and no

direct evidence shows that low frequency stimulation causes Arc to localize to endophilin and

dynamin. To fully explain how Arc can associate with and regulate both LTP and LTD, additional

research is needed to better understand the cellular functions of Arc and how they are regulated by

different stimuli. Together, these studies suggest that Arc expression can be utilized to gauge the

changes in the synaptic activity and dendritic dynamics in the diseased brain tissues.

II) Nuclear functions of Arc

Although the majority of Arc’s cellular impact is exerted through its role at the synapse,

nuclear expression of Arc is also critical and increases in response to stimuli 102, 103. Arc gene

expression is regulated by several second messengers (e.g. kinases) that amplify signaling

downstream of synaptic activity. Arc transcripts appear within 5 minutes of neural activity, hence

it’s classification as an IEG.106

Through its effects inside the nucleus, Arc also exerts a cell-wide change that is implicated

in homeostatic plasticity. This critical, regulatory mechanism functions to balance the effects of

synaptic plasticity (i.e. LTP and LTD). Through homeostatic scaling, a neuron is able to adjust its

response to long-term changes in activity, so that it can maintain the same firing rate. In

experiments of Arc KO or overexpression, this scaling ability was shown to be compromised110.

Arc translocates to the nucleus hours after activity induced translation at the synapse. There is a

direct, positive correlation between time and the ratio of nuclear to cytoplasmic Arc in response to

stimuli. Arc is localized to the nucleus via phosphorylation by the MEK-ERK signaling

29

pathway.104,109 Once inside the nucleus, Arc is involved in a series of actions that have been shown

to be responsible for Arc’s critical function in homeostatic scaling. In cultured hippocampal

neurons, Arc co-localizes with βSpIVΣ5, a spectrin scaffolding protein107 (figure 12). The

association of both Arc and βSpIVΣ5 (and neither Arc nor βSpIVΣ5 alone) in nuclear puncta

results in increased promyelocytic leukemia nuclear bodies (PML-nb), a known transcription

regulator108. Further, it has been shown that Arc acts indirectly through PML-nb’s to reduce GluA1

transcription resulting in a decreased surface level expression of GluA1103,107.

III) Regulatory mechanisms of Arc: posttranslational modifications

The majority of eukaryotic proteins undergo some form of post-translational modification

(PTM), a processing event that results in chemical changes to proteins after translation. Generally,

PTMs occur through proteolytic cleavage or the reversible addition of a modifying group (e.g.

acetyl, phosphate) to a specific sequence of amino acids within the protein target. Some of the key

regulatory functions of PTMs include altering protein conformation for targeting and localizing to

subcellular regions, activation, and degradation123. The role of PTM in Arc function and its protein-

protein interactions is of particular interest because Arc is a dynamic protein: it binds to several

Figure 13: Sites of Post-translational modifications on Arc. Several residues within the Arc protein

have been identified as target sites for key post-translational modifications. Among them are

phosphorylation (P), SUMO1-ylation (S1) or ubiquitination (Ub). ? indicates predicted target sites.

Source: Carmichael, R. E., & Henley, J. M. Transcriptional and post-translational regulation of Arc in

synaptic plasticity. In Seminars in cell & developmental biology. Academic Press. (2017).

30

proteins and has versatile functions that change based on cellular location and neuronal

conditions103,105,111,118. These characteristics point to the high probability of PTM involvement in

directing Arc localization, function, and other aspects of Arc regulation as well as sites of

dysfunction in diseases (figure 13). However, PTMs of Arc have mostly not been revealed.

SUMOylation, an important PTM used in cellular regulation of protein localization and

function, is the addition of an approximately 10kD protein from the small ubiquitin-like modifier

(SUMO) family of proteins124. Arc has been shown to be a substrate of SUMO, with two

SUMOylation sites125. Overexpression of a double mutant for both SUMOylation sites in cultured

hippocampal neurons prevented Arc’s localization patterns to the expected subcellular regions126.

When LTP was induced by activity in rats, SUMOylated Arc accumulates in large amounts to the

synaptoneurosomal and cytoskeletal fraction of the dentate gyrus127. These results show that newly

synthesized Arc likely requires modification by SUMO to be targeted to the actin cytoskeleton in

the consolidation phase of LTP.

Arc is a tightly regulated protein. It is unsurprising, then, that the PTM type that has the

most identified consensus sequence sites on Arc to date are ubiquitination sites. In a 2014 study,

researchers showed that Triad3A, a ubiquitin ligase, ubiquitinates Arc and induces its proteasomal

degradation. In a series of RNAi experiments, Triad3A loss-of-function analysis showed a

decrease in Triad3A by 75%, with a subsequent increase in Arc accumulation. When Triad3A was

overexpressed, Arc levels were reduced and this led to an increase in the level of synaptic AMPA

receptors128. Experimentally, Arc is also ubiquitinated by ubiquitin E3 ligase, Ube3A94,129. RNAi

knockdown of Ube3A experiments result in elevated Arc protein levels129. Like Triad3A,

overexpression of Ube3A leads to a decreased Arc-mediated AMPAR endocytosis.

31

The most common type of PTM in eukaroytes is phosphorylation, as it has been well documented

that phosphorylation is a key regulator of protein activity (i.e. kinase activation). Phosphorylation

of key proteins is a crucial component of the pathways between synaptic activity and cellular

responses in various transduction mechanisms, including during Arc transcription and

translation104,106. While four phosphorylation sites have been identified based on consensus

sequence, direct phosphorylation of Arc has not been directly demonstrated. Arc has been shown

to interact with calcium/calmodulin-dependent kinase CamKII in a method termed inverse

synaptic tagging, thus indicating that there is a phosphorylation site for CamKII130. In addition,

there are putative phosphorylation sites for protein kinase C (PKC) and casein kinase II (CKII)38.

However, phosphorylation at all of these sites and possibly other potential phosphorylation sites

remain to be verified or identified. More importantly, the function of phosphorylation of Arc must

be elucidated to fully appreciate the role of Arc in the dynamic regulation of PSD and the dendritic

field in health and disease.

h) Arc expression and function is altered in disease

A balance between excitation and inhibition are critical to proper, overall neuronal

function. Therefore, it is unsurprising that Arc, a critical regulator of synaptic function, has been

implicated in a large number of neurological disorders. More specifically, a wide range of

neurological diseases, including schizophrenia, AD, and autism spectrum disorders (ASD) among

others have pathological manifestations that include dendritic spine abnormalities132,133. These

include dysfunctional actin and cytoskeletal dynamics132,133. Despite the fact that there are no

known Arc mutations that are directly implicated in disease conditions, many animal models of

disease have shown altered Arc expression95. Indeed, Arc itself was discovered during a search for

genes that were expressed in an individual after a seizure, indicating a role for Arc in epilepsy.

32

Since its discovery, research into Arc has shown that it may be a central protein in a variety of

other diseases. In animal models of major depressive disorder, Arc expression is significantly

increased upon exposure to chronic stress134. Changes in Arc mRNA are seen in the PFC after even

a single exposure to addictive drugs, as Fumagalli and colleagues showed in a rat model. This may

indicate a significant role for Arc in addictive behaviors138. In schizophrenia, genes encoding

proteins that are associated with Arc have been found through genome wide association studies

(GWAS) and other large scale analyses139. Arc and its protein complexes are of interest in

schizophrenia because NMDAR hypofunction and downstream signaling dysfunction may be

etiologically important in this disease86.

I) Cognitive disorders, aging and AD

Arc dysfunction has been strongly implicated in diseases with cognitive defects. The

neurodevelopmental disease Angelman syndrome is characterized by neurodevelopmental delays

including speech impairments resulted from a mutation in Ube3A129,136. Mutations and copy

number variants for Ube3A can also result in ASDs. Mouse models of ASD with altered Ube3A

indicate decreased levels of Arc along with elevated Ube3A due to Ube3A’s modulatory role in

Arc protein degradation136.

Fragile X Syndrome (FXS) is the most common inherited form of mental retardation140.

The FMR1 gene, which produces the FMRP protein, has been experimentally shown to be

responsible for FXS. FXS is pathologically associated with elevated levels of mGluR-LTD and

overexpression of several proteins including Arc. This abnormally heightened Arc expression

leads to increased endocytosis of AMPARs and increased mGluR-LTD. importantly, altered

dendritic spine morphology is noted in animal models of FXS and humans with this disorder.

33

While the role for Arc in diseases with synaptic pathologies and brain dysfunction in these

neurodevelopmental disorders is postulated, little is known about the changes of Arc during normal

aging and age-related disorders with synaptopathy such as AD. More importantly, it remains

elusive whether change in Arc is the result or the cause of dysfunction and loss of synapses in the

disease progression in aged and AD brains. Although milder in magnitude, some AD pathologies

can be found in brains of non-demented elderly144. In support, brains of the older wild-type mice

that showed reduced synaptic function as indicated by the attenuated receptor function also

accompanied by elevated basal but diminished stimulated Arc expression54. Among various

pathogenic factors, soluble Aβ even when added exogenously can inhibit activity-dependent Arc

expression54. Addition of Aβ oligomers (Aβo) at µM or near μM range to neurons inhibits LTP

and directly induces LTD, spine loss, and eventual cell death94,95,145. Arc has been shown to be

directly involved in production of the Aβ peptide through its association with endosomes that

contain the secretases BACE1 and presenilin that are responsible for the cleavage of APP into

Aβ146. Several investigative studies have also shown reciprocal relationships between Aβ and Arc.

Wu et al. showed that the number of amyloid plaques were reduced in a mouse model of AD when

Arc was deleted65. In human AD tissues, Arc protein levels are elevated146. When soluble Aβo are

added directly to cultured neurons, an increase in Arc protein level is accompanied by a lengthened

but reduced number of dendritic spines. In a transgenic mouse model of AD (hAPPFAD), Arc

mRNA expression was shown to be reduced in the hippocampus together with behavioral

deficits147. Moreover, a study investigating changes in Arc expression due to age showed a

reduction in Arc expression induced by spatial exploration in granule cells in the hippocampus143.

Although these data link Arc to spine morphology and behavioral abnormalities during aging and

in AD, the cause-effect relationship during AD pathogenesis is still not clear.

34

This proposed study therefore aims to fill the knowledge gaps by systemically investigating

the regulatory mechanisms on Arc during normal aging and in AD. Using varying ages of wild-

type and 3x Tg AD mice, the age-dependent and AD effects on non-stimulated (basal) and activity-

dependent Arc expression were determined in both prefrontal cortex (PFC) and hippocampal

formation (HF). To gain insight into the regulatory mechanism of Arc and whether the regulation

of Arc alters during normal aging and in AD, we assessed the phosphorylation and nitration of Arc

under basal and receptor-stimulated conditions in both brain regions from wild-type and 3x Tg AD

at various ages. The inter-relationship of Arc with various signaling complexes and the kinases

that mediate Arc phosphorylation were also elucidated and confirmed using enzyme inhibitors,

The relevancy of the data obtained in mouse AD model to AD pathogenesis was further determined

using postmortem hippocampal formation from 8 sets of age- and gender-matched non-demented

controls, AD as well as MCI-AD and MCI-sNAP cases.

35

Experimental Procedures

Human Postmortem Hippocampal formation sections

This study protocol conformed to the tenets of the Declaration of Helsinki as reflected in a

previous approval by the City College of New York and City University of New York School of

Medicine human research committee. De-identified neurologically normal controls and patients

with diagnosis validated MCI-sNAP, MCI-AD and AD cases were obtained from NIH-sponsored

brain banks: the RUSH Alzheimer’s disease center (Chicago, IL), the University of Pennsylvania

Brain Tissue Resource Center (Philadelphia, PA), the Harvard Brain Tissue Resource Center

(HBTRC, Belmont, MA) and the UCLA Brain Tissue Resource Center (UBTRC, Los Angeles,

CA).

Description of case cohorts

The AD cases from University of Pennsylvania were from a subset of a cross-sectional

study of AD pathology. The MCI cases obtained from RUSH Alzheimer’s disease center were

part of a longitudinal epidemiological and clinico-pathological study of aging called Religious

orders study (ROS) and Memory and Aging Project (MAP), including MCI and AD. ROS cases

are elderly Catholic clergy (nuns, priests, and brothers) who enter the study without signs of

cognitive impairment. MAP is a community based aging study. All ROS and MAP subjects were

examined annually with full clinical and neuropsychological evaluations and donate their brains

upon death. The follow-up rate for annual testing is above 95%, and the autopsy rate exceeds

90%148, 149. These MCI subjects were selected randomly from the 350 deceased ROS subjects who

had validated MCI diagnosis and were similar in age (< 5 year), postmortem interval and gender

with control and AD cases. None of the cases included have history of diabetes, stroke or

36

psychiatric disorders. There was no evidence of vascular anomalies in the hippocampal formation

of any case used in the current study. Diagnoses of AD and MCI studied met clinical criteria for

that disorder specified by NINCDS-ADRDA150 based on the assessments in the consensus

conferences after review of medical records, direct clinical assessments, and interviews of care

providers. Clinical diagnosis stipulates that an individual showed clear cognitive decline from his

or her previous levels as verified in the memory tests with at least one other cognitive domain (e.g.,

perceptual speed). The diagnoses were confirmed by postmortem examination of neuritic plaque

densities in midfrontal gyrus (dorsolateral prefrontal cortex), superior + inferior temporal gyrus,

inferior parietal gyrus, hippocampus, and substantia nigra as specified by the Consortium to

Establish a Registry for AD151. The final diagnoses were consistent with Braak scores for

neurofibrillary tangle (NFT) pathology as recommended by the NIA-Reagan Institute consensus

on diagnosis of AD152. Diagnostic neuropathological examination was also done in all tissues

using fixed sections stained with hematoxylin and eosin and with modified Bielschowsky silver

staining153 to establish any disease diagnosis according to defined criteria152. The presence of both

amyloid plaques and NFTs in all AD brains was also confirmed by Nissl and Bielschowsky

staining and characterized immunohistochemically with anti-Aβ42 and -NFT staining in both

frontal and entorhinal cortex as well as hippocampus, as described previously49, 51. Control tissues

typically exhibited only minimal, localized microscopic neuropathology of AD (0 –3 neuritic

plaques/10% field and 0 – 6 NFTs/10% field in hippocampus).

Diagnosis of MCI 154,155was purely clinical and indicates that an individual was rated at the last

examination as cognitively impaired according to neuropsychological tests. A diagnosis of

amnestic MCI (aMCI, MCI-AD) indicates that the individual displayed prominent deficits in

episodic memory at the final evaluation. As this implies, a diagnosis of non-amnestic MCI (naMCI,

37

MCI-sNAP) indicates that the individual displayed predominant cognitive deficits other than

memory (i.e., perceptual speed and/or visuospatial ability) at the last evaluation. Cognitive Testing

Yearly evaluations of the ROS subjects include neuropsychological testing, as well as completion

of a medical history, neurological examination, and ratings on psychiatric scales. The average time

between the last neuropsychological evaluation and death is 6-7 months. As previously

described156,157, such evaluation included the MMSE and 7 tests of episodic memory, 4 of semantic

memory, 4 of working memory, 2 of perceptual speed, and 2 of visuospatial ability. For data

reduction in each subject, raw scores on individual tests were converted to z scores relative to the

baseline mean and standard deviation for the entire ROS cohort. These were averaged to yield

composite scores on the cognitive domains noted above (e.g., episodic memory), which were in

turn averaged to yield a composite global cognition score.

