Close
The page header's logo
About
FAQ
Home
Collections
Login
USC Login
Register
0
Selected 
Invert selection
Deselect all
Deselect all
 Click here to refresh results
 Click here to refresh results
USC
/
Digital Library
/
University of Southern California Dissertations and Theses
/
A new model of neurodegeneration: integrating molecular, electrochemical, and neuronal behavior into a novel paradigm
(USC Thesis Other) 

A new model of neurodegeneration: integrating molecular, electrochemical, and neuronal behavior into a novel paradigm

doctype icon
play button
PDF
 Download
 Share
 Open document
 Flip pages
 More
 Download a page range
 Download transcript
Contact Us
Contact Us
Copy asset link
Request this asset
Transcript (if available)
Content A NEW MODEL OF NEURODEGENERATION:
INTEGRATING MOLECULAR, ELECTROCHEMICAL, AND NEURONAL BEHAVIOR
INTO A NOVEL PARADIGM
by
Robin Talamas
A Thesis Presented to the
FACULTY OF THE USC MANN SCHOOL OF PHARMACY
UNIVERSITY OF SOUTHERN CALIFORNIA
In Partial Fulfillment of the
Requirements for the Degree
MASTER OF SCIENCE
PHARMACEUTICAL SCIENCES
December 2024



ii
Acknowledgements
I would like to thank my advisor, Dr. Daryl Davies, for providing his insights into my
work and for giving me the opportunity to explore this area of research.
I would like to thank my mother for all of the emotional support that she has provided me
as I worked to complete my Master’s Degree.
I would like to thank my fellow members of the Davies, with special mention to
Samantha Skinner and Isis Janilkarn-Urena, for helping me to feel like part of a team.
I would like to thank the fellow members of my cohort, who helped me to develop a
sense of community during my time with USC.
And I would also like to thank my friend, Zachary Manning. I could not have done this
without your help.



iii
Table of Contents
Acknowledgements......................................................................................................................... ii
List of Figures................................................................................................................................ iv
Abstract............................................................................................................................................v
Chapter 1: Introduction....................................................................................................................1
1.1 Hippocampal Dysfunction .............................................................................................1
1.2 Neuronal Pathways ........................................................................................................2
1.3 Current Unknowns.........................................................................................................2
1.4 Creating a Coherent Model............................................................................................3
Chapter 2: Ion Transport Gradients .................................................................................................4
Chapter 3: Hippocampal Molecular Pathway ................................................................................10
3.1 Stress and Isolation ......................................................................................................10
3.2 Disruptions in the Urea Cycle and Metabolism...........................................................13
3.3 ATP in the Synapse......................................................................................................16
Chapter 4: Neural Pathways...........................................................................................................23
4.1 A Primary Neural Pathway ..........................................................................................23
4.2 Signaling Anticipation and Reward .............................................................................27
4.3 Rewiring of the VTA ...................................................................................................32
Chapter 5: Future Investigations....................................................................................................36
5.1 Insulin Resistance and the Brain..................................................................................36
5.2 Influence of the Gut-Brain Axis ..................................................................................37
5.3 Liver Secretion of FGF21 ............................................................................................37
Chapter 6: Conclusions..................................................................................................................39
Bibliography ..................................................................................................................................41



iv
List of Figures
Figure 1: Transport Dynamics Across the Cell Membrane .............................................................4
Figure 2: Ion Transport in the Synapse............................................................................................7
Figure 3: Interactions of the TCA and Urea Cycles ......................................................................15
Figure 4: Adenosine Signaling and Synaptic Function..................................................................19
Figure 5: Neural Pathway Regulating Behavior............................................................................24
Figure 6: MOR-CRFR1 Synaptic Modulation...............................................................................28
Figure 7: KOR-mediated Blockade of GABA Signaling ..............................................................30
Figure 8: CRFR1-CRFR2 Action in the VTA ...............................................................................34



v
Abstract
Neurodegenerative diseases such as Alzheimer’s and Parkinson’s are complex disorders that are
poorly understood. They incorporate a broad range of dysfunctions, including the formation of
Tau and Amyloid aggregates in the hippocampus. This results in changes in behavior and
impairment to cognitive function. However, given the complexity of these disorders, it is
difficult to characterize the active mechanisms and how they contribute to disease progression.
This thesis aims to provide a new model that can better describe the cellular and neurological
changes associated with neurodegenerative disorders. This includes an investigation into the
molecular mechanisms that contribute to impaired neuronal function. In particular, this thesis
proposes a neural pathway that connects multiple regions of the brain in order to characterize
changes in behavior. This thesis integrates current research into neurodegeneration in order to
synthesize a model that can guide future research.



1
Chapter 1: Introduction
Neurodegenerative diseases such as Alzheimer’s and Parkinson’s are complex disorders
characterized by loss of neuronal populations and the accumulation of cytotoxic
proteinopathies.1,2 These conditions are progressive and are often associated with comorbid
psychiatric disorders, such as addiction, anxiety, and other behavioral disorders.2,3,4,5 These
disorders affect complex molecular and signaling pathways within the brain, making it difficult
to characterize their pathologies. This presents a significant challenge in the treatment of these
disorders, as novel therapeutics cannot be developed without knowledge of the mechanisms
involved. Furthermore, little is known about the development of these disorders; this is
especially true for accumulating cytotoxic proteinopathies, as these proteins serve otherwise
unknown functions under physiological conditions. Given the complexity of these systems, a
broader understanding is necessary in order to characterize these disorders, and with mounting
evidence of comorbidities and overlapping mechanisms, the need for a coherent and
comprehensive model becomes more and more pressing. By understanding the mechanisms of
cellular dysfunction in the affected regions, it is possible to develop a model which recognizes
the coordinated actions of these regions and their contributions to neuropsychiatric pathology.
1.1 Hippocampal Dysfunction
Cellular dysfunction in the brain is observed in animals experiencing behavioral
impairments. Animal models of behavior are common techniques used in researching the
progression of psychiatric and neurological disorders as well as identifying risk factors in their
development.6,7 In particular, single-housing models of social isolation (SI) is a common method
for studying the development of anxiety and cognitive decline in mice.6,8,9,10 Previous
experiments have demonstrated that SI-treated mice experience cellular disruptions in the



2
hippocampus that were consistent with models of stress and chronic corticosterone
administration, implicating corticosterone as a key mediating factor.7,10,11 These experiments
have demonstrated various alterations in the cellular metabolic function of the hippocampus,
including disruptions to urea metabolism as well as aberrations in neurotransmitter signaling.
1.2 Neuronal Pathways
The complex role of the hippocampus in behavioral and neurological disorders points to
its intricate connectivity with other regions of the brain, in which dysfunction at this region
radiates outwards to other parts of the brain. Notably, investigations into behavioral deficits such
as anxiety and impaired social interaction have shown interactions between the hippocampus and
the ventral tegmental area (VTA) as well as with bed nucleus of the stria terminalis
(BNST).12,13,14 The VTA is notable for its involvement in stress and addiction as well as for
being a primary region of dopaminergic activity,15,16,17 whereas the BNST is a complex region of
diverse subnuclei with distinct connections across the brain.18,19 Understanding the roles and
connectivity of these regions as well as changes during disease is necessary to recognize the
impacts had by cellular dysfunction and how such dysfunction influences behavior. From this, it
is possible to expand the prospective model of neuropsychiatric disorders into a more
comprehensive framework, one that encapsulates a variety of pathologies.
1.3 Current Unknowns
Various unknowns still exist within the proposed model due to a lack of evidence to
describe causal interactions between regions and signaling pathways. Because of the complex
mechanisms and pathologies underlying neurodegenerative and psychiatric disorders, it is
difficult to characterize the roles played by regions outside of the brain. Hormones which are
secreted into circulation play a key role in maintaining healthy brain function, a key example



3
being liver-secreted FGF21 which declines in concentration with age and thereby presenting a
possible link to age-related cognitive decline.20 In addition to this is the complex behavior of the
human gut microbiome. Although the gut microbiome has been recognized as having a major
role in neurodevelopment and behavior, there are still many unknowns as to its interactions with
the brain. Recently, it has been reported that depletion of the gut microbiota was able to impair
social behavior in mice by altering signaling of corticosterone and oxytocin in the brain,12,13 thus
providing a tentative link between cognitive dysfunction and environmental factors such as diet
as well as alcohol-associated dysbiosis. However, the exact links between these regions and their
interactions remain unclear. To that end, it is paramount to characterize the activity of the brain
and how it is influenced by hormonal signaling. From this understanding, a clear link between
these regions can be deduced.
1.4 Creating a Coherent Model
The goal of this thesis is to present a model framework to describe the functions of the
hippocampus and its connected regions, including the VTA and the BNST, in modulating
behavior and thereby provide directions for current and future research. A clear, comprehensive
model of neurodegeneration and its related disorders can provide researchers with a direct
pathway towards developing novel therapeutics. Such a model should identify distinct changes in
the molecular and signaling pathways of the brain, namely in the hippocampus and its connected
regions. Beginning with changes in ion transport across the cell membrane, this thesis will
investigate dysfunction in the hippocampus and its subsequent role in a connected neural
pathway. From this framework, this thesis will describe key points for future research into
neurodegeneration.



4
Chapter 2: Ion Transport Gradients
Before realizing a clear model of neurodegeneration, it is necessary to characterize
cellular behavior and how it changes under inflammatory conditions. Aberrant signaling
behavior is a prominent feature in multiple models of neuroinflammation, including AD and
stroke, so it is important to understand the intracellular changes under these conditions.8,21,22
These changes reflected significant disruptions in inhibitory gamma-Aminobutyric Acid
(GABA) signaling, which included altered neuronal sensitivity and an increase in astrocytic
GABA synthesis and release.6,8,21 Astrocytes are both GABA sensitive and GABA secreting
Figure 1: Transport Dynamics Across the Cell Membrane
Stoichiometry of ion-dependent transport across the cell membrane. Movement of GABA and
Glutamate are mediated by GABA Transporters (GATs) and Excitatory Amino Acid
Transporters (EAATs), respectively. Ion concentrations are listed next to respective transporters.
Sodium and Calcium concentrations are maintained by the Sodium-Calcium Exchanger (NCE).
Created using BioRender.com



5
cells, allowing them to respond to changes in the extracellular environment and fine-tune
synaptic signaling.22,23 This process is primarily mediated by the activity of GABA Transporters
(GATs) and Excitatory Amino Acid Transporters (EAATs), which allow for the uptake of
GABA and glutamate from the synaptic space and limit their activity.22,24 Transporters such as
GATs use ion-dependent transport to carry neurotransmitters across the cell membrane and are
characterized by the Nernst Equation, which calculates the membrane potential at which ion
transport reaches equilibrium.24,25
1) Nernst Equation: 𝐸𝑟𝑒𝑣 = −
26.7
2𝑧𝑁𝑎 + 𝑧𝐶𝑙
𝐿𝑛[(
𝐶𝑙𝑖
−
𝐶𝑙𝑜
−
) (
𝐺𝐴𝐵𝐴𝑖
𝐺𝐴𝐵𝐴𝑜
) (
𝑁𝑎𝑖
+
𝑁𝑎𝑜
+
)
2
]
However, these ion gradients are shown to be disrupted in neuroinflammatory disorders,
with a notable increase in cellular calcium concentrations.8,26 Intracellular calcium becomes
cytotoxic at elevated concentrations,26,27 therefore prompting its efflux from the cell via the
Sodium-Calcium Exchanger (NCE).22,24 Because sodium concentration is relevant to the
transport of both GABA and glutamate, this counter transport activity subsequently changes their
respective equilibrium values and transport behavior.
21,22,24 However, because astrocytes are
responsible for the uptake of multiple signaling molecules, it becomes difficult to describe
changes in cellular transport across multiple channels. To that end, it was necessary to prepare a
modified version of the Nernst Equation.22,24,25 This version of the formula organizes ion
movements across the respective transporters as though acting as a single concerted mechanism.
This technique balances movement across the three transporters by using a net zero change in
Sodium concentration, as it is involved in the activity of each of them.22,24 This paradigm can be
used to better describe interactions between membrane transporters in response to extreme
changes in ion concentrations, such as increases in intracellular calcium. Three formulae were



6
developed to describe the different combinations of GABA and glutamate efflux/influx: GABAGlutamate Efflux, Glutamate-Dominant Efflux, and GABA-Dominant Efflux.
2) GABA-Glutamate: 𝐸𝑟𝑒𝑣 = −
26.7
4
𝐿𝑛[(
𝐶𝑎𝑖
2+
𝐶𝑎𝑜
2+
)
5
(
𝐺𝑙𝑢𝑖
−
𝐺𝑙𝑢𝑜
−
)
3
(
𝐶𝑙𝑖
−
𝐶𝑙𝑜
−
)
3
(
𝐺𝐴𝐵𝐴𝑖
𝐺𝐴𝐵𝐴𝑜
)
3
(
𝐻𝑖
+
𝐻𝑜
+
)
3
(
𝐾𝑜
+
𝐾𝑖
+)
3
]
3) Glutamate-Dominant: 𝐸𝑟𝑒𝑣 = −
26.7
4
𝐿𝑛[(
𝐶𝑎𝑖
2+
𝐶𝑎𝑜
2+
)
3
(
𝐺𝑙𝑢𝑖
−
𝐺𝑙𝑢𝑜
−
)
5
(
𝐶𝑙𝑜
−
𝐶𝑙𝑖
−
)
3
(
𝐺𝐴𝐵𝐴𝑜
𝐺𝐴𝐵𝐴𝑖
)
3
(
𝐻𝑖
+
𝐻𝑜
+
)
5
(
𝐾𝑜
+
𝐾𝑖
+)
5
]
4) GABA-Dominant: 𝐸𝑟𝑒𝑣 = −
26.7
1
𝐿𝑛[(
𝐶𝑎𝑖
2+
𝐶𝑎𝑜
2+
)
3
(
𝐺𝑙𝑢𝑜
−
𝐺𝑙𝑢𝑖
−
)(
𝐶𝑙𝑖
−
𝐶𝑙𝑜
−
)
6
(
𝐺𝐴𝐵𝐴𝑖
𝐺𝐴𝐵𝐴𝑜
)
6
(
𝐻𝑜
+
𝐻𝑖
+)(
𝐾𝑖
+
𝐾𝑜
+
)]
For the majority of astrocytes, intracellular glutamate remains at a relatively low
concentration of 0.3 mM.24 This has been suspected to be due to high activity of Glutamine
Synthetase in astrocytes, which provides a vital step in the recycling of neurotransmitters from
the extracellular space. However, a distinct population of hippocampal astrocytes has been found
to closely mediate synaptic plasticity through the rapid and localized release of cellular
glutamate.28 These astrocytes were determined to have the necessary cellular machinery for
vesicular glutamate release similar to neurons, demonstrating increased transcription of genes
associated with synaptic structure, ion transport, and exocytosis. These astrocytes were found to
play a significant role in synaptic long-term potentiation (LTP) as well as pathological processes
such as seizures and addiction.28,29 As such, it is necessary to include this subset within the
following calculations. However, because these astrocytes demonstrate a specialized molecular
profile, it is unlikely that the intracellular glutamate concentration will match with those
observed in most astrocyte populations. For this reason, the following calculations will be made
under the assumption that this astrocyte population possesses intracellular glutamate
concentrations similar to neurons (10 mM).30
Using Equations 2, 3, and 4 above, the reversal potentials can be determined for each
efflux mode. Under physiological conditions, these patterns would not be frequently observed as



7
part of astrocyte behavior, as the calculated reversal potentials would greatly exceed the
astrocyte resting potential of -80 mV.24 However, efflux can be predicted to become much more
prevalent under conditions of cytotoxic intracellular calcium, in which concentrations rise
beyond an estimated five-fold increase (>500 nM).26 More specifically, the reversal potential for
GABA-Dominant Efflux shows a significant drop from 12.9 mV to -116 mV in standard
Figure 2: Ion Transport in the Synapse
Mediation of astrocytes in synaptic signaling. GABA (orange) and Glutamate (purple) are
transported across the astrocyte membrane to modulate availability in the synapse. Astrocytes
perform Glutamate-Dominant (left) or GABA-Dominant (right) efflux depending on intracellular
ion concentrations. Stoichiometry of each transporter in the efflux pattern is shown. Created
using BioRender.com



8
astrocytes, far below the astrocyte resting potential. This aberrant efflux behavior has been
implicated in multiple neuroinflammatory conditions in which increases in intracellular calcium
have been observed.8,21,26 Conversely, astrocytes predicted to have a higher cellular glutamate
concentration have been estimated to be more prone towards GABA-Glutamate and GlutamateDominant Efflux, with reversal potentials dropping to a respective -35.4 mV and -39.1 mV
during increased cellular calcium. Furthermore, Glutamate-Dominant Efflux is predicted to
become more active under increased extracellular GABA, suggesting a compensatory
mechanism against astrocytic GABA secretion. In the context of the hippocampus, this presents
a possible means of balancing excitatory and inhibitory signaling under cellular stress,
maintaining normal synaptic activity and avoiding impairments in memory.
However, this behavior is no longer apparent in the case of AD, in which efflux of
astrocytic GABA appears predominant.8,21,22 This suggests an additional change to cellular ion
concentration in AD that impairs astrocyte efflux of glutamate. This is most likely due to an
elevation in intracellular chloride, which helps to mediate GABA transport and signaling.22
Chloride concentration gradients play little role in the activity of EAAT or NCE, meaning that
changes in concentration primarily affect GABA transport via GAT.22,24 Chloride concentration
gradients are maintained by the activity of transporters KCC2 and NKCC1, which have been
found to show changes in function due to stress and Amyloid exposure, increasing intracellular
chloride concentration as a result.31,32 Under a modest change of 30 mM to 40 mM, this is
sufficient to reduce the reversal potential of GABA-Dominant Efflux from -22.4 mV to -68.5
mV in glutamate-secreting astrocytes, well below the reversal potentials for glutamate-based
efflux. Additionally, these disruptions in cellular concentrations present a significant risk to
neuronal function, as ionotropic GABA signaling is largely dependent on chloride influx.31



9
Under moderate increases, this effect may blunt GABAergic inhibition, desensitizing the neuron
to astrocytic GABA as a result. However, under extreme increases from 5 mM to 15 mM, this
can be predicted to shift the chloride reversal potential from hyperpolarizing (-82.5 mV) to
potentially depolarizing (-53.2 mV). From these estimations, it is possible to predict a clear
change in cellular behavior, one which becomes increasingly prevalent in the research of
neurodegeneration.



10
Chapter 3: Hippocampal Molecular Pathway
3.1 Stress and Isolation
To understand the overlap between psychiatric and neurodegenerative disorders, it is
important to recognize the signs of neurodegeneration in behavioral models. Social Isolation (SI)
is an established model for the development of cognitive impairment in mice.6,8,9,10 In particular,
socially isolated mice have shown distinct behavioral changes such as anxiety and impaired
memory as well as neurological changes including neuroinflammation, impaired metabolic
energy, and even the accumulation of hyperphosphorylated Tau fibrils, a prominent marker in
Alzheimer’s Disease (AD).8,9,10,33
The hippocampus is critical for the consolidation of long-term
memory and is one of the primary regions affected by AD.6,8,9,10 Previous studies using SI
models have investigated cellular dysfunction within the hippocampus.6,8,9,10 These studies have
demonstrated a consistent pattern of cellular dysfunction within the hippocampus, including
impaired GABAergic signaling as well as a phosphorylation of Tau protein at the PHF-1
region.
6,8,10,33 Notably, this pattern is consistent with other mouse models of stress, including
models of Repeated Acute Stress as well as injection of corticosterone into the
Hippocampus.7,10,11
These effects are further compounded by chronic alcohol consumption. The combination
of stress and alcohol consumption yielded greater changes in plasma corticosterone and GABA
dysfunction.34,35,36 This is distinct from conditions in which stress alone was not sufficient to
induce significant changes, indicating that alcohol consumption provides a cumulative effect.34,36
The role of alcohol abuse in the development of AD is evidenced by alcohol-induced increases in
Tau phosphorylation and Amyloid-Beta content in the hippocampus.35,37 Additionally, chronic
alcohol consumption reveals the involvement of glycogen synthase kinase 3B (GSK-3B), an