Tissue Collection Autopsy consent was obtained from the brain donors, next-of-kin, or legal

guardians in all cases. Postmortem cases were stored at 2-4°C until autopsy. After sagittal bisection

of the forebrain, the brain was cut into coronal slabs. One hemisphere was sampled for tissues to

be examined microscopically, including the cerebellar cortex, HF, and other brain areas used for

diagnostic neuropathological assessments indicated above. Samples were fixed in neutral-buffered

formalin for 24-48 h, and embedded in paraffin. The remaining tissue was frozen overnight at -

80°C and sealed in plastic bags for long-term storage at -80°C. HF tissue were later dissected from

the frozen hemispheres of 8 matched pairs of normal and AD cases for the ex vivo stimulation

with, NMDA/glycine, insulin and BDNF as described below. Surfaces were shaved before thawing

to remove oxidized surfaces. Based on the stimulating postmortem studies in rodents and

postmortem brain studies that tested the effect of PMI on ex vivo stimulation paradigm86,51, 158, the

postmortem time intervals for collecting these brains were <12 hr. Of the selected cases in the

38

current study, the mean postmortem intervals for collection of control, MCI-AD, MCI-sNAP and

AD brain samples were 6.8 ± 0.6hr, 5. 9 ± 0.6 hr, 6.6 ± 0.5 hr and 6.3 ± 1.0 hr, respectively). One-

gram blocks from hippocampal formation were dissected using a band saw from fresh frozen

coronal brain sections maintained at -80°C. All experiments were performed as a best-matched set

that consist of control-MCI-AD, MCI-sNAP and AD without knowledge of clinical information.

Postmortem interval (PMI) is defined as the period of time between death and when the tissue

is frozen1. Previous research149,150 has shown how varying PMD can affect tissue integrity.

Specifically, DNA, RNA and protein expression are known to be compromised as temperature and

PMD increase. For each cohort, a total of n = 8 samples were selected.

3xTg AD Mouse Model Tissue Preservation and Selection

While AD is a uniquely human disease, animal models are useful for investigating specific

pathologic mechanisms during aging and diseases of cognitive impairment151.

3xTg AD mutant mice, also known as the LaFerla mouse, were obtained from the LaFerla lab

at University of California at Irvine152. This mutant strain harbors 3 mutations that facilitate the

production of AD pathological markers: APPswe mutations result in a substitution of lysine and

methionine with arginine and leucine, respectively, leading to an increased production of Aβ152.

The second mutation is PS1M146V which changes methionine to valine at exon 5 that also promotes

Aβ42 secretion. The third mutation is MAPTP301L. This mutation accelerates the pathology of tau

helical filaments seen in AD although tau pathology in AD is not due to a mutation but the result

of hyperphosphorylation and consequent loss of function. The 3xTg AD mouse exhibits age-

dependent changes that partially resembles some changes seen in human AD although there is little

neurodegeneration until at least 12-month of age152.

39

PFC and HF were collected from wild-type E129 (from Taconic in Germantown, NY) and

3xTg AD mice to evaluate under basal and ex-vivo stimulation conditions. For the 3xTgAD mice,

mice were sacrificed at 6-, 10-, and 15-months of age (n=5). For the wild-type E129 mice, tissue

samples were collected at 4-, 6-, 10- and 15-months of age. In the earlier reports, Aβ plaque

pathology has been shown to develop at 6 months of age in the 3xTg AD mouse model, with tau

pathology observed at 10 months or older4,54. The 4-month-old E129 mice were used as the

references for age-related changes. In all experimental series, WTs were assessed simultaneous

with age-matched 3xTg AD mice. .

a) Ex-Vivo Stimulation

For in vitro assessments, postmortem tissues were gradually thawed (from -80°C to -

20°C), sliced using a chilled McIlwain tissue chopper (100μm X 100μm X 3 mm) and suspended

in ice-cold oxygenated low Mg2+ K-R (LMKR), containing (in mM): 25 HEPES, pH 7.4, 118

NaCl, 4.8 KCl, 1.3 CaCl2, 1.2 KH2PO4, 0.3 MgSO4, 25 NaHCO3, 10 glucose, 100 μM ascorbic

acid and protease inhibitor cocktail (Roche). After centrifugation and two additional washes with

1 ml of ice-cold LMKR, brain slices were suspended in 1 ml of LMKR.

Using a well-established ex vivo stimulation method139, human postmortem HP from non-

demented controls, MCI-sNAP, MCI-AD, and AD cases were immediately suspended in ice-cold

oxygenated LMKR with protease inhibitors, washed and distributed into 5 tubes each with

approximately 5 mg. Each was incubated at 37°C with either Kreb’s-Ringer (K-R), NMDA (10

μM)/glycine (1 μM) for 10 min, 1 nM insulin for 30 min, 50 ng/ml BDNF for 30 min or PNU

282987 (PNU282987 is a potent and selective agonist for α7nAChRs) for 10 min. In the cases of

NMDA/glycine and PNU282987, slices were briefly centrifuged (at 25°C) and re-suspended and

40

incubated in LMKR for an additional 20 min. The reaction was terminated by addition of protein

phosphate inhibitors, diluting with ice-cold Ca2+-free LMKR and centrifugation at 4°C.

Similarly, slices of HF and PFC derived from WT* and 3xTg AD mice at age 4-, 6-, 10-

and 15- months (n=5) were prepared using a chilled McIlwain tissue chopper (100μm X 100μm X

3 mm) and suspended in ice-cold oxygenated LMKR containing protease inhibitor cocktails ,

washed and distributed into 3 tubes (approximately 2.5 mg/tube) each was incubated at 37°C with

either LMKR, NMDA (10 μM)/glycine (1 μM) for 10 min followed by a 20-min incubation with

LMKR or 1 nM insulin for 30 min. The reaction was terminated by addition of protein phosphate

inhibitors, diluting with ice-cold Ca2+-free and centrifugation at 4°C.

*The 4 month cohort had WT tissue only, there was no 3xTgAD matched samples.

b) Immunoprecipitation and co-immunoprecipitation

These assessments used previously described co-immunoprecipitation methods51, 53, 158. Two

hundred micrograms of postmitochondrial fractions from either postmortem human HF, PFC or

HF of mice. Brain slices pelleted by centrifugation were solubilized by brief sonication in 250 μl

of immunoprecipitation buffer (containing 25mM HEPES, pH 7.5, 200mM NaCl, 1mM EDTA,

protease and protein phosphatase inhibitor cocktails (Roche) and 0.1% 2-mercaptoethanol

containing 0.5% digitonin, 0.2% sodium cholate, and 0.5% NP-40) and incubated at 4°C with

end-to-end shaking for 1 hr. After dilution with 750 μl of ice-cold immunoprecipitation buffer

and centrifugation (4°C) to remove insoluble debris, Arc and its associated signaling complexes

including FLNA and NMDARs in the lysate were isolated by immunoprecipitation with 16 hr

incubation at 4°C with rabbit anti-Arc (1μg) immobilized on protein A/G-conjugated agarose

beads. The resultant immunocomplexes were pelleted by centrifugation at 4°C.After three washes

with 1ml of ice-cold PBS, pH 7.2, and centrifugation, the isolated Arc complexes were dissociated

41

by resuspended in 90 μl of elution buffer (pH2.8) for 10 min and centrifuged to obtained the

supernatant that contains Arc and its associated proteins. The resultant supernatant was

neutralized by addition of 10 μl 1.5M Tris HCl, pH8.8 and neutralized by adding 100 μl of 2 X

SDS-PAGE sample preparation buffer (125 mM Tris-HCl, pH 6.8, 20% glycerol, 4% SDS, 10%

2-mercaptoethanol, 0.2% bromophenol blue) and boiling for 5 min. The content of Arc and its

associated proteins in the anti-Arc immunoprecipitate was determined by Western blotting with

monoclonal antibodies directed against the indicated target. The Arc complex blots were stripped

and reprobed with monoclonal anti-Arc to assess immunoprecipitation efficiency and loading. To

determine Arc levels in response to stimuli, immobilized rabbit anti-actin (0.5 μg)–protein A-

conjugated agarose was added together with anti-Arc in the co-immunoprecipitation process. The

content of β-actin in resultant immunoprecipitates was analyzed by Western blotting using

monoclonal anti–β-actin to illustrate even immunoprecipitation efficiency and loading.

c) Western Blot Procedure

Samples of both HF and PFC, under basal and stimulated conditions, were size-fractionated

on a 10% polyacrylamide gel with Tris/glycine/SDS buffer at 100 V for approximately 100

minutes at room temperature. The gels were electrophorectically transferred onto BioRad 0.2μm

nitrocellulose membranes in 20% (v/v) methanol transfer buffer. Membranes were blocked with

10% nonfat milk in phosphate buffered saline containing 0.1% Tween-20 (0.1%PBST) for 1 hour

at room temperature. The membranes were incubated with primary antibodies of choice such as

anti-Arc monoclonal antibody were diluted in PBST and at 4°C overnight or at 25ºC for 2 hr.

After incubation, the primary antibodies were removed and membranes were washed 3 times with

0.1%PBST and the incubated with HRP-conjugated anti-mouse (1:7,500-10,000 in PBST) for 60

minutes. Membranes were washed with 0.1%PBST twice and distilled H2O once and the

42

immunocomplexes were detected using chemiluminescent method (SuperSignal West Peco or

Femto Maximum Sensitivity substrate, and visualized by exposure to X-Ray film. The specific

protein bands were quantified using densitometric scan using ImageJ software. The same blot

was stripped and re-probed with different antibodies to assess up to three additional parameters

such as loading control (anti-β-Actin antibody) and specific phosphoepitopes (phosphotyrosine

(pY), phosphothreonine (pT) and phosphoserine (pS) on Arc. Additionally, Arc modified by

oxidative stress was interrogated by probing the blot with antibodies direct against nitrotyrosine

to assess the nitrated Y-Arc levels in immunopurified Arc.

43

d) Statistical Evaluation

Statistical analyses of 2 conditions were done using a Student’s t-test. Statistical analyses of

multiple comparisons were done using a 1-way ANOVA. These were computed in Microsoft

Excel. These results are represented as the mean ± SEM (unless otherwise stated). P values less

than 0.05 are considered significant.

Results

1) Isolation of Arc by immunoprecipitation with anti-Arc is complete and

specific. The specificity and completeness of isolating Arc by immunoprecipitation with anti-Arc

antibodies was tested in hippocampi and prefrontal cortices from wild-type mice. Mouse

hippocampi and prefrontal cortices were homogenized in 500 μl of ice-cold immunoprecipitation

medium. Following centrifugation to remove nucleus and mitochondria, 200 μg of the resultant

post-mitochondrial fraction (adjust to 150 μl with immunoprecipitation medium) was sonicated on

ice for 10 sec and solubilized at 4°C for 1 hr by 0.5% NP-40/0.2% Na cholate/0.5% digitonin with

end-to-end shaking as indicated above. Following centrifugation to remove insoluble debris, the

obtained lysate was diluted with incubated with anti-Arc (Santa Cruz Biotechnology SC-15325),

-rabbit IgG immobilized onto protein A/G-conjugated agarose beads or protein A/G-conjugated

agarose beads alone. Separately, brain lysate (200 μg) was combined with 6X sample preparation

buffer and boiled for 5 min (Lysate). The obtained immunocomplexes were obtained by

centrifugation. The supernatant was removed and concentrated using 10-KDa molecular weight

cut-off filter and combined with 6X sample preparation buffer and boiled for 5 min. The

immunocomplex containing pellet was washed with PBS and Arc proteins are eluted from

immunocomplexes by 90 μl of antigen elution buffer (pH2.8). The obtained Arc-containing eluate

44

was neurolized with 10 μl 1.5M Tris HCl, pH8.8 and solubilized by adding 100 μl of 2X sample

preparation buffer and boiling for 5 min. The solubilized lysate and immunoprecipitates were

analyzed by Western blotting to evaluate the completeness and specificity of the Arc purification

procedure.

To affirm the completeness and specificity of the anti-Arc immunoprecipitation, 200 μg of

solubilized post-mitochondrial fractions derived from control HF and PFC slices were also

assessed by a two-step immunoprecipitation. Lysates were immunoprecipitated first with 1 μg of

immobilized anti-Arc, -rabbit IgG or protein A/G-conjugated agarose beads. The resultant

supernatants were then immunoprecipitated with 1 μg of immobilized anti-Arc.

The results summarized in Figure 14 indicate that 1 μg of anti-Arc antibodies completely

and specifically purified Arc from brain lysate. The completeness is supported by the data showing

the abundance of Arc in anti-Arc immunoprecipitates from 100 μg of brain lysate is similar to that

in equal amounts of brain lysate. In contrast, the supernatant has negligible residual Arc. Further

supports for complete immunoprecipitation by anti-Arc can also be drawn from the data indicating

that there is no more Arc in the Anti-Arc immunoprecipitates derived from supernatant of the first

anti-Arc immunoprecipitation.

The specificity of anti-Arc is supported by the data showing Arc only present in anti-Arc

but not anti-IgG or protein A/G precipitates in the first round of immunoprecipitation. The fact

that equal amounts of Arc can be immunoprecipitated from supernatants of the first

immunoprecipitation step by anti-Arc clearly illustrate immunoprecipitation with 1 μg of anti-Arc

is complete and specific.

45

Figure 14: Anti-Arc immunoprecipitation in the HF and PFC is

complete and specific.

The completeness and specificity of anti-Arc immunoprecipitation was

determined in mouse HF and PFC. The blots shown are the representatives

of three experiments.

46

2) Assessing Arc Expression

2a) Basal and Activity-driven Arc expression is decreased during normal aging

and in AD.

Since Arc expression is strongly related to synaptic activity and synaptic activity is clearly

declined during normal aging and even more sharply in AD, it is of interest to assess changes in

Arc expression, especially in response to stimuli that regulate postsynaptic activities. To this end,

Arc protein expression under non-stimulated basal conditions and in response to 10 μM NMDA/1

μM glycine or 1 nM insulin was assessed in both HF and PFC derived from varying ages of WT

and 3xTg AD mice. Following incubation, total Arc proteins were purified by

immunoprecipitation along with actin that are used to gauge the efficiency of immunoprecipitation

and serve as the loading control. The data summarized in figure 15a and 15b clearly indicate Arc

expression increases in response to stimulation of HF and PFC mouse tissue by activating

NMDARs and IRs with NMDA/glycine and insulin, respectively. The NMDA/glycine- and

insulin-driven Arc expression was significantly decreased in an age dependent manner in WT brain

by 10-month of age. The reductions in activity-induced Arc expression are more robust in the 3xTg

AD animal. By 6 months old, 3xTg AD animals display a greater than 60% reduction in

NMDA/glycine- and insulin-evoked Arc expression and further reduction in this stimulation-

driven activity is evidenced at 10- and 15-month of age. The reduced activity-driven Arc

expression in brains of older (>10-month-old) WT and 3XTg AD mice was accompanied by and

coincided with elevated basal Arc levels. Increased AD pathologies are found in aged cognitively

normal subjects144. Together with heightened synaptic dysfunction illustrated as reduced LTP and

increased LTD at old age and in AD, the data presented in figure 16 suggest that elevated basal

Arc levels and reduced activity-induced Arc expression are associated with and contributed to a

47

decline in synaptic functions in aged and AD brains. Together with heightened AD pathologies

and synaptic defects at old age and in AD, these data indicate that increased basal Arc may function

as a compensatory change in response to altered, activity driven Arc expression.