11
ethanol-sensitive protein which induces Tau phosphorylation at the PHF-1 region.37,38,39 GSK-3B
showed increased activation in the above models of stress, suggesting a causal effect between
stress-induced elevations in corticosterone and Tau phosphorylation.7,11,39 Furthermore, changes
in GSK-3B activation following alcohol exposure were observed in the hippocampus as well as
in the prefrontal cortex (PFC) and the nucleus accumbens, with activation being associated with
increases in alcohol-seeking behavior and anxiety following abstinence.38,40,41 Using this
information, a clear relationship presents itself between GSK-3B and development of
neurodegeneration and alcohol abuse.
GSK-3B has various functions within the cell, but its most recognized role is as part of
the Beta-Catenin destruction complex. Beta-Catenin is a signaling molecule which induces gene
transcription in response to external stimuli but is limited by its degradation by the destruction
complex.42,43 In addition to this, Beta-Catenin is also involved in what is called the Noncanonical
Pathway, in which Beta-Catenin triggers a signaling cascade which releases calcium stored
within the endoplasmic reticulum into the cell.43 Under high intracellular concentrations, calcium
becomes cytotoxic, requiring efflux from the cell as well as negative feedback to prevent further
increase. This behavior is consistent with the existing data, as increased hippocampal calcium
influx was reported in mouse models of stress.8,44, Furthermore, it has been observed that DKK1,
a natural inhibitor of the Wnt-Beta-Catenin signaling pathway, is induced by corticosterone,
further implicating Beta-Catenin in mediating the rise in cellular calcium.42,43,45 This negative
feedback is likely to be mediated by calcium-induced proteolysis of GSK-3B. Activation of
GSK-3B is controlled by serine phosphorylation at the S9 site, with GSK-3B being deactivated
upon phosphorylation.42,43 However, Calpain has been identified as a candidate protease for this



12
site, indicating that calcium activation of Calpain can indirectly induce GSK-3B activity by
preventing phosphorylation.46,47,48
Overactivity of GSK-3B poses significant risk to cell survival, diminishing the cell’s
antioxidant capacity through degradation of NRF2.49,50 NRF2 is commonly regarded as a “master
regulator” for the cell’s antioxidant response as well as induction of mitochondrial function.51,52
Under normal conditions, NRF2 is readily degraded after modification by KEAP1, thereby
limiting its activity in the cell. However, this process is prevented by the presence of reactive
oxygen species, allowing NRF2 to induce an antioxidant response.52 GSK-3B, however, has been
found to induce degradation independent of KEAP1, thereby reducing the cell’s antioxidant
capacity as well as its sensitivity to reactive oxygen species.49,50 Furthermore, GSK-3B helps to
impair neuronal GABAergic signaling through degradation of Gephyrin, a scaffolding protein at
the postsynaptic site.48,53 Gephyrin scaffolding binds GABA-A receptors and closely groups
them at the synapse. However, SI mice were observed to have reduced tonic as well as
spontaneous inhibition by GABA, which was associated with reduced GABA-A receptor
clustering.6 This indicates that, consistent with the reported data, long term overactivity of GSK3B significantly disrupts neuronal antioxidant capacity as well as signaling behavior.
The effects of GSK-3B on GABA sensitivity underscores a broader disruption in
GABAergic signaling with significant implications for neuronal health. While impaired GABA
signaling is a common hallmark of memory impairments in AD and is reported in SI mouse
models, the dynamics of this change are more complex and allude to a broader cellular
dysfunction.8,21,54,55 In one experiment using a similar model of social isolation, SI mice
experienced an increase in tonic inhibition but not spontaneous inhibition, contrary to previous
reports in which both were reduced.6,8 This was identified as being due to an increased release of



13
astrocytic GABA, with hippocampal astrocytes demonstrating elevated intracellular Calcium as
well as increased expression of MAO-B, the primary enzyme involved in astrocytic GABA
synthesis.8 Furthermore, this effect was found to be dependent on two transporters: TRPA1, a
Calcium channel which was upregulated in the astrocytes of SI mice,8,44 as well as BEST1, a
Calcium-sensitive Chloride channel which has been regarded as a primary mechanism of
astrocytic GABA efflux.8,21 Similar changes in GABA synthesis were also observed in astrocyte
cultures treated with Amyloid-Beta as well as in mice subjected to chronic ethanol
consumption.35,36,56 Additionally, mice treated with chronic corticosterone showed an increase in
peroxide content in the hippocampus that was alleviated by TRPA1 inhibition.9.44 Contrary to
this, however, TRPA1 content in the hippocampus was reduced by chronic corticosterone
content, suggesting a negative feedback mechanism in response to the increased intracellular
calcium.8 This would provide clearer context for the reported decrease in GABA sensitivity in
the hippocampus, as prolonged SI would require negative feedback. This presents a key role for
GSK-3B in this model, as activation of GSK-3B would mediate GABA desensitization through
breakdown of Gephyrin.
3.2 Disruptions in the Urea Cycle and Metabolism
This pattern of aberrant GABA signaling presents a pathology with significant
implications for neuronal health in the hippocampus, as indicated by reported increases in
intracellular calcium and oxidative stress. In a mouse model of ischemic stroke, overproduction
of astrocytic GABA was observed in cortical regions affected by ischemia, with elevated
astrocytic GABA as well as increased expression of MAO-B and inducible Nitric Oxide
Synthase (iNOS).21 The latter, iNOS, is an enzyme that has been identified as involved in the
development of anxiety and depressive symptoms in SI and corticosterone-treated mice.57,58



14
Statistical analysis showed that there was a negative correlation between astrocytic GABA and
neuronal glucose metabolism in these regions.21 This association was further confirmed by sitespecific manipulation of MAO-B, which demonstrated MAO-B to be both necessary and
sufficient to impair neuronal glucose metabolism.21 Additionally, MAO-B poses significant risk
to cell survival through both its byproducts, which are ammonia and hydrogen peroxide, as well
as its substrate, putrescine, which is converted from ornithine in the Urea Cycle.56,59,60 This effect
was observed both in amyloid-beta-treated astrocyte cultures as well as in post-mortem
hippocampal samples, as the expression of several enzymes involved in the Urea Cycle had been
elevated.22,56 These samples showed increased concentrations of ammonia, which is the starting
substrate for the Urea Cycle and has been demonstrated to induce amyloidogenesis in
astrocytes.56,60
From this, activity of the Urea Cycle can be identified as a key factor in constructing a
model of neurodegeneration. As observed from previous studies, multiple enzymes involved in
the Urea Cycle were up-regulated in models of AD and ischemic stroke, and that selective
knockdown of these enzymes was able to restore cellular and cognitive function and improved
autophagy of amyloid-beta.21,56 Several studies have linked hyperammonemia with neurological
dysfunction, as treatment of astrocytes with ammonia resulted in increased production of
Amyloid Precursor Protein (APP) as well as a reduced rate of APP degradation.56,60 This
indicates a causal relationship between dysfunction of the Urea Cycle and amyloidogenesis, with
amyloid-beta seemingly produced as a response to increases in ammonia concentration.
Although the function of Amyloid-Beta under physiological conditions is not well understood, it
has been identified as interacting with multiple neuronal and non-neuronal surface proteins,61,62



15
suggesting that it acts to initiate a global shift in cellular machinery under hyperammonemic
conditions.
Figure 3: Interactions of the TCA and Urea Cycles
Representation of the TCA (left) and Urea (right) cycles within the cell. Pathways interact at the
regeneration of fumarate from aspartate with side reactions resulting in the production of
neurotransmitters GABA and Glutamate. Efflux of these neurotransmitters in disease states
results in dysregulation of both processes, impairing energy production and ammonia processing.
Created using BioRender.com
These shifts in Urea Cycle activity have further implications for cellular metabolism
through the TCA Cycle and the cell’s generation of energy, with the pathways intersecting



16
through the reactions involving aspartate and fumarate.56,63 In addition to fumarate being a
primary metabolite in the TCA Cycle, fumarate is also regenerated from aspartate as a byproduct
of the Urea Cycle.56,63 This reaction is mediated by Aspartate Transaminase, which
simultaneously metabolizes the excitatory neurotransmitter glutamate for use in the TCA
Cycle.63,64 Multiple investigations into neurodegeneration have reported significant decreases in
cellular energy, including decreased glucose metabolism,56 depleted ATP concentrations,6,65,66,
and down-regulations of Complexes I, II, and IV of the Electron Transport Chain (ETC).9,67
Notably, Complex II also demonstrated reduced capacity in comparison to Complexes I and
IV.67 Complex II, also known as Succinate Dehydrogenase, is directly involved in the TCA
Cycle, as it mediates the conversion of succinate to fumarate and is a rate-limiting step in the
production of ATP.63,67 Additionally, in the 5xFAD model of AD, neuronal concentrations of
both fumarate and aspartate were found to be reduced in the hippocampus of 5xFAD mice.63 This
effect was limited to metabolites of the TCA Cycle, as the concentrations of GABA and
glutamate synthesized from these reactions were found to vary between experimental models.63
As such, dysfunction to the Urea Cycle can be observed having a parallel, even causal
relationship with impaired production of cellular energy, with a consequent effect on cognitive
function.
3.3 ATP in the Synapse
The importance of energy homeostasis in the brain represents a significant risk factor in
the development of cognitive dysfunction, as reductions to ATP generation and increased efflux
of adenosine were observed in AD models.68,69 Changes in ATP production present an additional
risk to neurological function through the involvement of adenosine as an extracellular signaling
molecule: multiple classes of purine receptors are expressed on the cell surface, including the A1,



17
A2A, P1, and P2 receptors.68,70 These receptors are activated in response to extracellular
adenosine and other purines which have been effluxed from the cell, which can occur under
various conditions.68,70,71,72 Notably, this phenomenon has been observed to be further induced in
the hippocampus under pathological conditions such as anxiety, ischemia, and traumatic brain
injury, with hippocampal astrocytes having been identified as a major candidate for a primary
source.71,72,73 Furthermore, efflux of adenosine was influenced by changes in intracellular
calcium, suggesting a causal mechanism between elevated calcium as a result of cellular stress
and the subsequent release of adenosine as an inflammatory response.72,73,74 Under normal
physiological conditions, this reaction is likely to serve as a signaling mechanism in response to
stress, as the release of astrocytic adenosine was found to display an anxiolytic effect in animal
models of stress and resulted in increased anxious behavior when efflux was prevented.73
Conversely, however, this phenomenon appears to be a contributing factor under chronic models
of inflammation, including multiple models of AD.
The involvement of purine receptors in AD pathology has been largely recognized as
both a potential mechanism as well as a therapeutic target for disorder, with observed
correlations between the progression of AD and the activity of purine receptors.70,75, Of these
receptors, the receptor P2X7 has been identified as a promising candidate,70,75,76 with P2X7
activation being linked to the formation of Amyloid-Beta plaques.75,77,78 Moreover, activation of
the P2X7 receptor has been linked to increased activity of GSK-3B, which has also been linked
to cellular dysfunction as well Amyloid secretion.77,79 This association has led to the hypothesis
that the Amyloid-Beta peptide acts as an agonist for the receptor and induces neuroinflammation
through activation of glial cells.75,76 However, this proposal has been called into question, as
recent investigations have failed to demonstrate that the amyloid-beta peptide acts as a direct



18
P2X7 agonist.76 As such, Amyloid-Beta is more likely to indirectly mimic the effects of P2X7
activation, as incubation with amyloid-beta was able to induce release of cellular ATP as well as
the subsequent activation of P2X receptors.80 While this demonstrates the involvement of P2X
receptors in the pathogenesis of AD, it also illustrates their role as part of a larger signaling
cascade rather than as a direct target or mechanism. Therefore, while the P2X7 receptor presents
a means through which the ATP-mediated signaling cascade may be perpetuated, it fails to
demonstrate how this process is initiated, nor does it adequately illustrate the mechanism by
which Amyloid-Beta affects this process.
An alternative candidate would be the A2AR receptor, which has been shown to be
significantly upregulated in the brains of AD patients.81,82 Overexpression of the A2AR receptor
in the Hippocampus is an age-associated phenomenon that is further exacerbated in AD.82,83,84
This has prompted the investigation of A2AR as a potential biomarker for AD as well as a novel
therapeutic target,81 as memory impairments observed in AD mice were ameliorated by
pharmacological blockade of A2AR.82,85 Conversely, induction of A2AR was found to exacerbate
memory deficits in the same model, with overexpression being shown to induce age-associated
cognitive deficits in young mice.82,84 This suggests a causal mechanism between upregulation of
the A2AR receptor with age and the development of AD pathology. However, the A2AR receptor
also appears to play a significant role in injury, as A2AR activation was found to be linked to
models of neurological dysfunction including traumatic brain injury and kainate-induced
convulsions.86,87,88 A2AR mediated neurotoxicity under kainate-induced convulsions through
regulation of glutamate release and homeostasis, suggesting that the contribution of A2AR to AD
pathology is cumulative with injury.87,88,89 Moreover, the neurodegenerative effects of A2AR
activation were associated by increased activation of GSK-3B and subsequent Tau



19
phosphorylation at the PHF-1 region,86 strongly suggesting that A2AR is a key factor in AD
pathogenesis.
Figure 4: Adenosine Signaling and Synaptic Function
Schematic of A2AR activation in the synapse. Efflux of adenosine (red) via ENT is induced by
TrkB-BDNF signaling. A2AR activation induces presynaptic release of glutamate (purple) and
inhibits astrocytic uptake by GLT-1. A2AR activation sensitizes GR activation by corticosterone
(orange) against inhibition by BDNF. Combined GR activation and excitotoxicity induce
calpain-mediated cleavage of GSK-3B and TrkB, altering signaling within the cell. Created using
BioRender.com
A2AR activation plays an important role in synaptic activity and memory encoding.88,90 In
the dentate gyrus (DG), which is the primary region of input for the hippocampus, A2AR



20
activation was found to mediate LTP via retrograde signaling.88 Repeated burst firing in the DG
was accompanied by an increase in amplitude of excitatory postsynaptic currents (EPSCs) at the
affected synapse. This effect was found to be dependent on the presynaptic expression of A2AR
and the postsynaptic expression of tyrosine receptor kinase B (TrkB).88 TrkB is the primary
receptor of brain-derived neurotrophic factor (BDNF), a neurotrophin which promotes neuronal
cell survival and has been observed to be decreased in the brains of AD patients.91,92 The role of
A2AR in mediating LTP was determined to be through a retrograde signaling pathway in which
activation of postsynaptic TrkB induced the release of adenosine into the synaptic cleft with a
subsequent A2AR-induced release of glutamate.88 This pathway was also found to be involved in
the initiation and neurodegenerative effects of kainate-induced seizures, as kainate treatment
resulted in an upregulation of A2AR and a consequential increase in glutamate release.87,88
Furthermore, A2AR has been shown to play an important role in astrocytic glutamate
homeostasis, which is necessary for the clearance of neurotransmitters from the synapse.
Knockout of astrocytic A2AR was found to induce schizophrenia-like symptoms in mice through
upregulation of Glutamate Transporter-1 (GLT1), increasing the rate of glutamate clearance and
producing cognitive and locomotor impairments.89 This demonstrates that A2AR plays an
important role in mediating glutamate signaling in the synapse and that aberrant A2AR activity is
sufficient to induce a pathological state.
In the case of AD, A2AR demonstrates a significant increase in activation, which has been
demonstrated by increased expression in the hippocampus of aged adults and AD patients.81,82,83
Similar changes in A2AR expression were observed in multiple models of AD, in which blockade
of A2AR alleviated cognitive deficits and restored cellular survival and LTP in the
hippocampus.82,84,85 Furthermore, upregulation of A2AR induced in a transgenic mouse model of



21
AD was found to further exacerbate AD pathology, with greater impairments in memory and
increased Tau phosphorylation at the AT8 region. However, these changes were modest, with no
changes observed in Amyloid content or in phosphorylation of other Tau regions.84 This suggests
that, while A2AR activation plays a significant role in AD pathogenesis, the effect already
approaches its maximum in AD models. This relationship is further supported by the role of
BDNF in its neuroprotective effects against Amyloid toxicity. As previously observed, BDNF
acts as part of a retrograde signaling pathway with A2AR to induce LTP.88 However, although
BDNF exerted a neuroprotective effect in cultured neurons, this effect was found to be
independent of A2AR, suggesting a decoupling of the BDNF-TrkB-A2AR pathway.93 This was
consistent with reports finding that Amyloid interferes with BDNF signaling through the TrkB
receptor, as neuron cultures incubated with Amyloid-Beta demonstrated a shift from the fulllength TrkB receptor to its truncated non-functional isoform (TrkB.T).92,93 Given the
involvement of TrkB in the neurodegenerative effects of kainate-induced convulsions,87,88 this
represents a potential compensatory mechanism against inflammatory processes.
This shift in TrkB function indicates an intracellular signaling pathway which is induced
by Amyloid-Beta. A partial mediator of this effect was the increased transcription of the TrkB.T
isoform in response to excitotoxic conditions, which is a characteristic of neuroinflammatory and
neurodegenerative disorders.92,94 The increased expression of TrkB.T was found to exert an
inhibitory effect on the full length TrkB, further diminishing normal BDNF-TrkB signaling.94
Conversely, full length TrkB was subjected to proteolytic cleavage by Calpain, which was
induced by Amyloid-Beta.92,93,94 Activation of Calpain has been previously described as part of
the cellular response to stress and the subsequent activation of GSK-3B, indicating that TrkB
modification acts in parallel to this pathway.46,47,48 This is further confirmed by reported



22
interactions between TrkB and the Glucocorticoid Receptor (GR), which demonstrate a mutually
antagonistic relationship.91,95,96 Animals subjected to chronic stress displayed a widespread shift
from the full length to the truncated TrkB isoform across multiple brain regions.96 Furthermore,
BDNF was found to induce site-specific phosphorylation of GR, which was necessary for
synaptic maintenance and learning.95,97 This phosphorylation was disrupted under chronic stress
and resulted in impaired memory and increased mortality in AD models without affecting
Amyloid-Beta deposition.95 These effects were further amplified by overexpression of A2AR, as
it induced transcriptional activity of GR in the nucleus and sensitized the hippocampus to GR
agonism.98,99 In the context of synaptic activity, this suggests that A2AR activates GR as a
negative feedback mechanism against BDNF-TrkB signaling. Additionally, this provides a
substantial link between the cellular effects of chronic stress and the pathogenesis of AD, as both
are mediated by the age-associated increase in A2AR activation. And although the direct
relationship between Amyloid-Beta and A2AR activation remains unclear, it presents a novel
image of Amyloid-Beta deposition as part of the brain’s natural inflammatory response.



23
Chapter 4: Neural Pathways
4.1 A Primary Neural Pathway
Changes to cellular function in the hippocampus radiate outwards onto connected regions
of the brain, thereby causing complex changes in behavior. Understanding the links between
these connected regions is a necessary point in developing a clear model of neurodegeneration,
as the complex interactions between them shed light on how these disorders neuronal damage
local to the hippocampus rather than its signaling to other regions of the brain. Therefore, the
implications on behavior cannot be properly understood without the context of hippocampal
connectivity. Previous studies investigating the neurological action of the gut microbiome on
social behavior found significant roles by the hippocampus in conjunction with the hypothalamus
and the bed nucleus of the stria terminalis (BNST).12,13 This demonstrated a causal relationship
between depletion of the gut microbiome and elevated plasma corticosterone, resulting in
impaired social behavior.100 Similar studies into the ventral tegmental area (VTA) found similar
behavioral impairments, though these studies had investigated the role of oxytocin, specifically
onto the dopaminergic neurons of the VTA.13,14 Both induction of corticosterone signaling and
ablation of oxytocin signaling were observed to be sufficient to induce the observed social
impairments, while impairments caused by depletion of the gut microbiome could be reversed by
ameliorating these signaling mechanisms.100,101 Given previous findings that both corticosterone
and oxytocin were shown to induce neuronal spiking in the hippocampus13,100,101,102 as well as
projections by the VTA onto the hippocampus, it is likely that corticosterone and oxytocin act as
redundant signaling mechanisms in social behavior.
This redundant signaling is likely to have multiple factors which coordinate and regulate
the intensities of each pathway. One such factor is expected to be FGF21. FGF21 plays an