48

0

0.25

0.5

0.75

1

0

200

400

600

Arc

/-A

cti

n r

ati

o%

Sti

mu

lati

on

*

* * ** **

*

**

*

**

*

**

*

**

+ ++

# #

# #

# #

# #

# #

++++

++

^^^^

4-month W

T6-m

onth WT

6-month TG

10-month W

T10-m

onth TG15-m

onth WT

15-month TG

0

0.25

0.5

0.75

1

0

200

400

600

Arc

/-A

cti

n r

ati

o%

Sti

mu

lati

on

*

* * ** **

*

**

*

**

*

**

*

**

+ ++

# #

# #

# #

# #

# #

++++

++

^^^^

4-month W

T6-m

onth WT

6-month TG

10-month W

T10-m

onth TG15-m

onth WT

15-month TG

0

0.25

0.5

0.75

1

0

200

400

600

Arc

/-A

cti

n r

ati

o%

Sti

mu

lati

on

*

* * ** **

*

**

*

**

*

**

*

**

+ ++

# #

# #

# #

# #

# #

++++

++

^^^^

4-month W

T6-m

onth WT

6-month TG

10-month W

T10-m

onth TG15-m

onth WT

15-month TG

0

0.25

0.5

0.75

1

0

200

400

600

4-month W

T6-m

onth WT

6-month TG

10-month W

T10-m

onth TG15-m

onth WT

15-month TG

Kreb’s Ringer

10 μM NMDA + 1 μM Glycine

1 nM Insulin

*

*** *

**

*

**

*

**

*

**

*

**

+ +

# ## #

## # #

# #+++

^^ ^^

^

0

0.25

0.5

0.75

1

0

200

400

600

4-month W

T6-m

onth WT

6-month TG

10-month W

T10-m

onth TG15-m

onth WT

15-month TG

Kreb’s Ringer

10 μM NMDA + 1 μM Glycine

1 nM Insulin

*

*** *

**

*

**

*

**

*

**

*

**

+ +

# ## #

## # #

# #+++

^^ ^^

^

0

0.25

0.5

0.75

1

0

200

400

600

4-month W

T6-m

onth WT

6-month TG

10-month W

T10-m

onth TG15-m

onth WT

15-month TG

Kreb’s Ringer

10 μM NMDA + 1 μM Glycine

1 nM Insulin

*

*** *

**

*

**

*

**

*

**

*

**

+ +

# ## #

## # #

# #+++

^^ ^^

^

0

0.25

0.5

0.75

1

0

200

400

600

4-month

WT

6-month

WT

6-month

TG

10-month

WT

10-month

TG

15-month

WT

15-month

TG

Kreb’s Ringer

10 μM NMDA + 1 μM Glycine

1 nM Insulin

*

*** *

**

*

**

*

**

*

**

*

**

+ +

# ## #

## # #

# #+++

^^ ^^

^

a)

b)

Figure 15: Activity dependent Arc expression changes as a function of age and disease.

a: Representative blots showing activation-induced Arc expression in HF (LEFT) and PFC (RIGHT) of aged wild-

type (WT) and 3x AD transgenic (TG) mice.

b: Quantitative assessment of the activation-induced Arc expression in HF and PFC from 5 sets of 4-, 6-, 10-, and

15-month 3xTg AD mice with age matched WT. Data are expressed as the mean ± SEM of Arc/β-Actin optical

density ratios or % increase above basal Arc expression under non-stimulated, Kreb’s-Ringer incubated condition.

β-Actin levels are used as the immunoprecipitation/loading controls. HF = Hippocampal Formation, PFC =

Prefrontal Cortex.

*p<0.01 Compared to Kreb’s Ringer treated 4-month old WT group, #p<0.01 Compared to respective level in 4-

month old WT group, +p<0.01, ++p<0.05 Compared to respective level in 6-month old 3xTg group, ^p<0.01,

^^p<0.05 Compared to respective level in 10-month old WT group, Δp<0.01 Compared to respective level in

10-month old 3xTg group

49

2b) Assessment of the overall Arc protein levels.

Since Arc is an important synaptic regulator, we determined whether the overall Arc

protein expression in the HF and PFC is altered during normal aging and in AD. To achieve this

goal, the post-mitochondrial fractions prepared from 4-, 6-, 10-, and 15-month-old (there is no

3xTg AD cohort at 4 months) WT and 3xTg AD mice were solubilized by boiling in SDS-PAGE

sample preparation buffer (n=5). The Arc protein levels were assessed using Western blotting

with anti-Arc antibodies (Santa Cruz Biotechnology: SC-17839). The blots were stripped and re-

probed with anti-β-actin antibody (SC-517582) to ascertain even loading. The data shown in the

representative blots (figure 16a) and quantification (figure 16b) indicate that Arc protein

expression increases as mice age in both brain regions of WT mice. While the Arc protein levels

in HF did not differ between 4- and 6-month of age, Arc expression increased by 44% at 10- and

15-month of age. Similarly, the Arc expression in FCX did not change at 6-month of age but

increased by 12 and 24% respectively at the 10- and 15-month of age. This change in Arc protein

levels is even more severe in 3xTg AD mice. In the HF of 3xTg AD cohort, a 64-67 % elevation

in Arc expression level was evident by 6- and 10-month of age. At 15month-old, there is a 76%

increase in the Arc protein level. A robust, age-dependent increase in Arc expression is observed

in PFC of the 3xTg AD mice as Arc protein levels increased by 44%, 51% and 62% at 6-, 10- and

15-month of age, respectively. Together with the data showing markedly reduced activity-

dependent Arc expression in both HF and PFC (figure 15), these data may suggest that increased

Arc expression levels is an adaptive change in response to the reduced activity-driven Arc

expression. The elevated Arc protein levels in aged and 3xTg AD brains may suggest degradation

and/or increased gene expression of Arc occur during aging and in AD.

50

55

70

3529

130

55

35

Arc

β-Actin

55

70

3529

130

55

35

Arc

β-Actin

55

70

3529

130

55

35

Arc

β-Actin

0

0.5

1

1.5

4 Month W

T6 M

onth WT

6 Month 3xTG

10 Month W

T

10 Month 3xTG

15 Month W

T15 M

onth 3xTG

Arc

/-A

cti

n

Rati

o

*

**

*

*

*# ##

55

70

3529

130

55

35

Arc

β-Actin

0

0.5

1

1.5

4 Month W

T6 M

onth WT

6 Month 3xTG

10 Month W

T

10 Month 3xTG

15 Month W

T15 M

onth 3xTG

Arc

/-A

cti

n

Rati

o

**

**

*# ##

##

a)

b)

Figure 16: Basal Arc Expression levels in aging and AD

a: Representative blots showing basal Arc expression in HF (LEFT) and PFC (RIGHT) of wild-type (WT) and

3x AD transgenic (TG) mice at varying ages.

b: Quantitative assessment of the basal Arc expression in HF (LEFT) and PFC (RIGHT) from 5 sets of 4-, 6-,

10-, and 15-month-old WT and age-matched 3xTg AD mice. Data are expressed as the mean ± SEM of Arc/β-

Actin optical density ratios.

β-Actin levels are used as the immunoprecipitation/loading controls. *P < 0.01 Compared to 4-month WT #p

< 0.01, ##p < 0.05 compared to respective WT. HF = Hippocampal Formation, PFC = Prefrontal Cortex.

51

2c) Altered basal and activity-driven Arc expression in Human postmortem HF

from well-matched subjects with Alzheimer’s disease (AD), mild cognitive

impairment (MCI) and cognitively normal control subjects.

The data derived in AD mouse model suggest that reduced activity-driven Arc expression

may be related to synaptic dysfunction during AD pathogenesis. To assess the relationship

between the changes in Arc expression, especially in response to receptor stimulation, basal and

receptor activation-induced Arc expression was measured in the HF of 8 patients with AD

compared to well-matched subjects in control, MCI-sNAP and MCI-AD cohorts. In addition to

stimulation of the NMDAR by NMDA/glycine, activation of the α7nAchRs by PNU 282987 (a

potent, selective α7nAchR agonist), the trkBs by BDNF and insulin receptors by insulin was also

determined. The α7nAchR is the known upstream regulator of the NMDARs 145 and BDNF-trkB

signaling is intimately associated with NMDARs52,53. Upon incubation of the HF slices from

cognitive normal controls with NMDA/glycine, insulin, BDNF and PNU 282987, marked

increases in Arc expression was observed (figure 17a).

This receptor stimulation-evoked Arc expression is reduced most robustly in MCI-AD and

AD than in MCI-sNAP cases (figure 17b). In addition to the reduced stimulation-driven Arc

expression, significant increase in Arc levels in MCI-SNAP and AD under non-stimulated basal

condition was also noted. The increased Arc protein levels under basal conditions may partially

contribute to the observed reduction in activity-driven Arc expression in MCI-SNAP and AD.

More importantly, the dampened stimulation-induced Arc expression can occur without changes

in Arc protein levels under non-stimulated conditions as found in MCI-AD supports the notion

that reduced activity-driven Arc expression occurs earlier during the course of AD pathogenesis.

The elevated Arc protein levels under the basal condition is the result of adaptive increase in Arc

transcription in response to increased synaptic dysfunction and destruction in later phases of AD.

52

Moreover, the increased Arc protein levels under non-stimulated basal conditions noted in MCI-

AD hints that the homeostastic Arc levels are regulated by multiple factors including transcription

and degradation which are likely different between AD and MCI-SNAP. While the expression

in response to all stimuli was reduced most dramatically in MCI-AD and AD, the dampened

NMDA/glycine- and insulin-induced Arc response was evidenced together with relatively milder

decreases in BDNF and PNU 282987 responses in MCI-SNAP. These data together indicate that

reduced activity-induced Arc expression is an integral part of synaptic dysfunction in diseases with

cognitive impairments. The differential changes in MCI-AD and MCI-SNAP further highlight the

differences in disease progression and possibly the contribution of various pathogenic factors to

the synaptic/dendritic dysfunction in these brain diseases.

53

-Actin

Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

70

100

55

35

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

a)

b)

Figure 17: Arc expression is altered in cognitively impaired human HF.

a: Representative blots showing basal and activation-induced Arc expression in HF from cognitive normal

control, MCI-AD (amnestic MCI), MCI-SNAP (non-amnestic MCI) and AD (Alzheimer’s disease) subjects. β-

Actin levels are used as the immunoprecipitation/loading controls.

b: Quantitative assessment of the basal and activation-induced Arc expression in HF from 10 sets of cognitive

normal control, MCI-AD (amnestic MCI), MCI-SNAP (non-amnestic MCI) and AD (Alzheimer’s disease)

subjects. β-Actin levels are used as the immunoprecipitation/loading controls. Data are expressed as the mean

± SEM of Arc/β-Actin optical density ratios or % increase above basal Arc expression under non-stimulated,

Kreb’s-Ringer incubated condition.

54

3) Arc is modified post-translationally by phosphorylation.

Post-translational modification such as phosphorylation of a given protein not only

influences its function greatly but also often is incorporated into the signaling pathway as the

regulatory mechanism for controlling the protein’s localization, interaction with other molecules

and even degradation. Given that Arc is one of the more prominent synaptic regulators in the

postsynaptic field, it is of interest whether post-translational modifications of Arc exist, how this

process is regulated in physiological conditions and whether these mechanisms are altered during

aging and AD pathogenesis. To this end, the phosphorylation state of Arc was determined by

immunoblotting in HF and PFC from WT and 3xTg AD mice and human postmortem HF tissues.

The completeness and specificity of Arc phosphorylation was first tested by reciprocal

immunoprecipitation methods. The phosphorylated Arc in the anti-Arc immunoprecipitates was

first tested by Western blotting using antibodies directed against phospho-serine (pS), -tyrosine

(pY) or –threonine (pT). In addition, the presence and abundance of the phosphorylated Arc in

the anti-pS, -pY or –pT immunoprecipitates was determined by Western blotting with anti-Arc.

The results summarized in figure 18 demonstrate again that anti-Arc specifically and

completely immunoprecipitates Arc which contains pS- and pY- but not pT-Arc in both HF and

PFC of the WT mice. In support of complete and specific immunoprecipitation of Arc, the contents

of pS- and pY-Arc in the supernatants from anti-rabbit IgG or protein A/G-agarose beads are

similar to anti-Arc in the anti-Arc immunoprecipitates.

Further support of the idea that Arc is modified only at the serine and tyrosine but not

threonine sites as well as the completeness and specificity of immunoprecipitation procedure can

be drawn from the data presented in figure 19. The data showing pS- and pY- but not pT-Arc is

55

presented in the anti-pS and -pY immunoprecipitates indicate the immunoprecipitation by anti-pS

and –pY and –pT is specific.

The establishment of the immunoprecipitation method enables us to assess the effects of

age and AD pathologies on post-translational modification of Arc in brains from mouse model of

AD and humans.

56

Figure 18: Arc immunoprecipites contain serine- and tyrosine- but not

threonine-phosphorylated Arc in the HF and PFC.

a: Immunopurified Arc from HF is phosphorylated at both serine and

tyrosine but not threonine sites.

b: Immunopurified Arc from PFC is phosphorylated at both serine and

tyrosine but not threonine sites.

57

Figure 19: Immunoprecipitation with antibodies against the indicated

phosphoepitopes in the HF and PFC from WT mice.

Anti-pS and anti-pY but not anti-pT immunoprecipitates contain

phosphorylatedArc in both HF and PFC.

58

3a) Phosphorylation of Arc at serine (pS) and tyrosine (pY) is responsive to

receptor stimulation.

The phosphorylation states of Arc in the HF and PFC of both 3xTg AD and WT mice at

varying ages (4-, 6-, 10-, and 15-months old) under non-stimulated, basal conditions and following

stimulation of the NMDARs and IRs respectively by NMDA/glycine and insulin, was assessed in

the anti-Arc immunoprecipitates by Western blotting using phosphoepitope-specific antibodies.

Similarly, we also determined the phosphorylation states of Arc in postmortem human HF from

non-demented controls, MCI-SNAP, MCI-AD, and AD subjects (n=8) under non-stimulated basal

conditions and following stimulation by NMDA/glycine, insulin, BDNF and PNU 282987. The

data shown in figure 20-23 indicate that Arc is phosphorylated on tyrosine (Y) and Serine (S) but

not threonine (T) sites in mouse HF and PFC and postmortem human HF. Arc phosphorylation

on tyrosine and serine is increased following activation by all stimuli tested in both mouse and

human samples, suggesting that Arc’s function is regulated by phosphorylation.

3b) Arc phosphorylation states are altered during normal aging and in AD

pathogenesis.

Under the non-stimulated basal condition, pY- and pS-Arc levels are increased in both HF

and PFC of wild-type mice at age ≥ 10-month of age as well as all ages of 3xTg AD mice. In

accord with the reduced receptor stimulation induced Arc expression in old WT and 3xTg AD

mice, activity-dependent pY- and pS-Arc is reduced in an age- and disease pathology-dependent

manner. 3xTg AD mice exhibited greater elevations in basal Arc phosphorylation and greater

reductions in activity-dependent Arc phosphorylation compared to age-matched WT. This is most

evident in the 10- and 15-month old 3xTg AD mice (relative to the WT).

Phosphorylation of Arc is also observed in postmortem human HF. The data shown in

figure 22 indicate that under the non-stimulated basal condition, pY- and pS-Arc levels are

59

increased in MCI-SNAP and AD cases and activation-dependent pY- and pS-Arc is reduced in

MCI-AD, MCI-SNAP and most severely in AD.

These results indicate for the first time that Arc function is modulated by phosphorylation,

and these phosphorylation states are modified by advancing age and progression of AD

pathologies. The fact that changes in Arc phosphorylation are similar in pattern to the alterations

in basal and activity-dependent Arc expression, strongly suggests that phosphorylation is a

physiological feedback regulatory mechanism of Arc. The elevated levels of pS- and pY-Arc are

likely the result of a dysfunctional protein phosphatase system that is failing to dephosphorylate

Arc and other proteins at old age and in pathological conditions such as AD, thereby leading to its

accumulation.