24
Figure 5: Neural Pathway Regulating Behavior
A diagram mapping the connections in the proposed neural pathway. Above pathway shows
signaling cascade by CORT and OXT. Dotted lines represent brain regions that are involved in
both pathways. FGF21: Fibroblast Growth Factor 21; CORT: Corticosterone; OXT: Oxytocin;
VTA: Ventral Tegmental Area; DR: Dorsal Raphe Nuclei; BNSTDM: Bed Nucleus of the Stria
Terminalis, Dorsomedial; BNSTAL: Bed Nucleus of the Stria Terminalis, Anterolateral; BNSTPR:
Bed Nucleus of the Stria Terminalis, Principal; BNSTFU: Bed Nucleus of the Stria Terminalis,
Fusiform; BNSTOV: Bed Nucleus of the Stria Terminalis, Oval; CeA: Central Amygdala; NAcc:
Nucleus Accumbens; PVNCRF: Paraventricular Nucleus, CRF-Secreting; PVNOXT:
Paraventricular Nucleus, Oxytocin-Secreting. Created using BioRender.com



25
interesting role in this signaling pathway because it induces the transcription of both
corticosterone103 and oxytocin,104 thereby increasing and synchronizing their respective
signaling. In particular, FGF21 and corticosterone appear to express a bi-directional feedback, in
which FGF21 induces the synthesis of corticosterone from the adrenal glands which
subsequently induces FGF21 synthesis in the liver.103 In addition, transcription of both oxytocin
and FGF21 appear to be NRF2 dependent, indicating that induction of NRF2 is able to induce
both upstream and downstream effectors of this pathway.104 The importance of FGF21 in the
induction and coordination of this neuronal pathway provides a possible mechanism for agedependent cognitive decline, as levels of FGF21 in the plasma, liver, and brain are reported to
experience an age-related decrease.20
As previously commented, a recent study demonstrated a joint action between the
hippocampus and the BNST modulating social behavior under conditions of microbial depletion
as well as induction of corticosterone signaling.100 Notably, however, observations regarding
blockade of corticosterone signaling onto the BNST was more nuanced: whereas social behavior
was restored in microbe-depleted mice, the opposite effect was observed in mice with intact
microbiomes, indicating paradoxical changes in activity at this region. It is important to note that
the BNST is a complicated region of the brain, with distinct subnuclei projecting to diverse
regions of the brain, including the VTA, the amygdala, and the hypothalamus, giving it
involvement in various signaling pathways.18 This is important to note, as manipulation of
corticosterone signaling was performed through injection of a viral vector into the BNST.100 As
such, transfection was likely distributed across the subnuclei of the BNST, thereby affecting the
different signaling pathways and yielding paradoxical results.



26
Fortunately, many of the BNST subnuclei have already been well characterized.18,105,106
Two important regions are the dorsomedial105 and anterolateral areas106 of the BNST. These
regions are of particular interest because of their mutual outputs onto the parvocellular neurons
of the PVN,105,106 which controls secretion of CRF and initiates the Hypothalamic-PituitaryAdrenal (HPA) axis.107 The HPA axis regulates the secretion of glucocorticoid hormones into
circulation, which is a major contributor to the cellular dysfunction observed in models of
chronic stress. Furthermore, the HPA axis has been demonstrated to be subject to dysregulation
in age-associated increases A2AR expression, which further exacerbates the effects of
corticosterone in the synapse.98 The dorsolateral and anterolateral areas of the BNST possess a
high number of inhibitory GABAergic projections,18,105,106 indicating that their signaling onto the
PVN serves as an important negative feedback mechanism onto the HPA. Importantly, both of
these regions receive input from the hippocampus and the VTA105,108,109 and display mutual
innervations with the dorsal raphe nucleus,105,110 indicating a coordinated signaling. Notably,
however, the dorsomedial nucleus of the BNST also projects onto the magnocellular neurons of
the PVN,105 thereby halting further secretion of the oxytocin signaling and limiting the negative
feedback onto the HPA axis.
An additional point of interest in the BNST is the principal nucleus, which receives input
from CRF secreted by the PVN and sends projections backwards to the upstream anterolateral
area.18,111 This region is of particular interest because of its strong dual-expression of the kappaopioid receptor receptor (KOR)18,112 and of CRF receptor type 2 (CRFR2).18,19,113 KOR is the
primary receptor to dynorphins, an endogenous opioid which is associated with stress-induced
behavior as well as the reinforcement of drug seeking and depression-like behaviors.114,115,116
Interestingly, CRFR2 was also found to be involved in response to aversive stimuli, as blockade



27
of CRFR2 was able to ameliorate stress response due to opioid withdrawal117 as well as improve
resilience against the development of PTSD-like symptoms under repeated negative stimuli.19
Additionally, CRFR2 was found to mediate KOR signaling, inducing conditioned-place aversion
(CPA) similar to treatment with KOR agonists. However, CRFR2 was indicated to be a
downstream signal to KOR, as blockade of KOR prevented CRFR2-induced aversion but not
vice versa.114 Furthermore, the role of CRFR2 in development of anxiety and PTSD-like
symptoms presents further complications, as conflicting reports conclude that the expression of
CRFR2, specifically in the BNST, reduces anxiety and improves response to stress.118,119
These apparent contradictions are likely due to differences between the anticipation of
and response to a negative stimulus. Activation of CRFR2 improves anxiety scores in tests such
as the elevated plus maze and open field tests,119 which measure behavior in anticipation to
danger. This is distinct to previously mentioned models such as opioid withdrawal and CPA, in
which negative associations are already encoded. This distinction is further emphasized between
models of PTSD, in which blockade of CRFR2 was found to improve symptoms in response to
repeated negative stimuli,19 whereas the opposite relationship was observed after incubation of
animals with a predator-associated odor.118 This presents the CRFR2 signaling of the principal
nucleus as a candidate for valence detection. Furthermore, it demonstrates a mechanism of
consolidating memory of negative stimuli, as it prolongs activation of the HPA axis through
inhibition of the upstream anterolateral area.18,111
4.2 Signaling Anticipation and Reward
This pathway presents a novel and concise model to better understand the connected roles
of these distinct regions as well as their involvement in pathological conditions, including
neurodegenerative diseases and disorders such as addiction or trauma. However, as previously



28
stated, this pathway appears to differentiate between the anticipation of a stimulus and the
stimulus itself,19,114,118,119 suggesting that further modulation occurs elsewhere in the brain before
feeding back into the above regions. In particular, this includes two additional regions of the
Figure 6: MOR-CRFR1 Synaptic Modulation
Co-localization of CRFR1 and MOR receptors at the Central Amygdala and the Anterolateral
Area. Release of CRF (blue) by the Oval and Fusiform Nuclei induces presynaptic release of
Glutamate (purple) with increases in postsynaptic excitation. Release of enkephalin (green) by
the Oval Nucleus eliminates postsynaptic excitation by inhibition of CRFR1 receptors. Created
using BioRender.com
BNST: the oval and fusiform nuclei.120 Both regions receive innervation by the nucleus of the
tractus solitarius (NTS),105,120 which is a major source of norepinephrine in the brain. However,



29
in a cue-conditioned model of drug delivery, release of norepinephrine was associated with the
cued stimulus followed by release of dopamine onto the oval nucleus upon drug
administration.121,122 This demonstrates a sequential signaling process in the BNST in which the
anticipation of reward precedes the rewarding stimulus, thereby enabling distinction between the
two. Additionally, of the oval and fusiform nuclei, the oval nucleus displays greater outputs onto
the fusiform than vice versa.120 This could help to serve as a negative feedback during the
anticipatory phase, whereas subsequent dopamine release onto the oval nucleus would create a
shift in balance between the two.
Understanding the interactions between these nuclei requires identification of their
mutual signaling targets. Both the fusiform and oval nuclei have been observed to have
projections onto the central amygdala and the anterolateral area of the BNST,120 the latter being a
core region in the previously described pathway. The central amygdala and the anterolateral area
share a distinct similarity in their receptor distributions, as both were found to have a significant
co-localization of the CRF-receptor type 1 (CRFR1) and the mu-opioid receptor (MOR).123 The
latter is the primary receptor for another class of endogenous opioids, enkephalins, and is
identified as the main mechanism of action for addictive opiates such as heroin and morphine.124
In both the central amygdala and the anterolateral area, approximately 63-66% of CRFR1-
expressing dendrites were found to co-localize with MOR, with a primary placement at synapses
which receive excitatory input.123 This suggests that both CRFR1 and MOR express postsynaptic roles in modulating the response to excitatory signals in these regions. However, these
roles were likely contradictory to each other, as presynaptic CRFR1 was largely found in the
axons of excitatory synapses whereas MOR was expressed in those that were inhibitory. Taken
together, this suggests that CRFR1 plays a role in increasing sensitivity to glutamate and other



30
excitatory signals whereas MOR subsequently desensitizes the cell. This reflects the sequential
activation of the fusiform and oval nuclei,121,122 as CRF is expressed in both regions while
enkephalin is mainly expressed in the oval nucleus.
Interestingly, mutual interactions between the anterolateral area and the central amygdala
appear to play an important role in this pathway: both the anterolateral and dorsomedial areas of
Figure 7: KOR-mediated Blockade of GABA Signaling
Synaptic signaling from the Central Amygdala onto the Anterolateral Area. Inhibitory signaling
is blocked by release of dynorphin (pink) onto presynaptic KOR receptors. Negative feedback is
restored upon cessation of dynorphin and release of GABA (orange). Created using
BioRender.com



31
the BNST project onto the central amygdala,105,106 which itself sends projections backwards to
the anterolateral area.125 The mutual inhibition by the anterolateral area and the central amygdala
potentially serves as a negative feedback loop, preventing overexcitation of either by the oval
and fusiform nuclei.120 However, these signals are not entirely symmetrical, as inhibitory
projections from the central amygdala onto the anterolateral area were found to express KOR at
the presynaptic site which prevented the release of GABA, thereby blocking inhibitory
signaling.118 This offsets the mutual inhibition of the two regions by preventing negative
feedback from the central amygdala onto the anterolateral area but not vice versa, placing
heavier restriction on the former. This effect may also be mediated by the oval and fusiform
nuclei, as both areas significantly express dynorphin112 in addition to their above roles in
glutamate sensitivity. This presents an additional means of differentiation between anticipation
and reward: similar to its role in the principal nucleus of the BNST,114 dynorphin signaling in the
anterolateral area may help in distinguishing anticipation, as its deactivation and the
simultaneous release of enkephalin would work in tandem to restore negative feedback.
The central amygdala as well as the fusiform nucleus both project onto the nucleus
accumbens,120,126 emphasizing the importance of dopamine signaling in this pathway. The
nucleus accumbens receives dopamine input from the VTA and has been identified as a key
region in reinforcement of reward as well as addiction.15 As a mediator in reward signaling,
inhibitory input into the nucleus accumbens during the anticipatory phase could represent a
priming in this pathway, as the subsequent disinhibition may produce a rebound in activity that is
cumulative to input from the VTA. This is observable on a cellular level, as reductions in
astrocytic glutamate release were a necessary condition for synaptic reinstatement of cocaineseeking behavior.29 This is supported by the proposed roles of dynorphin and enkephalin in