60

a)

b)

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pY-Arc55

NY-Arc

55

55

55

pT-Arc

0

0.25

0.5

0.75

1

1.25

0

200

400

600

800

pS

-Arc

/-A

cti

n r

ati

o%

In

cre

as

e

4-month

WT

6-month

WT

6-month

TG

10-month

WT

10-month

TG

15-month

WT

15-month

TG

*

**

**

**

*

*

*

*

**

*

**

** *++ ++

# #

#

#

# ## #

# #

++^^

^

^

0

0.25

0.5

0.75

1

1.25

0

200

400

600

800

pY

-Arc

/-A

cti

n r

ati

o%

In

cre

as

e

4-month

WT

6-month

WT

6-month

TG

10-month

WT

10-month

TG

15-month

WT

15-month

TG

*

***

* **

**

*

* *

*

**

*

**

# # # # # ## #

^ ^

^

Figure 20: Basal and activity dependent Arc phosphorylation expression are altered in HF

of aged WT and 3xTg AD mice.

a: Representative blots showing basal and activation-induced Arc phosphorylation expression in

HF of aged wild-type (WT) and 3x AD transgenic (TG) mice.

b: Quantitative assessment of basal and activity dependent changes in Arc phosphorylations in 5

sets of 4-, 6-, 10-, and 15-month 3xTg AD mice with age matched WT. Data are expressed as the

mean ± SEM of Arc/β-Actin optical density ratios or % increase above basal Arc expression under

non-stimulated, Kreb’s-Ringer incubated condition. LEFT: pY-Arc, RIGHT: pS-Arc

β-Actin levels are used as the immunoprecipitation/loading controls. HF = Hippocampal

Formation, PFC = Prefrontal Cortex.

61

-Actin

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WTIP

: A

nti

-Arc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pS-Arc

pY-Arc55

NY-Arc

55

55

55

pT-Arc

0

0.25

0.5

0.75

1

0

200

400

600

800p

S-A

rc/

-Acti

n r

ati

o%

In

cre

as

e

4-month

WT

6-month

WT

6-month

TG

10-month

WT

10-month

TG

15-month

WT

15-month

TG

** **

****

* * ** * *

*

*

**

*

# #

# #

### #

# #

^^^^

^

pY

-Arc

/-A

cti

n r

ati

o

0

0.25

0.5

0.75

1

0

200

400

600

800

% I

ncre

as

e

# #

# #

# #

# #

# #

4-month

WT

6-month

WT

6-month

TG

10-month

WT

10-month

TG

15-month

WT

15-month

TG

** **** ** * * ** * *

* *

*

*^

*

a)

b)

Figure 21: Basal and activity dependent Arc phosphorylation expression are altered in PFC of aged WT and

3xTg AD mice.

a: Representative blots showing basal and activation-induced Arc phosphorylation expression in PFC of aged wild-

type (WT) and 3x AD transgenic (TG) mice.

b: Quantitative assessment of basal and activity dependent changes in Arc phosphorylations in 5 sets of 4-, 6-, 10-,

and 15-month 3xTg AD mice with age matched WT. Data are expressed as the mean ± SEM of Arc/β-Actin optical

density ratios or % increase above basal Arc expression under non-stimulated, Kreb’s-Ringer incubated condition.

LEFT: pY-Arc, RIGHT: pS-Arc

β-Actin levels are used as the immunoprecipitation/loading controls. PFC = Prefrontal Cortex.

62

Figure 22: Human HF shows disease dependent changes in Arc phosphorylation expression.

a) Representative blots showing pY- and pS-Arc increase in response to indicated stimuli.

b) Quantitative assessment of basal and activity dependent changes in Arc phosphorylations in 8 sets of HF from age matched

cognitive normal controls and disease cohorts. While pY- and pS-Arc levels are increased in MCI-SNAP and AD, the

activation-induced tyrosine- and serine-phosphorylation of Arc is reduced in MCI-AD, MCI-SNAP and AD comparing to

controls.

Data are expressed as the mean ± SEM of Arc/β-Actin optical density ratios or % increase above basal Arc expression under

non-stimulated, Kreb’s-Ringer incubated condition. β-Actin levels are used as the immunoprecipitation/loading controls. pY-Arc: phosphotyrosine modified Arc; pS-Arc: phosphoserine-Arc. MCI-AD: amnestic MCI; MCI-SNAP: non-amnestic

MCI; AD: Alzheimer’s disease; HF = Hippocampal Formation. β-Actin levels serve as the immunoprecipitation/loading

controls.

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

a)

b)

Kreb’s Ringer

NMDA/Glycine

Insulin

BDNF

PNU282987

63

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pY-Arc55

NY-Arc

55

55

55

pT-Arc

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pY-Arc55

NY-Arc

55

55

55

pT-Arc

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pY-Arc55

NY-Arc

55

55

55

pT-Arc

-Actin

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pS-Arc

pY-Arc55

NY-Arc

55

55

55

pT-Arc

-Actin

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pS-Arc

pY-Arc55

NY-Arc

55

55

55

pT-Arc

-Actin

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pS-Arc

pY-Arc55

NY-Arc

55

55

55

pT-Arc

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

a)

c)

b)

Figure 23 a-c: Arc does not show phosphorylation on the phosphoThreonine epitope.

pT: phosphoThreonine-Arc; Representative blots showing pT-Arc is not present in basal, unstimulated conditions and does not

increase in response to indicated stimuli in either mice or human samples. MCI-AD: amnestic MCI; MCI-SNAP: non-amnestic

MCI; AD: Alzheimer’s disease. β-Actin levels serve as the immunoprecipitation/loading controls.

64

4) Nitration of Arc indicates oxidative stress occurs during AD pathogenesis

modifies Arc.

Oxidative stress is one of the most common and well documented pathogenic processes in

aging and neurodegenerative disorders146. The high level of reactive oxygen species (ROS) is

associated with synaptic damage and causes functional defects in affected proteins. To determine

whether Arc is influenced by oxidative stress, the level of nitration of Arc estimated by the

abundance of nitrotyrosine in immunopurified Arc was determined by Western blotting with a

specific antibody directed against nitrotyrosine. The level of nitrotyrosine on Arc was measured

under non-stimulated, basal conditions and following receptor activation in the immunopurified

Arc from WT and 3xTg AD mice as well as human postmortem disease. The data shown in figure

24 indicate that elevated nitrated Arc is evidenced in WT at age ≥10-month of age and 3xTg AD

mice without effects from receptor stimulation. The nitrated Arc levels in both brain regions of

3xTg AD mice are markedly higher comparing to their nitrated Arc expression levels relative to

age-matched WT (figure 24). These data suggests that the heightened oxidative stress during AD

pathogenesis can destroy Arc’s function thereby limiting Arc’s responsiveness to receptor

stimulation. The fact that nitrated Arc levels are much higher in HF compared to PFC may suggest

that HF has a greater severity in oxidative damage (figure 24b). The human postmortem tissue

results support the idea that oxidative stress correlates with AD pathologies since nitrated Arc is

absent in MCI-SNAP cases and the levels of nitrated Arc is lower in MCI-AD than AD (figure

25). These results demonstrate for the first time that Arc is modified by oxidative stress in MCI-

AD and AD. The levels of nitrated Arc may be used to gauge the severity of oxidative stress and

AD pathologies.

65

-Actin

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pS-Arc

pY-Arc55

NY-Arc

55

55

55

pT-Arc-Actin

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pS-Arc

pY-Arc55

NY-Arc

55

55

55

pT-Arc

-Actin

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pS-Arc

pY-Arc55

NY-Arc

55

55

55

pT-Arc

0

0.25

0.5

0.75

1

NY

-Arc

/-A

cti

n r

ati

o

4-month

WT

6-month

WT

6-month

TG

10-month

WT

10-month

TG

15-month

WT

15-month

TG

*** *** ** *

***

** *

###

## #

^^^## #

## #

## #

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pY-Arc55

NY-Arc

55

55

55

pT-Arc-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pY-Arc55

NY-Arc

55

55

55

pT-Arc

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

+

+

4M-WT

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

+

+

+

+

+

+

+

+

+

+

+

+

6M-WT 6M-TG 10M-WT 10M-TG 15M-WT 15M-TG

pY-Arc55

NY-Arc

55

55

55

pT-Arc

0

0.25

0.5

0.75

1

4-month

WT

6-month

WT

6-month

TG

10-month

WT

10-month

TG

15-month

WT

15-month

TG

***

NY

-Arc

/-A

cti

n r

ati

o

* **

* **

***##

#

## #

## ### #

^^^

Figure 24: Arc is modified by nitration in the HF and PFC of WT and 3xTg AD animals.

a: Representative blots showing nitrated Arc under basal and stimulation conditions in HF of aged wild-type

(WT) and 3x AD transgenic (TG) mice.

b: Quantitative assessment of basal and activity dependent changes in nY-Arc of 4-, 6-, 10-, and 15-month 3xTg

AD mice with age-matched WT. Data are expressed as the mean ± SEM of Arc/β-Actin optical density ratios or

% increase above basal Arc expression under non-stimulated, Kreb’s-Ringer incubated condition. LEFT: pY-

Arc, RIGHT: pS-Arc

β-Actin levels are used as the immunoprecipitation/loading controls. HF = Hippocampal Formation, PFC =

Prefrontal Cortex.

a)

b)

66

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

-Actin

pS-Arc

NMDA (10 μM) /Glycine (1 μM):

Insulin (1nM):

BDNF (50ng/ml):

+

+

+

Control

+

+

+

MCI-AD

+

+

+

MCI-SNAP

+

+

+

AD

55

IP:

An

ti-A

rc

An

ti-A

cti

n

55

35

PNU282987(0.1 μM):++++

pT-Arc55

pY-Arc55

NY-Arc55

Figure 25: Arc is modified by nitration in the human postmortem HF.

a: Representative blot showing nitration of Arc under basal and stimulated conditions in HF

from cognitive normal control, NY-Arc: Nitrotyrosine-Arc; MCI-AD: amnestic MCI; MCI-

SNAP: non-amnestic MCI; AD: Alzheimer’s disease. β-Actin serves as the loading control.

b: Quantitative assessment of nitrated Arc under basal and stimulation conditions in human

postmortem HF. Data are expressed as the mean ± SEM of Arc/β-Actin optical density ratios or

% increase above basal Arc expression under non-stimulated, Kreb’s-Ringer incubated

condition. β-Actin levels are used as the immunoprecipitation/loading controls. HF =

Hippocampal Formation.

Kreb’s Ringer

NMDA/Glycine

Insulin

BDNF

PNU282987

a)

b)

67

5) Altered Arc connection with key signaling cascades during aging and AD

pathogenesis contributes to its altered phosphorylation and dysfunction.

The fact that Arc is phosphorylated in response to receptor stimulation suggests that Arc is

associated with key signaling pathways in the postsynaptic density (PSD). To test this hypothesis,

we conducted an Arc co-IP experiment and first probed for key scaffold proteins and their

associated signaling molecules in aged 3xTg AD and WT mice. To achieve equal Arc protein

levels, the input anti-Arc immunoprecipitates for WT and 3xTg AD mice were adjusted according

to their Arc levels under basal conditions (figure 16). Our data presented in figures 27 and 28

indicate that Arc is predominantly associated with NMDARs and also Filamin A (FLNA) to a

lesser extent in both HF and PFC from young (< 6-month of age) WT mice. While the Arc and

PSD-95/NMDAR linkage is supported by the high levels of obligatory NR1 subunits and

regulatory NR2A subunits and scaffolding protein, PSD-95, Arc-FLNA connection is indicated by

the presence of JAK2, PAK1, PTEN and PP2A along with FLNA (figure 26). As the age increased

(≥10-month of age) in WT and especially with AD pathogenesis in 3xTg AD mice, the Arc-FLNA

connection is strengthened together with reduction in Arc-PSD-95/NMDAR linkage (figure 26-

28). Such rearrangement in binding partners for Arc during normal aging and in AD pathogenesis

may indicate age- and disease-related changes in regulatory mechanisms for Arc and possibly

consequent altered synaptic plasticity and dendritic dynamics.

NMDAR signaling complexes play an important role in the regulation of synaptic plasticity

in the postsynaptic density. NMDA receptors signaling dysfunction is linked to synaptic defects

and dendritic abnormalities in the AD pathogenesis and to a lesser extent during the aging

process51, 53, 54. This receptor functional downregulation appears to cause derangement of the

postsynaptic density with specifically, reduction of NR1 and NR2 levels158, ,159. The defects in

postsynaptic density during aging and in AD are supported by the data from the present study

68

showing reduced Arc associated NR1, NR2A and PSD-95 in 3xTg AD and to a lesser extent in

aged (≥ 10-month of age) WT mice. Consistent with the interaction of PSD95 with NR2A, PSD‐

95 may be required for NMDAR localization to synapses160,161. The NMDAR dysfunction coupled

with diminished Arc-NMDAR connections can lead to reduced LTP and failed synaptic activation

resulting in eventual cognitive impairments in elderly and AD.

Filamin A (FLNA) is a critical scaffolding protein that is associated with and regulates

dozens of receptors and signaling molecules and intracellular protein trafficking by modulating

actin polymerization166. FLNA is associated constitutively with the insulin receptor166. Altered

FLNA conformation in aging and AD is a critical mechanism of disease pathogenesis77. Among

key kinases of interest in the regulation of Arc is Jak2, a non-receptor tyrosine kinase linking to

FLNA166. Jak2, along with its primary downstream target, Stat3, are both located in dendritic

spines. The Jak2-Stat3 pathway has been implicated in synaptic plasticity.163 Pak1 is a member of

the conserved p21-activated serine-threonine kinase family and is a cytoplasmic protein that is

recruited to the membrane upon activation. Phosphatase and tensin homolog deleted on

chromosome 10 (PTEN) is a tyrosine phosphatase that has been extensively characterized for its

central role in cancer, but has also been implicated in ASDs and other diseases. PTEN has been

shown to be a critical link between Aβ and postsynaptic depression, although the mechanism of

this function is unclear156,157. Here, we show that the levels of FLNA-associated PTEN were

decreased with age in WT animals and an even more robust reduction observed in FLNA-

associated PTEN levels in 3xTg AD mice. The serine/threonine protein phosphatase PP2A is a

protein comprised of scaffold, catalytic and regulatory subunits. Reduced function of PP2A has

been implicated in the disrupted L-LTD in AD. Similar to PTEN, decreased FLNA-linked PP2A

69

as indicated by reduced PP2A-Cα/β was noted in the brains of aged WT at ≥ 10-month-old with

even greater reduction in the brains of 3xTg AD mice.

Together, the data presented confirm previous report that Arc is in the NMDAR signaling

complex and indicate for the first time that Arc is primarily associated with NMDARs and also

with FLNA signaling pathways as its physiological regulators. While the Arc-NMDAR

connection is deteriorated during normal aging and more robust with the presence of AD

pathologies, Arc-FLNA association is dramatically increased in AD and to a less extent in WT

mice at the age ≥ 10-month of age. Such alteration in Arc’s binding partners indicates a shift in

its regulation during pathogenic progression that may be significantly related with synaptic defects

in AD and aged brains.

70

Filamin A

JAK2

PAK1

PTEN

PP2A-C/

NR2A

PSD-95

NR1

100

100

130

35

55

70

70

250IP

: A

nti

-Arc

4 Month W

T

6 Month W

T

6 Month 3xTG

10 Month W

T

10 Month 3xTG

15 Month W

T

15 Month 3xTG

Arc55

Figure 26: Altered Arc linkages with signaling complexes in aged and 3xTg AD

mice.

Arc association with its interactors are altered in the HF and PFC of 3xTg AD mice

compared to WT controls. Tissue lysates were IP’ed for Arc complexes and probed for

FLNA, JAK2, PAK1, PTEN, PP2A-Cα/β, NR2A, NR1, and PSD95 by immunoblotting.

71

Figure 27: Altered Arc linkages with signaling complexes in the HF of aged and

3xTg AD mice.