32
switching from the anticipatory to the reward phases as well as their respective involvements in
the reinforcement of addictive behavior. To this end, the nucleus accumbens modulates this
pathway via its GABAergic projections from the nucleus accumbens shell to the dorsomedial
nucleus,127,128,129 in which inhibition of the nucleus accumbens during the anticipatory phase
consequently disinhibits the dorsomedial nucleus. This demonstrates a priming mechanism for
signaling onto the PVN, as both the anterolateral and dorsomedial areas see increased activation
during the anticipatory phase. This also demonstrates a strict control over the pathway, as
dopamine release by the VTA restores negative feedback between the anterolateral area and the
central amygdala, bringing activation of this pathway to an abrupt halt. Recognizing this nuanced
behavior in signal timing is important for modeling the interactions between brain regions under
neurodegeneration.
4.3 Rewiring of the VTA
From this information, the VTA can be identified as having a critical role in modulating
this pathway, as the release of dopamine from the VTA appears to initiate a shift in equilibrium
between opposing signals. This is further emphasized by plasticity of the VTA on the cellular
level, as the VTA shows a bimodal response to CRF input, as excitation of the VTA in response
to CRF was diminished over increasing time and concentration. This effect was identified to be
due to differential action between presynaptic CRFR1 and CRFR2 receptors, with an initial
increase in glutamate release mediated by CRFR1 which was subsequently attenuated by
CRFR2, which induced the release of GABA onto the presynaptic site.130 As a source of CRF
which projects onto the VTA, this has particular implications for the dorsomedial nucleus,105,112
as output from the VTA onto the hippocampus and the nucleus accumbens send competing
excitatory and inhibitory signals back towards the dorsomedial nucleus.105,109,129 As a primary



33
source of dopamine and a key region in drug addiction, the impact to the VTA cannot be
understated: in animals which self-administered cocaine, the attenuating effect of CRFR2 was
diminished, as administration of a CRFR2 agonist saw a diminished effect on both glutamate and
GABA tone.130 This indicates a significant change on the cellular level of the VTA’s ability to
regulate its response to CRF during the anticipatory phase, potentially sensitizing this signaling
pathway to drug-associated cues.
A further indicator to this was the response of the VTA under conditions of stress: in
contrast to healthy mice, animals which experienced both cocaine self-administration as well as
yohimbine-induced distress saw a shift in CRFR2 activity from attenuating to sensitizing.130 This
is a stark difference because the role of the CRFR2 receptor is essentially reversed in this
condition, potentially amplifying the excitatory effect of CRF over a longer period. The cause of
this shift was identified as a response to adenosine signaling, as blockade of the A1 receptor was
able to mitigate sensitization by CRFR2 despite showing no impact in previous test conditions.
This suggests that, under conditions of stress and drug-associated cues, there is an increase in
extracellular adenosine that is not observable under other conditions, and that this increase
mediates a decrease in GABAergic tone through activation of the A1 receptor. This increase in
extracellular adenosine may have been mediated by efflux of ATP from nearby astrocytes, as
observed in the hippocampus. Notably, the increase in glutamate signaling by CRFR2 was
ablated by blockade of the GABA-B receptor. Because of the drop in GABA tone by A1-CRFR2
interactions, the blockade of presynaptic GABA receptors would be anticipated to have the
opposite effect, further preventing the release of glutamate. This suggests a non-neuronal
mechanism, likely nearby astrocyte cells, as GABA-B receptor-mediated increase in intracellular
Calcium could potentially result in efflux of ATP.131 Astrocytic involvement is further



34
Figure 8: CRFR1-CRFR2 Action in the VTA
Differential effects of CRFR1 and CRFR2 receptors in the VTA. CRFR1-induced release of
Glutamate (purple) is attenuated by CRFR2-mediated release of GABA (orange) onto the
presynaptic site. Attenuation by CRFR2 is blocked by astrocytic release of adenosine (red) onto
the A1 receptor. Stress-induced release of adenosine by astrocytes is mediated by activation of
the CRFR2 and GABA-B receptors. Created with BioRender.com
implicated by evidence of a cocaine-induced decrease in number of cells in the VTA expressing
S100B, an astrocyte-specific marker, as well as increased colocalization of S100B and CRFR2,
suggesting increased astrocyte sensitivity to CRF.132
The changes in CRF signaling as well as the changes to CRFR116,17,130 and
CRFR2130,132,133 activity represent a distinct time-dependent mechanism for the development of



35
dependence. Acute withdrawal after prolonged chronic ethanol exposure was shown to attenuate
CRF-induced increases in excitatory signaling onto the VTA while producing a significant
increase in glutamatergic tone.16 Moreover, this increase in glutamatergic tone was found to be
CRFR1 dependent, thereby coinciding with observed changes in CRFR2 activity under stress.130
Blockade of CRFR1 in the VTA was shown to limit binge-like drinking as well as prevent
reinstatement of drug-seeking behavior after stress.16,17 However, as with previous observations,
this effect was prevented by simultaneous blockade of CRFR2.17,130 This change in activity
appears to extend beyond the VTA into the nucleus accumbens and the BNST, with excitability
in these regions increasing after social isolation and chronic ethanol exposure, respectively.133,134
In the nucleus accumbens, release of dopamine after depolarization is significantly increased
following social isolation, with blockade of local CRFR1 being unable to attenuate dopamine
release in contrast to control rats, indicating a decrease in sensitivity.133 Notably, a similar
behavior was observed in the central amygdala, as CRFR1 was found to be an important
mediator of GABAergic signaling during binge-like drinking and withdrawal. This is consistent
with the proposed role of CRFR1 in sensitizing the central amygdala to excitatory input. Taken
together, this reinforces the overlapping mechanisms in the pathology of addiction and social
isolation, as disruptions to CRF molecular signaling result in aberrant behavior. Furthermore, it
creates a concrete image of the impact of stress on neuropsychiatric disorders, which can thus be
used to model the progression of neurodegenerative disorders in response to environmental
stressors.



36
Chapter 5: Future Investigations
5.1 Insulin Resistance in the Brain
Insulin plays a significant role in the brain as it helps to modulate cellular energy and cell
survival. Conversely, insulin resistance has been shown to act as a mechanism for the
development of cognitive impairment, in particular during the early stages of AD.100,135,136 This
has been identified as being mediated by Insulin Receptor Substrate 1 (IRS1), a signaling protein
which mediates the intracellular response to insulin and shows reduced activation in multiple
models of AD.135,136 Multiple factors involved in the pathology of AD have been observed to
reduce the activation of IRS1, including incubation of Beta-Amyloid as well as phosphorylation
by GSK-3B, both of which were recognized in the sections above as playing a key role in the
pathogenesis of AD.137,138
This is notable as it demonstrates an overlapping pathology between animal models of
stress and diabetes, as diabetic models of mice showed similar patterns of Tau phosphorylation
to SI and corticosterone-treated mice.11,139 Conversely, administration of intranasal insulin was
able to rescue activation of IRS1 and reduce activity of GSK-3B through induction of the PI3KAKT axis,140 suggesting a negative feedback mechanism between the two proteins. This is not
the first example of impaired energy homeostasis in the brain in AD pathology, as was observed
with reductions to ATP generation and the efflux of adenosine as a signaling mechanism for
neuroinflammation.68,69,70 Additionally, IRS1 can be inferred to have a distinct role in the
pathogenesis of AD, notably through Tau phosphorylation, as the expression of Tau protein is
necessary for IRS1 signaling.141 As both proteins have been shown to be targets of GSK-3B
activity, this presents a potential pattern of enzyme activity within the cell during the early stages
of AD.34,138 However, the role of this shift in activity under physiological conditions, as well as



37
the interactions between Tau expression and insulin signaling in the brain, both remain largely
unclear.
5.2 Influence of the Gut Brain Axis
As previously commented, studies into the gut microbiome demonstrate a causal
mechanism between dysbiosis and impaired social behavior, disrupted neuroendocrine signaling,
and aberrant activity in the hippocampus, the VTA, and the BNST.12,13,18 These effects are
mediated by excess concentrations of plasma corticosterone and diminished secretion of
oxytocin from the PVN of the hypothalamus, which serve as part of a redundant signaling
mechanism to activate the proposed primary pathway.12,13 This phenomenon was found to occur
in animal models of microbial depletion as well as in genetic models in which the animal in
which the microbiome had been disrupted but not otherwise depleted. This presents a distinct
chain of events in which an environmental or physiological factor induces specific gut dysbiosis
which hampers endocrine signaling to the brain and results in aberrant neurological and
behavioral responses. More specifically, these symptoms were found to result from the loss of
two specific taxa of bacteria: L. reuteri and E. faecalis, both of which are Lactic Acid
Bacteria.142,143 This raises important questions into the behavior of Lactic Acid Bacteria in the
gut microbiome and its contribution to neurological health. Additionally, this raises questions
regarding the effects of conditions known to impact the gut microbiome, such as alcohol
consumption, which creates various shifts in microbial content while also disrupting the
intestinal environment.144,145
5.3 Liver Secretion of FGF21
Similar to the microbiome, liver secretion of FGF21 plays an important role in
maintaining cognitive health, in particular through the coordination of corticosterone and



38
oxytocin signaling to the brain.103,104 FGF21 has been shown to exert strong neuroprotective
properties in the hippocampus while also suppressing alcohol consumption by modulating
activity of the basolateral amygdala.20,104,146 Conversely, FGF21 secretion decreases with age,
presenting a possible link to age-related cognitive decline.20 Additionally, FGF21 has been found
to mediate the inactivation of GSK-3B,147 which has been previously addressed as being a key
mediator in multiple pathologies in AD models, including Tau hyperphosphorylation and
inactivation of IRS1.27,138 The action of FGF21 in the brain presents an important means in
regulating metabolic function under cellular stress. However, the role of FGF21 in maintaining
neurological health, as well as the broader interactions between the brain and liver function,
requires deeper investigation.