Arc association with its interactors are altered in the HF of 3xTg AD mice compared to

WT controls. HF tissue lysates were IP’ed for Arc complexes and probed for FLNA,

JAK2, PAK1, PTEN, PP2A-Cα/β, NR2A, NR1, and PSD95 by immunoblotting.

72

Figure 28: Altered Arc linkages with signaling complexes in the PFC of aged and

3xTg AD mice.

Arc association with its interactors are altered in the PFC of 3xTg AD mice compared to

WT controls. HF tissue lysates were IP’ed for Arc complexes and probed for FLNA,

JAK2, PAK1, PTEN, PP2A-Cα/β, NR2A, NR1, and PSD95 by immunoblotting.

73

6) Regulation of Arc by Phosphorylation is mediated by PKC/Src and

JAK2/PAK1 associated respectively with NMDARs and FLNA

The data showing Arc is regulated by phosphorylation of tyrosine and serine and the

revelation of Arc’s connection with NMDAR and FLNA signaling cascades suggest that kinases

associated with these signaling pathways are involved in phosphorylation of Arc. To test this

hypothesis, we incubated HF and PFC tissues with specific enzyme inhibitors.

Accordingly, HF and PFC slices from 6-month-old wild-type mice were incubated with

selected kinase inhibitors (table 1): Chelerythrine (PKC), PP1 (Src), Tryphostin B42 (JAK2) or

NVS PAK1 (PAK1) prior to exposure to either NMDA (10μM) + Glycine (1 μM) or Insulin (1nM).

PP1 is a potent, reversible and selective inhibitor of the Src family of protein tyrosine kinases.

Chelerythrine is a potent inhibitor of PKC. As expected, PP1 blocks NMDA/glycine-induced

tyrosine phosphorylation of Arc only (figures 29, 30). In contrast, inhibition of PKC by

chelerythrine blocks NMDA/glycine-induced phosphorylation of Arc on both tyrosine and serine

(figures 29, 30). This result is in accord with the previous observation that PKC can activate Src

via PyK2 in the NMDAR signaling163,164.

In addition, we also tested the role for kinases associated with FLNA in the regulation of

Arc by phosphorylation induced by insulin. Selectively inhibiting JAK2 by pre-treatment with

Tyrphostin B42 blocks insulin-induced phosphorylation of Arc on both tyrosine and serine sites.

In contrast, pretreatment with NVS PAK1, an allosteric inhibitor of p21-activated kinases prevents

insulin-induced serine phosphorylation exclusively. In agreement with an earlier report165, our

present data indicate that JAK2 is an upstream regulator of PAK1 in the FLNA signaling

cascade165.

74

Taken together, the data obtained using kinase inhibitors support the notion that Arc is

regulated by phosphorylation mediated through activation of NMDAR and FLNA signaling

pathways.

Name Target

PP1 Src family Tyrosine Kinase Inhibitor

Chelerythrine Potent inhibitor of Protein Kinase C (Ser/Thr)

Tyrphostin B42 Inhibitor of Jak2 (Tyr)

NVS PAK1 Potent allosteric inhibitor of PAK1 (Ser/Thr)

Table 1: Pharmacological agents used and targets of action.

75

Figure 29: Identification of critical

kinases responsible for pS-Arc and pY-

Arc in HF.

a: Representative blot showing pS-Arc and

pY-Arc in HF under stimulated conditions.

Tissue was treated with different enzyme

inhibitors to measure phosphorylation levels.

b: Quantitative assessment of stimulation

dependent changes in Arc phosphorylations

due to addition of kinase inhibitors in HF of

mice.

HF = Hippocampal Formation.

a)

b)

76

Figure 30: Revelation of the kinases that

mediateArc phosphorylation in PFC.

a: Representative blot showing pS-Arc and pY-

Arc in PFC under stimulated conditions. Tissue

was treated with different enzyme inhibitors to

measure phosphorylation levels.

b: Quantitative assessment of stimulation

dependent changes in Arc phosphorylations due

to addition of kinase inhibitors in PFC of mice.

PFC = Prefrontal Cortex

a)

b)

77

Conclusions

Deterioration in activity-dependent synaptic function during the aging process has been

proposed to be a risk factor in neurological disorders and especially in neurodegenerative diseases

with cognitive impairment. However, the underlying mechanisms remain largely unresolved

although defective synaptic regulatory mechanisms are likely involved. The postsynaptic activity

is modulated by the prominent regulatory protein Arc. Arc plays a critical role in regulation of

learning and memory by modulating stimulation-induced synaptic activity leading to L-LTP and

dendritic remodeling81-83. Given the pivotal role of Arc in sculpting neuronal function, in this thesis

project I studied the molecular mechanisms that regulate Arc expression. In particular, I examined

Arc under activity-dependent conditions during aging and in diseases of cognitive impairment

using two brain regions from a mouse model of AD and postmortem human HF tissue.

We demonstrate that in response to stimulation of the various neuronal receptors located

in the dendritic field, Arc expression dramatically increased in both brain regions of mice and in

postmortem human HF. The time frame of this rapid activity-elicited Arc expression is in

agreement with previous findings that mRNA for Arc is localized in the synaptic/dendritic field

rather than synthesized in the cell bodies and then shipped to the synapses for local activity172, 173.

Most importantly, we presented evidence that activity-dependent Arc expression is reduced during

the aging process and in the MCI phase with or without AD pathologies and most severely in AD.

The graded reduction in brains from aged, MCI and AD cases support the notion that age-related

deterioration of Arc responsiveness to stimuli is an integral part of synaptic/dendritic dysfunction.

In accord with the reduced receptor stimulation induced Arc expression, defected NMDAR,

α7nAChR and IR function have all been demonstrated in aged and AD brains174, 53, 54, 159. In

contrast, an increased TrkB activity in AD has been noted175. The reduced BDNF-induced Arc

78

expression in AD therefore supports the notion that synaptic activation by the activated TrkB is

mediated by the NMDARs through pY-TrkB-NMDAR linkage as previously demonstrated in

earlier studies175. The defected activity-dependent Arc expression in AD and MCI-SNAP but not

MCI-AD is accompanied by an elevated Arc protein level. These data suggest that basal Arc

protein level is driven by adaptive increases in Arc gene expression in cell bodies. Further, these

increases are related to the magnitude and/or dimension of synaptic dysfunction and/or destruction

rather than directly linked to the reduced activity-evoked Arc expression. In this regard, the

magnitude of synaptic dysfunction in the brains of MCI-AD is milder compared to AD and

therefore, most likely occurs earlier during AD pathogenesis elicited by Aβ, especially Aβ42. In

addition to the AD pathogenesis, reduced receptor function was also observed in the brains of

MCI-SNAP cases that are without significantly higher AD pathologies than their age-matched

peers. Hence, dampened activity-dependent Arc expression can be induced by pathological

changes other than, or in addition to, Aβ. The fact that impaired synaptic functions were observed

to be mediated, at least in part, by a failure to promote Arc expression by activation of multiple

receptors may suggest the damage occurs to common mechanisms required for receptor operation

such as adequate mitochondrial function, healthy synaptic membranes. Alternatively, reduced Arc

mRNA levels, resulting from: a failure to transport mRNA from cell bodies to the synaptic field,

improper mRNA storage, mRNA protection and/or defects in translational machinery may also

play a role in reduction in activity-driven Arc expression. Clearly, further research is required to

elucidate these unknowns.

Although activity-induced Arc expression has been well-established, the fate of Arc protein

once it is synthesized is largely unknown. We paid particular attention to the most common post-

translational modification: phosphorylation since phosphorylation of a protein not only changes

79

its behavior and disposal but also reflects its inter-relationship to and health of specific signaling

pathways. We show here, for the first time, that Arc is phosphorylated at tyrosine and serine but

not threonine sites. The fact that the levels of tyrosine- and serine-phosphorylated Arc increase

proportionally following receptor stimulation suggest that phosphorylation of Arc is a

physiological regulator of Arc that likely influences its function, localization, disposal and/or

interaction with its binding partners. The reduced activity-driven Arc phosphorylation in the brains

of MCI and AD cases as well as in 3xTg AD and aged WT mice indicate that Arc is tightly

regulated and closely associated with the synaptic activities. The highly coordinated

phosphorylation of Arc suggests that Arc is phosphorylated by the kinases within the signaling

pathways located in close proximity of or even physically associated with Arc. This hypothesis is

supported by our data showing that Arc is physically associated with the NMDARs probably

through PSD-95 and FLNA as Arc can be co-immunoprecipitated with PSD-95 and FLNA

although PSD-95 and FLNA are not directly connected. These data indicate there are at least two

populations of Arc located in close proximity but not physically associated with each other that

serve to modulate the synaptic activities within different synaptic/dendritic microdomains. In

accord, PSD-95 and its linked NMDARs are found in dendritic spines whereas FLNA is

predominantly located in dendritic shaft176, 177.

In an effort to elucidate the protein kinases that phosphorylate Arc, we induced Arc

phosphorylation by stimulation with NMDA/glycine and insulin to activate NMDARs and IRs,

respectively. Confirmed using selective kinase inhibitors, our data indicate that PKC and Src

tyrosine kinase are responsible for PSD-95 linked NMDAR activation induced Arc

phosphorylation, whereas JAK2 and PAK1 mediate FLNA-associated IR activation by insulin.

Blockade of PKC reduces both serine- and tyrosine phosphorylation of Arc confirming that PKC

80

activation can activate Src, as shown previously178. Inhibition of JAK2 prevents insulin-induced

tyrosine- and serine-phosphorylation on Arc thus demonstrating PAK1 can be activated by JAK2

activation166. Since Arc is synthesized on demand to orchestrate the dendritic remodeling in

response to increases in synaptic activities, phosphorylation of Arc with the kinases within the

signaling pathways may represent a mechanism to provide close monitoring of the selective

synaptic/dendritic regions by improving Arc’s interaction with specific protein(s) in the synapse

and/or ability to translocate to specific dendritic fields to monitor synaptic activity.

Most importantly, our data show that during aging and more prominently in AD, there is a

shift in the Arc connections from PSD-95/NMDARs to FLNA. This rearrangement of Arc

association during aging and in AD pathogenesis appears to occur gradually to provide a prominent

underlying mechanism by which the diminished Arc in the dendritic spines mediates deleterious

synaptic activities. Together with the data showing ex vivo exposure to Aβ42 markedly reduces

NMDA/glycine-induced Arc expression54, our data suggests that the increased Aβ42-elicited

pathology during AD pathogenesis reduces NMDAR signal transduction and disrupts Arc-PSD-

95/NMDAR linkage but promotes Arc-FLNA interaction leading to the dominant presence of Arc

in the FLNA signaling complexes. The reduced PTEN and PP2A in the FLNA signaling complexes

from 3x Tg AD mice and AD may, at least in part, be responsible for the elevated phosphorylated

Arc under basal conditions in AD.

An altered conformation of FLNA is closely linked to the amyloid and tau pathologies in

AD54. The altered FLNA can be induced by Aβ monomers or small oligomers since aberrant

FLNA is found prevalently in mice infused intracerebroventicularly with soluble Aβ54. In AD and

to a lesser extent aged brains, Aβ42 binding to α7nAChRs and TLR4 (CD14) recruits FLNA to

promote sustained Aβ42 toxic signaling to accelerate neurofibrillary lesions and

81

neuroinflammation, respectively53, 54. The abnormal FLNA may also play a role in insulin

resistance prevalently present in AD159. Hence, in addition to increased Aβ piling onto α7nAChRs,

the aberrant FLNA with altered conformation may also cause accumulation of Arc in the FLNA

associated signaling pathways including α7nAChRs, TLR4 and IRs.

It has been well-established that elevated Aβ monomers or small oligomers in AD

destabilize mitochondria, leading to oxidative stress and production of ROS/RNS that

deleteriously modify the function of multiple proteins such as tau. In this study, we show for the

first time that ROS/RNS also modify Arc as indicated by the presence of nitrated Arc in AD and

MCI-AD (but not MCI-SNAP). The oxidative stress modified Arc was also present in 3xTg AD

and to a lesser extent aged WT mice. The differential nitration of Arc observed in the HF and PFC

of the 3xTg AD mice suggests that susceptibility to oxidative stress is different in these two brain

regions. These data lend support to the notion that defected Arc plays a significant role in

mediating neuronal dysfunction induced by the escalated pathologies induced by soluble Aβ.

While the precise effect of nitration on Arc remains elusive, it is highly conceivable that nitration

on Arc impairs Arc’s function and consequently synaptic transmission and dendritic remodeling.

Lastly, reduction in activity-dependent Arc together with higher basal Arc expression are

observed in the brains from MCI-SNAP patients. Since the levels of Arc-PSD-95/NMDAR or -

FLNA connections in the MCI-SNAP cases are comparable to controls and there is no evidence

Arc is nitrated, the reduced activity-evoked Arc expression in MCI-SNAP is unlikely the result of

escalated Aβ-induced AD pathologies. Hence, the underlying mechanism(s) responsible for the

reduced receptor activation driven Arc is not known although universal attenuation of activity-

induced Arc expression across 4 distinct receptors suggests that defects in common factor(s) that

82

are vital to proper receptor function such as altered membrane fluidity may be the primary culprit.

Future studies are clearly required to fully address this change.

In summary, our results support the notion that activity-dependent Arc expression is a

pivotal event in the modulation of postsynaptic activities and dendritic remodeling to not only

meet the demands of a rapidly changing neuronal environment but also to support long-term

neuronal function such as memory consolidation. The data presented indicate that changes in

activity-dependent Arc expression as well as Arc’s regulation and connections during aging and

in diseases with cognitive impairment, especially AD, play an essential role in mediating decline

in postsynaptic/dendritic activity, diminished dendritic flexibility and neurodegeneration. Based

on the observed changes in Arc expression as well as its regulation and connections during aging

and in AD, we propose a hypothetical model (figure 31) to depict pathological consequences of

altered FLNA-enabled Aβ42 signaling via α7nAChR. Soluble Aβ42 monomers or small oligomers

bind α7nAChR thereby inducing recruitment of FLNA to these receptors. Dimers of native FLNA,

coupled to insulin receptors but not to α7nAChR or TLR4, are depicted as straight rods; red curly

FLNA depicts the conformation altered form that is recruited to α7nAChR and TLR4 and possibly

also associated with IRs. Enabled by altered FLNA’s new linkages, Aβ42 activates α7nAChR to

hyperphosphorylate tau subsequently leading to neurofibrillary lesions and neurodegeneration.

While Arc is associated with both PSD-95/NMDARs and FLNAs under the physiological

conditions, the elevated Aβ also promote the translocation of Arc from PSD-95/NMDARs to

FLNAs. This change is accompanied by an enhanced inflammatory cytokine release (TNFα, IL-

1β and IL-6) by reactive astrocytes resulting from persistently activated TLR453,54. This

neuroinflammation can contribute to insulin desensitization159. Although the insulin receptor is

constitutively associated with native FLNA, it is highly plausible that altered FLNA also

83

contributes to the insulin receptor dysfunction in AD. Aβ’s aberrant signaling through α7nAChR

impairs function of α7nAChR and of downstream NMDARs thereby limiting calcium influx

through both receptors. Increasing Aβ42 piling onto α7nAChR results in internalization and

accumulation of intraneuronal Aβ42-α7nAChR complexes. The hyperphosphorylation of tau

causes tau to dissociate from microtubules, disrupting microtubule stability, axonal/dendritic

transport and neuronal function. Along with dysfunctional tau, impaired NMDARs reduce Arc

expression and LTP with heightened LTD. The diminished postsynaptic activity in the dendritic

spines resulting from reduced activity-driven Arc expression and perhaps the increased nitrated

and/or phosphorylated Arc eventually can then cause shrinkage of dendritic spines and loss of

synapses. Along the AD pathogenic progression, cognitive impairment becomes more apparent

and neuropil treads and neurofibrillary tangles are accumulated, neurons degenerated and neuritic

plaques are formed. Thus, augmentation of postsynaptic activity mediated by activity-driven Arc

expression should be considered as one of the therapeutic goals for preventing or delaying age-

associated neurological diseases especially Alzheimer’s disease.

84

Figure 31: Proposed mechanism of action of Arc.

(a) Under physiological conditions. Arc associates with native conformation FLNA (blue sticks),

Jak2, Pak1 and NMDAR in the postsynaptic density.

(b) Under pathological conditions. Aβ42 and altered FLNA (curly orange springs) cause a host of

pathological changes downstream. Significantly, Arc is uncoupled from the complex it functions

in within the PSD. This leads to the altered expression seen.

a)

b)

85

Through studying changes in Arc expression level in relation to neurotransmitter-

and/neuroregulator-driven activities, alteration in Arc protein as well as Arc’s interaction with

other synaptic and neuronal proteins, this study has shed light on the role of Arc in the pathogenic

progression in the cognition relevant brain regions of MCI and AD. In addition, by investigating

the relationship between Arc and various pathogenic factors in MCI-SNAP, MCI-AD and AD, this

study has uncovered novel mechanisms that may alter Arc’s function during the course of

Alzheimer’s disease pathogenesis that may help to illuminate the differences between MCI-SNAP

and MCI-AD. The results obtained may facilitate the development of early diagnosis of AD and

novel treatment strategies. Altogether, the data obtained will help define the role of Arc in the

progression of postsynaptic/dendritic pathologies in key cognition regulatory brain areas in

diseases with cognitive defects.

86

References

1. Harada, C. N., Natelson Love, M. C., and Triebel, K.. “Normal Cognitive Aging.” Clinics in

geriatric medicine 29.4: 737–752. PMC. (2013).

2. Glisky EL. Changes in Cognitive Function in Human Aging. In: Riddle DR, editor. Brain

Aging: Models, Methods, and Mechanisms. Boca Raton (FL): CRC Press/Taylor & Francis;

2007. Chapter 1. Available from: https://www.ncbi.nlm.nih.gov/books/NBK3885/

3. Shi, L., Adams, M. and Brunso-Bechtold, J. K. Subtle Alterations in Glutamatergic Synapses

Underlie the Aging-Related Decline in Hippocampal Function. In: Riddle DR, editor. Brain

Aging: Models, Methods, and Mechanisms. Boca Raton (FL): CRC Press/Taylor & Francis;

2007. Chapter 8. Available from: https://www.ncbi.nlm.nih.gov/books/NBK3885/

4. Clayton DA, et al. A hippocampal NR2B deficit can mimic age-related changes in long-term

potentiation and spatial learning in the Fischer 344 rat. J Neurosci, 22: 3628. (2002).

5. Wenk GL, Barnes CA. Regional changes in the hippocampal density of AMPA and NMDA

receptors across the lifespan of the rat. Brain Res. 885:1. (2000).

6. Potier B, et al. NMDA receptor activation in the aged rat hippocampus. Exp Gerontol. 35:1185.

(2000).

7. Bach ME, et al. Age-related defects in spatial memory are correlated with defects in the late

phase of hippocampal long-term potentiation in vitro and are attenuated by drugs that enhance

the cAMP signaling pathway. Proc Natl Acad Sci USA. 96, 5280. (1999).

8. Salat DH, Kaye JA, Janowsky JS. Prefrontal gray and white matter volumes in healthy aging

and Alzheimer disease. Archives of neurology. 56, 338–44. (1999).

9. Sullivan P, Pary R, Telang F, Rifai AH, Zubenko GS. Risk factors for white matter changes

detected by magnetic resonance imaging in the elderly. Stroke; A Journal of Cerebral

Circulation. 21, 1424–8. (1990).

10. Meier-Ruge W, Ulrich J, Bruhlmann M, Meier E. Age-related white matter atrophy in the

human brain. Annals of the New York Academy of Sciences. 673, 260–9. (1992).

11. Rogalski E, Stebbins GT, Barnes CA, et al. Age-related changes in parahippocampal white

matter integrity: a diffusion tensor imaging study. Neuropsychologia. 50, 1759–65. (2012).

12. Petersen, R.C., Smith, G.E., Waring, S.C., Ivnik, R.J., Tangalos, E.G. and Kokmen, E. Mild

cognitive impairment: clinical characterization and outcome. Archives of neurology, 56(3),

303-308 (1999).

87

13. Petersen, Ronald C. Mild cognitive impairment. New England Journal of Medicine 364 (23),

2227-2234. (2011).

14. Schott, J. M. and Petersen, R. C. New criteria for Alzheimer’s disease: which, when and why?,

Brain, 138(5), 1134–1137, (2015).

15. Petersen, R. C., Caracciolo, B., Brayne, C., Gauthier, S., Jelic, V., & Fratiglioni, L. Mild

cognitive impairment: a concept in evolution. Journal of internal medicine, 275(3), 214-228.

(2014).

16. Petersen RC, Knopman DS, Boeve BF, et al. Mild Cognitive Impairment: Ten Years Later.

Archives of neurology. 66(12), 1447-1455. doi:10.1001/archneurol.2009.266. (2009).

17. Petersen, R. C., Aisen, P. , Boeve, B. F., Geda, Y. E., Ivnik, R. J., Knopman, D. S., Mielke, M.

, Pankratz, V. S., Roberts, R. , Rocca, W. A., Weigand, S. , Weiner, M. , Wiste, H. and Jack,

C. R. Mild cognitive impairment due to Alzheimer disease in the community. Ann Neurol., 74,

199-208. (2013).

18. Albert, M.S., DeKosky, S.T., Dickson, D, et al. The diagnosis of mild cognitive impairment

due to Alzheimer's disease: recommendations from the National Institute on Aging and

Alzheimer's Association Workgroup. Alzheimers Dementia 7:270–279. (2011).

19. De Reuck, J., Deramecourt, V., Cordonnier, C., Pasquier, F., Leys, D., Maurage, C. A., and

Bordet, R. The incidence of post-mortem neurodegenerative and cerebrovascular pathology in

mixed dementia. Journal of the Neurological Sciences, 366, 164-166. (2016).

20. Barker, W.W., Luis, C.A., Kashuba, A., Luis, M., Harwood, D.G., Loewenstein, D. et al.

Relative frequencies of Alzheimer's disease, Lewy body, vascular and frontotemporal

dementia, and hippocampal sclerosis in the State of Florida Brain Bank. Alzheimer Dis Assoc

Disord. 16: 203–212 (2002).

21. Alzheimer's Association. 2018 Alzheimer's disease facts and figures. Alzheimer's & Dementia

14.3, 367-429 (2018).

22. Jellinger, K.A. The enigma of mixed dementia. Alzheimers Dementia. 3: 40–53. (2007).

23. Prince, M., Bryce, R., Albanese, E., Wimo, A., Ribeiro, W. and Ferri, C.P. The global

prevalence of dementia: A systematic review and metaanalysis. Alzheimer’s & Dementia, 9:

63–75. (2013).

24. Hebert, L.E., Weuve J, Paul, A., Scherr, P.A. and Evans, D.A. Alzheimer disease in the United

States (2010–2050) estimated using the 2010 census. Neurology, 80: 1778–1783. (2013).

25. Arevalo-Rodriguez, I, Smailagic, N, Ciapponi A, Sanchez-Perez E, Giannakou A, Roqué i

Figuls M, et al. Mini-Mental State Examination (MMSE) for the detection of Alzheimer's

88

disease and other dementias in people with mild cognitive impairment (MCI). Cochrane

Database of Systematic Reviews, 10. (2013).

26. American Psychiatric Association. (2013). Diagnostic and statistical manual of mental

disorders (5th ed.). Arlington, VA: American Psychiatric Publishing.

27. Folstein, M. F., Folstein, S. E., and McHugh, P. R. “Mini-mental state”: a practical method for

grading the cognitive state of patients for the clinician. Journal of psychiatric research 12(3),

189-198. (1975).

28. Creavin, S.T., Wisniewski, S., Noel-Storr, A.H., et al. Mini-Mental State Examination

(MMSE) for the detection of dementia in clinically unevaluated people aged 65 and over in

community and primary care populations. Cochrane Database of Systematic Reviews. 1. DOI:

10.1002/14651858.CD011145.pub2. (2016).

29. Roalf, D. R. et al. Comparative Accuracies of Two Common Screening Instruments for the

Classification of Alzheimer’s Disease, Mild Cognitive Impairment and Healthy Aging.

Alzheimer’s & Dementia : The Journal of the Alzheimer’s Association 9(5), 529–537. (2013).

30. Trzepacz, P.T., Hochstetler, H., Wang, S., Walker, B., Saykin A.J. and “Alzheimer’s Disease

Neuroimaging Initiative.” Relationship between the Montreal Cognitive Assessment and Mini-

Mental State Examination for Assessment of Mild Cognitive Impairment in Older Adults.

BMC Geriatrics, 15:107. (2015).

31. Albert, M.S., DeKosky, S.T., Dickson, D., Dubois, B., Feldman, H.H., Fox, N.C., Gamst, A.,

Holtzman, D.M., Jagust, W.J., Petersen, R.C. and Snyder, P.J. The diagnosis of mild cognitive

impairment due to Alzheimer’s disease: Recommendations from the National Institute on

Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer's disease.

Alzheimer's & dementia, 7(3), pp.270-279. (2011).

32. Bejjani, C., Mehanna, R., & Shah, A. Biomarkers in Alzheimer’s Disease. Directions in

Psychiatry, 36 (4), 261-281. (2016).

33. Shaw, L. M., Vanderstichele, H., Knapik‐Czajka, M., Clark, C. M., Aisen, P. S., Petersen, R.

C., ... & Dean, R. Cerebrospinal fluid biomarker signature in Alzheimer's disease

neuroimaging initiative subjects. Annals of Neurology, 65(4), 403-413. (2009).

34. Gunnarsson, M. D., Lannfelt, L., Ingelsson, M., Basun, H., & Kilander, L. (2014). High tau

levels in cerebrospinal fluid predict rapid decline and increased dementia mortality in

Alzheimer's disease. Dementia and geriatric cognitive disorders, 37(3-4), 196-206.

35. Braak, H. and Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta

Neuropathologica, 82:239-259. (1991).

36. LeVine, H. and Walker, L.C., 2006. Models of Alzheimer's disease. In Handbook of Models

for Human Aging (pp. 121-134).

89

37. Bhat, A. H., Dar, K. B., Anees, S., Zargar, M. A., Masood, A., Sofi, M. A., & Ganie, S. A.

Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic

insight. Biomedicine & Pharmacotherapy, 74, 101-110. (2015).

38. Kim, G. H., Kim, J. E., Rhie, S. J., and Yoon, S. The role of oxidative stress in

neurodegenerative diseases. Experimental neurobiology, 24(4), 325-340. (2015).

39. Niedzielska, E., Smaga, I., Gawlik, M., Moniczewski, A., Stankowicz, P., Pera, J., & Filip, M.

Oxidative stress in neurodegenerative diseases. Molecular neurobiology, 53(6), 4094-4125.

(2016).

40. Fernandez, A.M. & Torres-Alemán, I. 2012. The many faces of insulin-like peptide signalling

in the brain. Nat. Rev. Neurosci. 13:225–239. doi:10.1038/nrn3209 (2012).

41. Arnold, S.E., Arvanitakis, Z., Macauley-Rambach, S.L., Koenig, A.M., Wang, H.Y., Ahima,

R.S., Craft, S., Gandy, S., Buettner, C., Stoeckel, L.E. and Holtzman, D.M.. Brain insulin

resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nature Reviews

Neurology, 14(3), 168. (2018).

42. Fraering, P.C. Structural and Functional Determinants of γ-secretase, an intramembrane

protease implicated in Alzheimer's disease. Current Genomics, 8(8), 531-549. (2007).

43. O'Brien, R. J. & Wong, P. C. Amyloid precursor protein processing and Alzheimer's disease.

Annual Review of Neuroscience, 34, 185-204. (2011).

44. Hardy, J., & Selkoe, D. J. The amyloid hypothesis of Alzheimer's disease: progress and

problems on the road to therapeutics. Science, 297(5580), 353-356. (2002).

45. Tanzi, R.E. & Bertram. L. Twenty years of the Alzheimer's disease amyloid hypothesis: a

genetic perspective. Cell, 120, 545-555. (2005).

46. McLean, C.A., Cherny, R.A., Fraser, F.W., Fuller, S.J., Smith, M.J., Vbeyreuther, K., Bush,

A.I. and Masters, C.L. Soluble pool of Aβ amyloid as a determinant of severity of

neurodegeneration in Alzheimer's disease. Annals of neurology, 46(6), 860-866. (1999).

47. Haass, C., and Selkoe, D. J. Soluble protein oligomers in neurodegeneration: lessons from the

Alzheimer's amyloid β-peptide. Nature reviews Molecular cell biology, 8(2), 101-112. (2007).

48. Näslund, J., Haroutunian, V., Mohs, R., Davis, K. L., Davies, P., Greengard, P., and Buxbaum,

J. D. Correlation between elevated levels of amyloid β-peptide in the brain and cognitive

decline. Jama, 283(12), 1571-1577. (2000).

49. Wang, H. Y., Lee, D. H., D'Andrea, M. R., Peterson, P. A., Shank, R. P., and Reitz, A. B. β-

Amyloid1–42 binds to α7 nicotinic acetylcholine receptor with high affinity implications for

Alzheimer's disease pathology. Journal of Biological Chemistry, 275(8), 5626-5632. (2000a).

90

50. Wang, H. Y., Lee, D. H., Davis, C. B., and Shank, R. P. Amyloid Peptide Aβ1‐42 Binds

Selectively and with Picomolar Affinity to α7 Nicotinic Acetylcholine Receptors. Journal of

Neurochemistry, 75(3), 1155-1161. (2000b).

51. Wang, H. Y., Stucky, A., Liu, J., Shen, C., Trocme-Thibierge, C., and Morain, P. Dissociating

β-amyloid from α7 nicotinic acetylcholine receptor by a novel therapeutic agent, S 24795,

normalizes α7 nicotinic acetylcholine and NMDA receptor function in Alzheimer's disease

brain. Journal of Neuroscience, 29(35), 10961-10973. (2009).

52. Wang, H. Y., Bakshi, K., Shen, C., Frankfurt, M., Trocmé-Thibierge, C., and Morain, P. S

24795 Limits β-Amyloid–α7 Nicotinic Receptor Interaction and Reduces Alzheimer's Disease-

Like Pathologies. Biological Psychiatry, 67(6), 522-530. (2010).

53. Wang, H. Y., Bakshi, K., Frankfurt, M., Stucky, A., Goberdhan, M., Shah, S. M., and Burns,

L. H. Reducing amyloid-related Alzheimer's disease pathogenesis by a small molecule

targeting filamin A. Journal of Neuroscience, 32(29), 9773-9784. (2012).

54. Wang, H. Y., Lee, K. C., Pei, Z., Khan, A., Bakshi, K., and Burns, L. H. PTI-125 binds and

reverses an altered conformation of filamin A to reduce Alzheimer's disease pathogenesis.

Neurobiology of Aging, 55, 99-114 (2017a).

55. Nagele, R. G., D’andrea, M. R., Anderson, W. J., and Wang, H. Y. Intracellular accumulation

of β-amyloid1–42 in neurons is facilitated by the α7 nicotinic acetylcholine receptor in

Alzheimer’s disease. Neuroscience, 110(2), 199-211. (2002).