39
Chapter 6: Conclusions
Neurodegenerative diseases such as Alzheimer’s and Parkinson’s are still poorly
understood, as their complex pathologies make it difficult to characterize the mechanisms behind
their progression. Similarly, much is unknown about the psychiatric disorders which are often
comorbid with them, presenting a significant obstacle in the treatment of patients afflicted with
one or multiple of these conditions. These disorders affect complex signaling pathways within
the brain, which results in a series of abnormalities cascading throughout the interconnected
regions. As such, the pathologies of these disorders must be understood in the global context of
how the individual regions influence each other in both healthy and in disease states.
The goal of this thesis has been to propose a viable model of neural activity across
multiple regions of the brain, including the hippocampus, the VTA, and the BNST, which
encapsulates a broad range of behaviors and neurological disorders. This model demonstrates
that the Hippocampus undergoes a series of complex changes in neuronal signaling, namely in
the release of astrocytic GABA responding to elevated intracellular calcium. This is an
instigating factor in the disruption of cellular energetics and the dismantling of the TCA and
Urea Cycles. These effects cascade downstream onto the BNST and the VTA, which mediate a
diverse network of interconnected regions involved in regulating behavior. The VTA plays an
important role in this network, providing a substantial driving force through the release of
dopamine and is shown to be inducible by several key factors. Moreover, it displays a distinct set
of plastic behaviors which are observable across various psychiatric disorders. This signaling is
further mediated by the diverse subsections of BNST, which helps to coordinate anticipation and
reward signaling through the timed release of dynorphin and enkephalin.



40
This model presents a significant step forward in understanding the brain, having
characterized not only neurodegeneration and addiction but also the brain’s responses to stress,
reward, and social interaction. Because of its complex structure, much is still unknown regarding
the various functions of the brain and their contributions to neuropsychiatric disorders.
Treatment for these disorders largely consists of pharmacological interventions which broadly
affect the brain’s signaling pathways, thereby resulting in diminished efficacy and a high
likelihood of side effects. Recent progress has evolved to include novel therapeutics such as
electrical stimulation of the brain as well as peptide-based drugs that are designed to target
specific proteins. However, these advances are limited by the available understanding of the
brain. This requires a more comprehensive understanding of the metabolic and signaling
pathways and their interactions at different regions. The model proposed by this thesis aims to
provide a foundation for future research into the brain’s complex processes.



41
Bibliography
1. Gupta, R., Advani, D., Yadav, D. et al. Dissecting the Relationship Between
Neuropsychiatric and Neurodegenerative Disorders. Mol Neurobiol 60, 6476–6529
(2023). https://doi.org/10.1007/s12035-023-03502-9
2. Briggs, Robert et al. “Drug treatments in Alzheimer's disease.” Clinical medicine
(London, England) vol. 16,3 (2016): 247-53. doi:10.7861/clinmedicine.16-3-247
3. Matej, Radoslav et al. “Alzheimer's disease and other neurodegenerative dementias in
comorbidity: A clinical and neuropathological overview.” Clinical biochemistry vol. 73
(2019): 26-31. doi:10.1016/j.clinbiochem.2019.08.005
4. Bayassi-Jakowicka, Martyna et al. “More than Addiction-The Nucleus Accumbens
Contribution to Development of Mental Disorders and Neurodegenerative Diseases.”
International journal of molecular sciences vol. 23,5 2618. 27 Feb. 2022,
doi:10.3390/ijms23052618
5. Padhi, Piyush et al. “Mechanistic Insights Into Gut Microbiome Dysbiosis-Mediated
Neuroimmune Dysregulation and Protein Misfolding and Clearance in the Pathogenesis
of Chronic Neurodegenerative Disorders.” Frontiers in neuroscience vol. 16 836605. 25
Feb. 2022, doi:10.3389/fnins.2022.836605
6. Silva, Joshua et al. “Modulation of Hippocampal GABAergic Neurotransmission and
Gephyrin Levels by Dihydromyricetin Improves Anxiety.” Frontiers in pharmacology
vol. 11 1008. 9 Jul. 2020, doi:10.3389/fphar.2020.01008
7. Joshi, Yash B et al. “Absence of ALOX5 gene prevents stress-induced memory deficits,
synaptic dysfunction and tauopathy in a mouse model of Alzheimer's disease.” Human
molecular genetics vol. 23,25 (2014): 6894-902. doi:10.1093/hmg/ddu412
8. Cheng YT, Woo J, Luna-Figueroa E, Maleki E, Harmanci AS, Deneen B. Social
deprivation induces astrocytic TRPA1-GABA suppression of hippocampal circuits.
Neuron. 2023 Apr 19;111(8):1301-1315.e5. doi: 10.1016/j.neuron.2023.01.015. Epub
2023 Feb 13. PMID: 36787749; PMCID: PMC10121837.
9. Al Omran, Alzahra J et al. “Dihydromyricetin ameliorates social isolation-induced
anxiety by modulating mitochondrial function, antioxidant enzymes, and BDNF.”
Neurobiology of stress vol. 21 100499. 30 Oct. 2022, doi:10.1016/j.ynstr.2022.100499
10. Al Omran, Alzahra J et al. “Social isolation induces neuroinflammation and microglia
overactivation, while dihydromyricetin prevents and improves them.” Journal of
neuroinflammation vol. 19,1 2. 4 Jan. 2022, doi:10.1186/s12974-021-02368-9
11. Dey, Aditi et al. “Glucocorticoid-mediated activation of GSK3β promotes tau
phosphorylation and impairs memory in type 2 diabetes.” Neurobiology of aging vol. 57
(2017): 75-83. doi:10.1016/j.neurobiolaging.2017.05.010
12. Wu, Wei-Li et al. “Microbiota regulate social behaviour via stress response neurons in
the brain.” Nature vol. 595,7867 (2021): 409-414. doi:10.1038/s41586-021-03669-y
13. Sgritta, Martina et al. “Mechanisms Underlying Microbial-Mediated Changes in Social
Behavior in Mouse Models of Autism Spectrum Disorder.” Neuron vol. 101,2 (2019):
246-259.e6. doi:10.1016/j.neuron.2018.11.018



42
14. Lee, H. J., Caldwell, H. K., Macbeth, A. H., Tolu, S. G., & Young, W. S., 3rd (2008). A
conditional knockout mouse line of the oxytocin receptor. Endocrinology, 149(7), 3256–
3263. https://doi.org/10.1210/en.2007-1710
15. Becker-Krail, Darius D et al. “The Ventral Tegmental Area and Nucleus Accumbens as
Circadian Oscillators: Implications for Drug Abuse and Substance Use Disorders.”
Frontiers in physiology vol. 13 886704. 27 Apr. 2022, doi:10.3389/fphys.2022.886704
16. Silberman, Yuval et al. “A corticotropin releasing factor pathway for ethanol regulation
of the ventral tegmental area in the bed nucleus of the stria terminalis.” The Journal of
neuroscience : the official journal of the Society for Neuroscience vol. 33,3 (2013): 950-
60. doi:10.1523/JNEUROSCI.2949-12.2013
17. Vranjkovic, Oliver et al. “Enhanced CRFR1-Dependent Regulation of a Ventral
Tegmental Area to Prelimbic Cortex Projection Establishes Susceptibility to StressInduced Cocaine Seeking.” The Journal of neuroscience : the official journal of the
Society for Neuroscience vol. 38,50 (2018): 10657-10671.
doi:10.1523/JNEUROSCI.2080-18.2018
18. Lebow, M., Chen, A. Overshadowed by the amygdala: the bed nucleus of the stria
terminalis emerges as key to psychiatric disorders. Mol Psychiatry 21, 450–463 (2016).
https://doi.org/10.1038/mp.2016.1
19. Lebow, Maya et al. “Susceptibility to PTSD-like behavior is mediated by corticotropinreleasing factor receptor type 2 levels in the bed nucleus of the stria terminalis.” The
Journal of neuroscience : the official journal of the Society for Neuroscience vol. 32,20
(2012): 6906-16. doi:10.1523/JNEUROSCI.4012-11.2012
20. Ren, Bo et al. “Methionine restriction alleviates age-associated cognitive decline via
fibroblast growth factor 21.” Redox biology vol. 41 (2021): 101940.
doi:10.1016/j.redox.2021.101940
21. Nam, Min-Ho et al. “Excessive Astrocytic GABA Causes Cortical Hypometabolism and
Impedes Functional Recovery after Subcortical Stroke.” Cell reports vol. 32,1 (2020):
107861. doi:10.1016/j.celrep.2020.107861
22. Kilb, Werner, and Sergei Kirischuk. “GABA Release from Astrocytes in Health and
Disease.” International journal of molecular sciences vol. 23,24 15859. 13 Dec. 2022,
doi:10.3390/ijms232415859
23. Liu, Jianhui et al. “Astrocytes: GABAceptive and GABAergic Cells in the Brain.”
Frontiers in cellular neuroscience vol. 16 892497. 10 Jun. 2022,
doi:10.3389/fncel.2022.892497
24. Verkhratsky A, Parpura V, Vardjan N, Zorec R. Physiology of Astroglia. Advances in
Experimental Medicine and Biology. 2019 ;1175:45-91. DOI: 10.1007/978-981-13-9913-
8_3. PMID: 31583584; PMCID: PMC7265998.
25. Chrysafides SM, Bordes SJ, Sharma S. Physiology, Resting Potential. [Updated 2023 Apr
10]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-.
Available from: https://www.ncbi.nlm.nih.gov/books/NBK538338/
26. Verma, M., Lizama, B.N. & Chu, C.T. Excitotoxicity, calcium and mitochondria: a triad
in synaptic neurodegeneration. Transl Neurodegener 11, 3 (2022).
https://doi.org/10.1186/s40035-021-00278-7
27. Kass, G E, and S Orrenius. “Calcium signaling and cytotoxicity.” Environmental health
perspectives vol. 107 Suppl 1,Suppl 1 (1999): 25-35. doi:10.1289/ehp.99107s125