56. Lewis, J., Dickson, D. W., Lin, W. L., Chisholm, L., Corral, A., Jones, G., ... & Eckman, C.

Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP.

Science, 293(5534), 1487-1491, (2001).

57. De Strooper, B., Saftig, P., Craessaerts, K., Vanderstichele, H., Guhde, G., Annaert, W., ... &

Van Leuven, F. (1998). Deficiency of presenilin-1 inhibits the normal cleavage of amyloid

precursor protein. Nature, 391(6665), 387. (1998).

58. Wolfe, M. S., Xia, W., Ostaszewski, B. L., Diehl, T. S., Kimberly, W. T., & Selkoe, D. J. Two

transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and γ-

secretase activity. Nature, 398(6727), 513. (1999).

59. Yu, Y., Run, X., Liang, Z., Li, Y., Liu, F., Liu, Y., ... & Gong, C. X. Developmental regulation

of tau phosphorylation, tau kinases, and tau phosphatases. Journal of Neurochemistry, 108(6),

1480-1494.). (2009).

60. Kovacs, G. G. Invited review: neuropathology of tauopathies: principles and practice.

Neuropathol. Appl. Neurobiol. 41, 3–23. doi: 10.1111/nan.12208 (2015).

91

61. Clavaguera, F., Bolmont, T., Crowther, R.A., Abramowski, D., Frank, S., Probst, A., Fraser,

G., Stalder, A.K., Beibel, M., Staufenbiel, M. and Jucker, M. Transmission and spreading of

tauopathy in transgenic mouse brain. Nature Cell Biology, 11(7), p.909. (2009).

62. Nonaka, T., Masuda-Suzukake, M., Arai, T., Hasegawa, Y., Akatsu, H., Obi, T., Yoshida, M.,

Murayama, S., Mann, D.M., Akiyama, H. and Hasegawa, M., 2013. Prion-like properties of

pathological TDP-43 aggregates from diseased brains. Cell Reports, 4(1), 124-134. (2013).

63. Hasegawa, M. Molecular mechanisms in the pathogenesis of Alzheimer’s disease and

tauopathies-prion-like seeded aggregation and phosphorylation. Biomolecules, 6(2), p.24.

(2016).

64. Kandel, Eric R., et al. Principles of Neural Science. New York, McGraw-Hill Health

Professions Division, (2013).

65. Hedden, T. & Gabrieli, J.D. Insights into the ageing mind: a view from cognitive

neuroscience. Nat. Rev. Neurosci. 5, 87–96. (2004).

66. Raz, N., Gunning-Dixon, F.M., Head, D., Dupuis, J.H. & Acker, J.D. Neuroanatomical

correlates of cognitive aging: evidence from structural magnetic resonance imaging.

Neuropsychology 12, 95–114. (1998).

67. Davies, C. A., Mann, D. M. A., Sumpter, P. Q., & Yates, P. O. A quantitative morphometric

analysis of the neuronal and synaptic content of the frontal and temporal cortex in patients with

Alzheimer's disease. Journal of the neurological sciences, 78(2), 151-164. (1987).

68. Scheff, S. W., Price, D. A., Schmitt, F. A., & Mufson, E. J. Hippocampal synaptic loss in early

Alzheimer's disease and mild cognitive impairment. Neurobiology of Aging, 27(10), 1372-

1384. (2006).

69. Bliss, T. V., & Lømo, T. Long‐lasting potentiation of synaptic transmission in the dentate area

of the anaesthetized rabbit following stimulation of the perforant path. The Journal of

Physiology, 232(2), 331-356. (1973).

70. Nicoll, R. A., & Roche, K. W. Long-term potentiation: peeling the onion. Neuropharmacology,

74, 18-22. (2013).

71. Nicoll, R. A., & Schmitz, D. Synaptic plasticity at hippocampal mossy fibre synapses. Nature

Reviews Neuroscience, 6(11), 863. (2005).

72. Gustafsson B, Wigstrom H. Long-term potentiation in the hippocampal CA1 region: its

induction and early temporal development. Prog Brain Res., 83:223–232. (1990).

73. Herring, B.E. and Nicoll. R. A. Long-Term Potentiation: From CaMKII to AMPA Receptor

Trafficking. Annu. Rev. Physiol., 78, 351-365. (2016).

92

74. Malenka, R. C., & Bear, M. F. LTP and LTD: an embarrassment of riches. Neuron, 44(1), 5-

21. (2004).

75. Nicoll, R. A. A brief history of long-term potentiation. Neuron, 93(2), 281-290. (2017).

76. Barria, A., Muller, D., Derkach, V., Griffith, L. C., and Soderling, T. R. Regulatory

phosphorylation of AMPA-type glutamate receptors by CaM-KII during long-term

potentiation. Science, 276, 2042-2045. (1997).

77. Malenka, R.C. Synaptic plasticity in the hippocampus: LTP and LTD. Cell, 78(4), 535-538.

(1994).

78. J.G. Valtschanoff, R.J. Weinberg. Laminar organization of the NMDA receptor complex

within the postsynaptic density. Journal of Neuroscience, 21, 1211-1217. (2001).

79. Okabe, S. Molecular anatomy of the postsynaptic density. Molecular and Cellular

Neuroscience, 34(4), 503-518. (2007).

80. J.C. Beique, R. Andrade. PSD-95 regulates synaptic transmission and plasticity in rat cerebral

cortex. Journal of Physiology, 546, 859-867 (2003).

81. K. Futai, M.J. Kim, T. Hashikawa, P. Scheiffele, M. Sheng, Y. Hayashi. Retrograde

modulation of presynaptic release probability through signaling mediated by PSD-95-

neuroligin. Nature Neuroscience, 10186-195. (2007).

82. M. Niethammer, E. Kim, M. Sheng. Interaction between the C terminus of NMDA receptor

subunits and multiple members of the PSD-95 family of membrane-associated guanylate

kinases. Journal of Neuroscience, 16, 2157-2163. (1996).

83. M. Sheng, M.J. Kim. Postsynaptic signaling and plasticity mechanisms. Science, 298, 776-

780. (2002).

84. V. Stein, D.R. House, D.S. Bredt, R.A. Nicoll. Postsynaptic density-95 mimics and occludes

hippocampal long-term potentiation and enhances long-term depression. Journal of

Neuroscience, 23, 5503-5506. (2003).

85. Vadlamudi, R. K., Li, F., Adam, L., Nguyen, D., Ohta, Y., Stossel, T. P., & Kumar, R. Filamin

is essential in actin cytoskeletal assembly mediated by p21-activated kinase 1. Nature Cell

Biology, 4(9), 681. (2002).

86. Hahn, C.G., Wang, H.Y., Cho, D.S., Talbot, K., Gur, R.E., Berrettini, W.H., Bakshi, K.,

Kamins, J., Borgmann-Winter, K.E., Siegel, S.J. and Gallop, R.J. Altered neuregulin 1–erbB4

signaling contributes to NMDA> receptor hypofunction in schizophrenia. Nature medicine,

12(7), 824. (2006).

93

87. Burns, L.H. and Wang, H.Y., Altered filamin A enables amyloid beta-induced tau

hyperphosphorylation and neuroinflammation in Alzheimer’s disease. Neuroimmunology and

Neuroinflammation, 4, 263-71. (2017).

88. Okuno, H. Regulation and function of immediate-early genes in the brain: beyond neuronal

activity markers. Neuroscience Research, 69, 175–186. doi: 0.1016/j.neures.2010.12.007

(2011).

89. Morgan, J. I., Cohen, D. R., Hempstead, J. L., and Curran, T. Mapping patterns of c-fos

expression in the central nervous system after seizure. Science, 237, 192–197. doi:

10.1126/science.3037702 (1987).

90. Lyford, G. L., Yamagata, K., Kaufmann, W. E., Barnes, C. A., Sanders, L. K., Copeland, N.

G., et al. Arc, a growth factor and activity-regulated gene, encodes a novel cytoskeleton-

associated protein that is enriched in neuronal dendrites. Neuron, 14, 433–445. doi:

10.1016/0896-6273(95)90299-6 (1995).

91. Guzowski, J. F., Setlow, B., Wagner, E. K., and McGaugh, J. L. Experience-dependent gene

expression in the rat hippocampus after spatial learning: a comparison of the immediate-early

genes Arc, c-fos, and zif268. Journal of Neuroscience, 21, 5089–5098. (2001).

92. Shepherd, J. D., & Bear, M. F. New views of Arc, a master regulator of synaptic plasticity.

Nature Neuroscience, 14(3), 279. (2011).

93. Korb, E., & Finkbeiner, S. Arc in synaptic plasticity: from gene to behavior. Trends in

Neurosciences, 34(11), 591-598. (2011).

94. Vazdarjanova, A., Ramirez‐Amaya, V., Insel, N., Plummer, T. K., Rosi, S., Chowdhury, S., ...

& Barnes, C. A. Spatial exploration induces ARC, a plasticity‐related immediate‐early gene,

only in calcium/calmodulin‐dependent protein kinase II‐positive principal excitatory and

inhibitory neurons of the rat forebrain. Journal of Comparative Neurology, 498(3), 317-329.

(2006).

95. McCurry, C. L., Shepherd, J. D., Tropea, D., Wang, K. H., Bear, M. F., & Sur, M. Loss of Arc

renders the visual cortex impervious to the effects of sensory experience or deprivation. Nature

Neuroscience, 13(4), 450. (2010).

96. El-Boustani, S., Ip, J. P., Breton-Provencher, V., Knott, G. W., Okuno, H., Bito, H., & Sur, M.

Locally coordinated synaptic plasticity of visual cortex neurons in vivo. Science, 360(6395),

1349-1354. (2018)

97. Myrum, C., Baumann, A., Bustad, H. J., Flydal, M. I., Mariaule, V., Alvira, S., ... & Márquez,

J. A. Arc is a flexible modular protein capable of reversible self-oligomerization. Biochemical

Journal, 468(1), 145-158. (2015).

94

98. Zhang, W., Wu, J., Ward, M. D., Yang, S., Chuang, Y. A., Xiao, M., ... & Worley, P. F.

Structural basis of arc binding to synaptic proteins: implications for cognitive disease. Neuron,

86(2), 490-500. (2015).

99. Campioni, M. R., & Finkbeiner, S. Going retro: ancient viral origins of cognition. Neuron,

86(2), 346-348. (2015).

100. Day, C., & Shepherd, J. D. (2015). Arc: building a bridge from viruses to memory. Biochemical

Journal, 469(1), e1-e3.

101. Korb, E., Wilkinson, C. L., Delgado, R. N., Lovero, K. L., & Finkbeiner, S. Arc in the nucleus

regulates PML-dependent GluA1 transcription and homeostatic plasticity. Nature

Neuroscience, 16(7), 874. (2013).

102. Epstein, I., & Finkbeiner, S. The Arc of cognition: Signaling cascades regulating Arc and

implications for cognitive function and disease. Seminars in cell & developmental biology

(Vol. 77, pp. 63-72). Academic Press. (2018).

103. Nikolaienko, O., Eriksen, M. S., Patil, S., Bito, H., & Bramham, C. R. Stimulus-evoked ERK-

dependent phosphorylation of activity-regulated cytoskeleton-associated protein (Arc)

regulates its neuronal subcellular localization. Neuroscience, 360, 68-80. (2017).

104. Ramírez-Amaya, V., Vazdarjanova, A., Mikhael, D., Rosi, S., Worley, P. F., & Barnes, C. A.

Spatial exploration-induced Arc mRNA and protein expression: evidence for selective,

network-specific reactivation. Journal of Neuroscience, 25(7), 1761-1768. (2005).

105. Bloomer, W. A., VanDongen, H. M., & VanDongen, A. M. Activity-regulated cytoskeleton-

associated protein Arc/Arg3. 1 binds to spectrin and associates with nuclear promyelocytic

leukemia (PML) bodies. Brain research, 1153, 20-33. (2007).

106. Bernardi, R. & Pandolfi, P.P. Structure, dynamics and functions of promyelocytic leukaemia

nuclear bodies. Nature Reviews Molecular Cell Biology, 8, 1006–1016 (2007).

107. Huang, F., Chotiner, J. K., & Steward, O. Actin polymerization and ERK phosphorylation are

required for Arc/Arg3. 1 mRNA targeting to activated synaptic sites on dendrites. Journal of

Neuroscience, 27(34), 9054-9067. (2007).

108. Shepherd, J. D., Rumbaugh, G., Wu, J., Chowdhury, S., Plath, N., Kuhl, D., ... & Worley, P.

F. Arc/Arg3. 1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron, 52(3), 475-

484. (2006).

109. Guzowski, J. F., Lyford, G. L., Stevenson, G. D., Houston, F. P., McGaugh, J. L., Worley, P.

F., & Barnes, C. A. Inhibition of activity-dependent arc protein expression in the rat

hippocampus impairs the maintenance of long-term potentiation and the consolidation of long-

term memory. Journal of Neuroscience, 20(11), 3993-4001. (2000).

95

110. Messaoudi, E., Kanhema, T., Soulé, J., Tiron, A., Dagyte, G., da Silva, B., & Bramham, C. R.

Sustained Arc/Arg3. 1 synthesis controls long-term potentiation consolidation through

regulation of local actin polymerization in the dentate gyrus in vivo. Journal of

Neuroscience, 27(39), 10445-10455. (2007).

111. Plath, N., Ohana, O., Dammermann, B., Errington, M. L., Schmitz, D., Gross, C., ... & Kobalz,

U. Arc/Arg3. 1 is essential for the consolidation of synaptic plasticity and

memories. Neuron, 52(3), 437-444. (2006).

112. Yuste, R. & Bonhoeffer, T. Morphological changes in dendritic spines associated with long-

term synaptic plasticity. Annual Review of Neuroscience, 24, 1071–1089. (2001)

113. Maletic-Savatic, M., Malinow, R. & Svoboda, K. Rapid dendritic morphogenesis in CA1

hippocampal dendrites induced by synaptic activity. Science 283, 1923–1927 (1999).

114. Fukazawa, Y., Saitoh, Y., Ozawa, F., Ohta, Y., Mizuno, K., & Inokuchi, K. Hippocampal LTP

is accompanied by enhanced F-actin content within the dendritic spine that is essential for late

LTP maintenance in vivo. Neuron, 38(3), 447-460. (2003).

115. Sarmiere PD, & Bamburg JR. Regulation of the neuronal actin cytoskeleton by ADF/cofilin.

Journal of Neurobiology, 58, 103–117. (2004).

116. Chowdhury, S., Shepherd, J. D., Okuno, H., Lyford, G., Petralia, R. S., Plath, N., ... & Worley,

P. F. Arc/Arg3. 1 interacts with the endocytic machinery to regulate AMPA receptor

trafficking. Neuron, 52(3), 445-459. (2006).

117. Man, H. Y., Lin, J. W., Ju, W. H., Ahmadian, G., Liu, L., Becker, L. E., ... & Wang, Y. T.

Regulation of AMPA receptor–mediated synaptic transmission by clathrin-dependent receptor

internalization. Neuron, 25(3), 649-662. (2000).

118. Ehlers, M. D. Reinsertion or degradation of AMPA receptors determined by activity-

dependent endocytic sorting. Neuron, 28(2), 511-525. (2000).

119. Waung, M. W., Pfeiffer, B. E., Nosyreva, E. D., Ronesi, J. A., & Huber, K. M. Rapid

translation of Arc/Arg3. 1 selectively mediates mGluR-dependent LTD through persistent

increases in AMPAR endocytosis rate. Neuron, 59(1), 84-97. (2008).

120. Bürkle, A. Posttranslational Modification. Brenner's Online Encyclopedia of Genetics, Four-

Volume Set (pp. 1533-1533). Elsevier Inc. (2001).

121. Martin, S., Wilkinson, K. A., Nishimune, A., & Henley, J. M. Emerging extranuclear roles of

protein SUMOylation in neuronal function and dysfunction. Nature Reviews Neuroscience,

8(12), 948. (2007).

96

122. Craig, T. J., and Henley, J. M. SUMOylation, Arc and the regulation homeostatic synaptic

scaling Implications in health and disease. Commun. Integr. Biol. 5, 634–636. doi:

10.4161/cib.21712. (2012).

123. Craig, T. J., Jaafari, N., Petrovic, M. M., Rubin, P. P., Mellor, J. R., & Henley, J. M.

Homeostatic synaptic scaling is regulated by protein SUMOylation. Journal of Biological

Chemistry, 287(27), 22781-22788. (2012).

124. Nair, R. R., Patil, S., Tiron, A., Kanhema, T., Panja, D., Schiro, L., ... & Bramham, C. R.

Dynamic Arc SUMOylation and selective interaction with F-actin-binding protein drebrin A

in LTP consolidation in vivo. Frontiers in Synaptic Neuroscience, 9, 8. (2017).

125. Mabb, A. M., Je, H. S., Wall, M. J., Robinson, C. G., Larsen, R. S., Qiang, Y., ... & Ehlers, M.

D. Triad3A regulates synaptic strength by ubiquitination of Arc. Neuron, 82(6), 1299-1316.

(2014).

126. Greer, P. L., Hanayama, R., Bloodgood, B. L., Mardinly, A. R., Lipton, D. M., Flavell, S. W.,

... & Ploegh, H. L. The Angelman Syndrome protein Ube3A regulates synapse development

by ubiquitinating arc. Cell, 140(5), 704-716. (2010).

127. Okuno, H., Akashi, K., Ishii, Y., Yagishita-Kyo, N., Suzuki, K., Nonaka, M., ... & Natsume,

R. Inverse synaptic tagging of inactive synapses via dynamic interaction of Arc/Arg3. 1 with

CaMKIIβ. Cell, 149(4), 886-898. (2012).

128. Link, W., Konietzko, U. W. E., Kauselmann, G., Krug, M., Schwanke, B., Frey, U., & Kuhl,

D. Somatodendritic expression of an immediate early gene is regulated by synaptic activity.

Proceedings of the National Academy of Sciences, 92(12), 5734-5738. (1995).

129. Blanpied, T. A., & Ehlers, M. D. Microanatomy of dendritic spines: emerging principles of

synaptic pathology in psychiatric and neurological disease. Biological Psychiatry, 55(12),

1121-1127. (2004).

130. Newey, S. E., Velamoor, V., Govek, E. E., & Van Aelst, L. Rho GTPases, dendritic structure,

and mental retardation. Journal of Neurobiology, 64(1), 58-74. (2005).

131. Li, Y., Pehrson, A. L., Waller, J. A., Dale, E., Sanchez, C., & Gulinello, M. A critical

evaluation of the activity-regulated cytoskeleton-associated protein (Arc/Arg3. 1)'s putative

role in regulating dendritic plasticity, cognitive processes, and mood in animal models of

depression. Frontiers in Neuroscience, 9, 279. (2015).

132. Smith, S. E., Zhou, Y. D., Zhang, G., Jin, Z., Stoppel, D. C., & Anderson, M. P. Increased gene

dosage of Ube3a results in autism traits and decreased glutamate synaptic transmission in mice.

Science Translational Medicine, 3(103), 103ra97-103ra97. (2011).

97

133. Niere, F., Wilkerson, J. R., & Huber, K. M. Evidence for a fragile X mental retardation protein-

mediated translational switch in metabotropic glutamate receptor-triggered Arc translation and

long-term depression. Journal of Neuroscience, 32(17), 5924-5936. (2012).

134. Managò, F., Mereu, M., Mastwal, S., Mastrogiacomo, R., Scheggia, D., Emanuele, M., ... &

Papaleo, F. Genetic disruption of Arc/Arg3. 1 in mice causes alterations in dopamine and

neurobehavioral phenotypes related to schizophrenia. Cell Reports, 16(8), 2116-2128. (2016).

135. Fumagalli F, Franchi C, Caffino L, Racagni G, Riva MA, Cervo L. Single session of cocaine

intravenous self-administration shapes goal-oriented behaviours and up-regulates Arc mRNA

levels in rat medial prefrontal cortex. International Journal of Neuropsychopharmacology,

12(3):423–429. (2009).

136. Takagi, S., Balu, D. T., & Coyle, J. T. Subchronic pharmacological and chronic genetic NMDA

receptor hypofunction differentially regulate the Akt signaling pathway and Arc expression in

juvenile and adult mice. Schizophrenia Research, 162(1-3), 216-221. (2015).

137. Garber, K. B., Visootsak, J., & Warren, S. T. Fragile X syndrome. European Journal of Human

Genetics, 16(6), 666. (2008).

138. Niere, F., Wilkerson, J. R., & Huber, K. M. Evidence for a fragile X mental retardation

protein-mediated translational switch in metabotropic glutamate receptor-triggered Arc

translation and long-term depression. Journal of Neuroscience, 32(17), 5924-5936. (2012).

139. Burke, S.N., Barnes, C.A. Aging ensembles:circuit contributions to memory deficits. In:

Mizumori SJY (ed) Hippocampal place fields: relevance to learning, memory. Oxford

University Press, Oxford, pp 364–384. (2008).

140. Hsieh, H., Boehm, J., Sato, C., Iwatsubo, T., Tomita, T., Sisodia, S., & Malinow, R. AMPAR

removal underlies Aβ-induced synaptic depression and dendritic spine loss. Neuron, 52(5),

831-843. (2006).

141. Lacor, P. N., Buniel, M. C., Chang, L., Fernandez, S. J., Gong, Y., Viola, K. L., ... & Krafft,

G. A. Synaptic targeting by Alzheimer's-related amyloid β oligomers. Journal of

Neuroscience, 24(45), 10191-10200. (2004).

142. Wu, J., Petralia, R. S., Kurushima, H., Patel, H., Jung, M. Y., Volk, L., ... & Kuhl, D.

Arc/Arg3. 1 regulates an endosomal pathway essential for activity-dependent β-amyloid

generation. Cell, 147(3), 615-628. (2011).

143. Palop, J. J., Chin, J., Bien-Ly, N., Massaro, C., Yeung, B. Z., Yu, G. Q., & Mucke, L.

Vulnerability of dentate granule cells to disruption of arc expression in human amyloid

precursor protein transgenic mice. Journal of Neuroscience, 25(42), 9686-9693. (2005).

98

144. Bennett, D. A., Schneider, J. A., Arvanitakis, Z., Kelly, J. F., Aggarwal, N. T., Shah, R. C.,

and Wilson, R. S. Neuropathology of older persons without cognitive impairment from two

community-based studies. Neurology, 66(12), 1837-1844. (2006).

145. Fulda, S., Gorman, A. M., Hori, O., & Samali, A. Cellular stress responses: cell survival and

cell death. International journal of cell biology, 2010. (2010).

146. Wang, H. W., Pasternak, J. F., Kuo, H., Ristic, H., Lambert, M. P., Chromy, B., ... &

Trommer, B. L. Soluble oligomers of β amyloid (1-42) inhibit long-term potentiation but not

long-term depression in rat dentate gyrus. Brain Research, 924(2), 133-140. (2002).

147. Durrenberger, P. F., Fernando, S., Kashefi, S. N., Ferrer, I., Hauw, J. J., Seilhean, D., ... &

Palkovits, M. Effects of antemortem and postmortem variables on human brain mRNA quality:

a BrainNet Europe study. Journal of Neuropathology & Experimental Neurology, 69(1), 70-

81. (2010).

148. Ferrer, I., Martinez, A., Boluda, S., Parchi, P., & Barrachina, M. Brain banks: benefits,

limitations and cautions concerning the use of post-mortem brain tissue for molecular studies.

Cell and Tissue Banking, 9(3), 181. (2008).

149. Bennett, D. A., Wilson, R. S., Schneider, J. A., Evans, D. A., Beckett, L. A., Aggarwal, N. T.,

... & Bach, J. Natural history of mild cognitive impairment in older persons. Neurology, 59(2),

198-205. (2002).

150. Bennett, D. A., Schneider, J. A., Buchman, A. S., de Leon, C. M., Bienias, J. L., & Wilson,

R. S. The Rush Memory and Aging Project: study design and baseline characteristics of the

study cohort. Neuroepidemiology, 25(4), 163-175. (2005).

151. McKhann, G., Drachman, D., Folstein, M., Katzman, R., Price, D., & Stadlan, E. M. Clinical

diagnosis of Alzheimer's disease Report of the NINCDS‐ADRDA Work Group* under the

auspices of Department of Health and Human Services Task Force on Alzheimer's Disease.

Neurology, 34(7), 939-939. (1984).

152. Mirra, S. S., Heyman, A., McKeel, D., Sumi, S. M., Crain, B. J., Brownlee, L. M., ... & Berg,

L. The Consortium to Establish a Registry for Alzheimer's Disease (CERAD) Part II.

Standardization of the neuropathologic assessment of Alzheimer's disease. Neurology, 41(4),

479-479. (1991).

153. Hyman, B. T., & Trojanowski, J. Q. Editorial on consensus recommendations for the

postmortem diagnosis of Alzheimer disease from the National Institute on Aging and the

Reagan Institute Working Group on diagnostic criteria for the neuropathological assessment

of Alzheimer disease. Journal of neuropathology and experimental neurology, 56(10), 1095.

(1997).

99

154. Yamamoto, T., & Hirano, A. A comparative study of modified Bielschowsky, Bodian and

thioflavin S stains on Alzheimer's neurofibrillary tangles. Neuropathology and applied

neurobiology, 12(1), 3-9. (1986).

155. Nelson, A. P., & O'Connor, M. G. Mild cognitive impairment: a neuropsychological

perspective. CNS spectrums, 13(1), 56-64. (2008).

156. Petersen, R. C., & Negash, S. Mild cognitive impairment: an overview. CNS spectrums, 13(1),

45-53. (2008).

157. Wilson, R. S., Barnes, L. L., De Leon, C. M., Aggarwal, N. T., Schneider, J. S., Bach, J., ...

& Bennett, D. A. Depressive symptoms, cognitive decline, and risk of AD in older persons.

Neurology, 59(3), 364-370. (2002a)

158. Wilson, R. S., Beckett, L. A., Barnes, L. L., Schneider, J. A., Bach, J., Evans, D. A., &

Bennett, D. A. Individual differences in rates of change in cognitive abilities of older persons.

Psychology and aging, 17(2), 179. (2002b)

159. Talbot, K., Wang, H. Y., Kazi, H., Han, L. Y., Bakshi, K. P., Stucky, A., ... & Arvanitakis, Z.

Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-

1 resistance, IRS-1 dysregulation, and cognitive decline. The Journal of clinical investigation,

122(4), 1316-1338. (2012).

160. LeVine, H., & Walker, L. C. Models of Alzheimer's disease. In Handbook of Models for

Human Aging (pp. 121-134). (2006).

161. Oddo, S., Caccamo, A., Shepherd, J. D., Murphy, M. P., Golde, T. E., Kayed, R., ... & LaFerla,

F. M. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular

Aβ and synaptic dysfunction. Neuron, 39(3), 409-421. (2003).

162. Talbot, K., Wang, H. Y., Kazi, H., Han, L. Y., Bakshi, K. P., Stucky, A., ... & Arvanitakis, Z.

Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-

1 resistance, IRS-1 dysregulation, and cognitive decline. The Journal of Clinical Investigation,

122(4), 1316-1338. (2012).

163. Nicolas C. S., Peineau, S., Amici, M., Csaba, Z., Fafouri, A., Javalet, C…Collingridge, G.L.

The Jak/STAT pathway is involved in synaptic plasticity. Neuron. 73(2):374-90. (2012).

164. Huang Y. Aβ-independent roles of apolipoprotein E4 in the pathogenesis of Alzheimer’s

disease. Trends in Molecular Medicine.16:287–94. (2010).

165. Bakshi, K., Gennaro, S., Chan, C. Y., Kosciuk, M., Liu, J., Stucky, A., ... & Wang, H. Y.

Prenatal cocaine reduces AMPA receptor synaptic expression through hyperphosphorylation

of the synaptic anchoring protein GRIP. Journal of Neuroscience, 29(19), 6308-6319. (2009).

100

166. Rider, L., Shatrova, A., Feener, E. P., Webb, L., & Diakonova, M. JAK2 tyrosine kinase

phosphorylates PAK1 and regulates PAK1 activity and functions. Journal of Biological

Chemistry, 282(42), 30985-30996. (2007).

167. Burns, L. H., & Wang, H. Y. Altered filamin A enables amyloid beta-induced tau

hyperphosphorylation and neuroinflammation in Alzheimer’s disease. Neuroimmunology and

Neuroinflammation, 4:263-71. (2017).

168. A.J. Mishizen-Eberz, R.A. Rissman, T.L. Carter, M.D. Ikonomovic, B.B. Wolfe, D.M.

Armstrong. Biochemical and molecular studies of NMDA receptor subunits NR1/2A/2B in

hippocampal subregions throughout progression of Alzheimer's disease pathology.

Neurobiology of Disease, 15, 80-92, 10.1016/j.nbd.2003.09.016 (2004).

169. N. Amada, K. Aihara, R. Ravid, M. Horie. Reduction of NR1 and phosphorylated

Ca2+/calmodulin-dependent protein kinase II levels in Alzheimer's disease. Neuroreport, 16,

1809-1813. (2005).

170. Buller AL,Monaghan DT. Pharmacological heterogeneity of NMDA receptors:

Characterization of NR1a/NR2D heteromers expressed in Xenopus oocytes. European Journal

of Pharmacology, 320, 87–94. (1997).

171. Hoe HS, Pocivavsek A,Chakraborty G,Fu Z,Vicini S,Ehlers MD,Rebeck GW. Apolipoprotein

E receptor 2 interactions with the N‐methyl‐D‐aspartate receptor. Journal of Biological

Chemistry, 281, 3425–3431. (2006).

172. Steward, O., & Worley, P. F. Selective targeting of newly synthesized Arc mRNA to active

synapses requires NMDA receptor activation. Neuron, 30(1), 227-240. (2001).

173. Farris, S., Lewandowski, G., Cox, C. D., & Steward, O. Selective localization of arc mRNA

in dendrites involves activity-and translation-dependent mRNA degradation. Journal of

Neuroscience, 34(13), 4481-4493. (2014).

174. Battaglia, F., Wang, H. Y., Ghilardi, M. F., Gashi, E., Quartarone, A., Friedman, E., & Nixon,

R. A. Cortical plasticity in Alzheimer’s disease in humans and rodents. Biological Psychiatry,

62(12), 1405-1412. (2007).

175. Wang, H. Y., Stucky, A., Hahn, C. G., Wilson, R., Bennett, D., & Arnold, S. BDNF-trkB

signaling in late life cognitive decline and Alzheimer’s disease. Translational Neuroscience,

2(2), 91-100. (2011).

176. Pak, D. T., Yang, S., Rudolph-Correia, S., Kim, E., & Sheng, M. Regulation of dendritic spine

morphology by SPAR, a PSD-95-associated RapGAP. Neuron, 31(2), 289-303. (2001).

177. Noam, Y., Phan, L., McClelland, S., Manders, E. M., Ehrengruber, M. U., Wadman, W. J., ...

& Chen, Y. Distinct regional and subcellular localization of the actin‐binding protein filamin

a in the mature rat brain. Journal of Comparative Neurology, 520(13), 3013-3034. (2012).

101

178. Huang Y. Aβ-independent roles of apolipoprotein E4 in the pathogenesis of Alzheimer’s

disease. Trends in Molecular Medicine. 16:287–94. (2010).