43
28. de Ceglia, Roberta et al. “Specialized astrocytes mediate glutamatergic gliotransmission
in the CNS.” Nature vol. 622,7981 (2023): 120-129. doi:10.1038/s41586-023-06502-w
29. Scofield, Michael D et al. “Gq-DREADD Selectively Initiates Glial Glutamate Release
and Inhibits Cue-induced Cocaine Seeking.” Biological psychiatry vol. 78,7 (2015): 441-
51. doi:10.1016/j.biopsych.2015.02.016
30. Featherstone, David E. “Intercellular glutamate signaling in the nervous system and
beyond.” ACS chemical neuroscience vol. 1,1 (2010): 4-12. doi:10.1021/cn900006n
31. Kim, Haram R, and Marco Martina. “Bidirectional Regulation of GABAA Reversal
Potential in the Adult Brain: Physiological and Pathological Implications.” Life (Basel,
Switzerland) vol. 14,1 143. 19 Jan. 2024, doi:10.3390/life14010143
32. Zhou, Yuan et al. “Soluble β-amyloid impaired the GABA inhibition by mediating KCC2
in early APP/PS1 mice.” Bioscience trends vol. 15,5 (2021): 330-340.
doi:10.5582/bst.2021.01245
33. Ren, Qing-Guo; Gong, Wei-Gang; Wang, Yan-Juan; Zhou, Qi-Da; Zhang, Zhi-Jun
(2015). Citalopram Attenuates Tau Hyperphosphorylation and Spatial Memory Deficit
Induced by Social Isolation Rearing in Middle-Aged Rats. Journal of Molecular
Neuroscience, 56(1), 145–153. doi:10.1007/s12031-014-0475-4
34. Ben-Azu, Benneth et al. “Alcohol-exacerbates post-traumatic stress psychiatric behavior
and its neuropathological sequalae in experimental mice: preventive effects of morin.”
Alcohol (Fayetteville, N.Y.), S0741-8329(24)00111-3. 31 Jul. 2024,
doi:10.1016/j.alcohol.2024.07.009
35. Gong, Yu-Shi et al. “Comparative effects of EtOH consumption and thiamine deficiency
on cognitive impairment, oxidative damage, and β-amyloid peptide overproduction in the
brain.” Free radical biology & medicine vol. 108 (2017): 163-173.
doi:10.1016/j.freeradbiomed.2017.03.019
36. Ben-Azu, Benneth et al. “Alcohol exacerbates psychosocial stress-induced
neuropsychiatric symptoms: Attenuation by geraniol.” Neurochemistry international vol.
177 (2024): 105748. doi:10.1016/j.neuint.2024.105748
37. Hoffman, Jessica L et al. “Alcohol drinking exacerbates neural and behavioral pathology
in the 3xTg-AD mouse model of Alzheimer's disease.” International review of
neurobiology vol. 148 (2019): 169-230. doi:10.1016/bs.irn.2019.10.017
38. van der Vaart, Andrew et al. “Glycogen synthase kinase 3 beta regulates ethanol
consumption and is a risk factor for alcohol dependence.” Neuropsychopharmacology :
official publication of the American College of Neuropsychopharmacology vol. 43,13
(2018): 2521-2531. doi:10.1038/s41386-018-0202-x
39. Wang, Jian-Zhi et al. “Kinases and phosphatases and tau sites involved in Alzheimer
neurofibrillary degeneration.” The European journal of neuroscience vol. 25,1 (2007):
59-68. doi:10.1111/j.1460-9568.2006.05226.x
40. Ji, Zhe et al. “Binge Alcohol Exposure Causes Neurobehavioral Deficits and GSK3β
Activation in the Hippocampus of Adolescent Rats.” Scientific reports vol. 8,1 3088. 15
Feb. 2018, doi:10.1038/s41598-018-21341-w
41. Neasta, Jérémie et al. “AKT signaling pathway in the nucleus accumbens mediates
excessive alcohol drinking behaviors.” Biological psychiatry vol. 70,6 (2011): 575-82.
doi:10.1016/j.biopsych.2011.03.019



44
42. Wu, Dianqing, and Weijun Pan. “GSK3: a multifaceted kinase in Wnt signaling.” Trends
in biochemical sciences vol. 35,3 (2010): 161-8. doi:10.1016/j.tibs.2009.10.002
43. Qin, Kevin et al. “Canonical and noncanonical Wnt signaling: Multilayered mediators,
signaling mechanisms and major signaling crosstalk.” Genes & diseases vol. 11,1 103-
134. 24 Mar. 2023, doi:10.1016/j.gendis.2023.01.030
44. Pereira GC, Piton E, Bornholdt J, Dos Santos BM, de Almeida AS, Dalenogare DP,
Fialho MFP, Becker G, da Silva Brum E, Sampaio TB, Oliveira SM, Oliveira MS,
Trevisan G, Bochi GV. TRPA1 participation in behavioral impairment induced by
chronic corticosterone administration. Psychopharmacology (Berl). 2023 Jan;240(1):157-
169. doi: 10.1007/s00213-022-06290-7. Epub 2022 Dec 15. PMID: 36520197.
45. Matrisciano, Francesco et al. “Induction of the Wnt antagonist Dickkopf-1 is involved in
stress-induced hippocampal damage.” PloS one vol. 6,1 e16447. 27 Jan. 2011,
doi:10.1371/journal.pone.0016447
46. Arnsten, Amy F T et al. “Hypothesis: Tau pathology is an initiating factor in sporadic
Alzheimer's disease.” Alzheimer's & dementia : the journal of the Alzheimer's
Association vol. 17,1 (2021): 115-124. doi:10.1002/alz.12192
47. Jin, Nana et al. “Truncation and activation of GSK-3β by calpain I: a molecular
mechanism links to tau hyperphosphorylation in Alzheimer's disease.” Scientific reports
vol. 5 8187. 2 Feb. 2015, doi:10.1038/srep08187
48. Tyagarajan, Shiva K et al. “Regulation of GABAergic synapse formation and plasticity
by GSK3beta-dependent phosphorylation of gephyrin.” Proceedings of the National
Academy of Sciences of the United States of America vol. 108,1 (2011): 379-84.
doi:10.1073/pnas.1011824108
49. Hayes, John D.; Chowdhry, Sudhir; Dinkova-Kostova, Albena T.; Sutherland, Calum
(2015). Dual regulation of transcription factor Nrf2 by Keap1 and by the combined
actions of β-TrCP and GSK-3. Biochemical Society Transactions, 43(4), 611–620.
doi:10.1042/bst20150011
50. Rada, Patricia et al. “SCF/{beta}-TrCP promotes glycogen synthase kinase 3-dependent
degradation of the Nrf2 transcription factor in a Keap1-independent manner.” Molecular
and cellular biology vol. 31,6 (2011): 1121-33. doi:10.1128/MCB.01204-10
51. Vomund, Sandra et al. “Nrf2, the Master Regulator of Anti-Oxidative Responses.”
International journal of molecular sciences vol. 18,12 2772. 20 Dec. 2017,
doi:10.3390/ijms18122772
52. Baird, L., & Yamamoto, M. (2020). The Molecular Mechanisms Regulating the KEAP1-
NRF2 Pathway. Molecular and cellular biology, 40(13), e00099-20.
https://doi.org/10.1128/MCB.00099-20
53. Battaglia, Sereina et al. “Activity-Dependent Inhibitory Synapse Scaling Is Determined
by Gephyrin Phosphorylation and Subsequent Regulation of GABAA Receptor
Diffusion.” eNeuro vol. 5,1 ENEURO.0203-17.2017. 18 Jan. 2018,
doi:10.1523/ENEURO.0203-17.2017
54. Wu, Zheng et al. “Tonic inhibition in dentate gyrus impairs long-term potentiation and
memory in an Alzheimer's [corrected] disease model.” Nature communications vol. 5
4159. 13 Jun. 2014, doi:10.1038/ncomms5159
55. Jo, Seonmi et al. “GABA from reactive astrocytes impairs memory in mouse models of
Alzheimer's disease.” Nature medicine vol. 20,8 (2014): 886-96. doi:10.1038/nm.3639



45
56. Ju, Yeon Ha et al. “Astrocytic urea cycle detoxifies Aβ-derived ammonia while impairing
memory in Alzheimer's disease.” Cell metabolism vol. 34,8 (2022): 1104-1120.e8.
doi:10.1016/j.cmet.2022.05.011
57. Amiri, Shayan et al. “Co-occurrence of anxiety and depressive-like behaviors following
adolescent social isolation in male mice; possible role of nitrergic system.” Physiology &
behavior vol. 145 (2015): 38-44. doi:10.1016/j.physbeh.2015.03.032
58. Song, Lingling et al. “Antidepressant effect of catalpol on corticosterone-induced
depressive-like behavior involves the inhibition of HPA axis hyperactivity, central
inflammation and oxidative damage probably via dual regulation of NF-κB and Nrf2.”
Brain research bulletin vol. 177 (2021): 81-91. doi:10.1016/j.brainresbull.2021.09.002
59. Jo D, Kim BC, Cho KA, Song J. The Cerebral Effect of Ammonia in Brain Aging:
Blood-Brain Barrier Breakdown, Mitochondrial Dysfunction, and Neuroinflammation. J
Clin Med. 2021 Jun 24;10(13):2773. doi: 10.3390/jcm10132773. PMID: 34202669;
PMCID: PMC8268635.
60. Komatsu, Ayaka et al. “Ammonia induces amyloidogenesis in astrocytes by promoting
amyloid precursor protein translocation into the endoplasmic reticulum.” The Journal of
biological chemistry vol. 298,5 (2022): 101933. doi:10.1016/j.jbc.2022.101933
61. Mezler M, Barghorn S, Schoemaker H, Gross G, Nimmrich V. A β-amyloid oligomer
directly modulates P/Q-type calcium currents in Xenopus oocytes. Br J Pharmacol. 2012
Mar;165(5):1572-83. doi: 10.1111/j.1476-5381.2011.01646.x. PMID: 21883149;
PMCID: PMC3372738.
62. Jarosz-Griffiths HH, Noble E, Rushworth JV, Hooper NM. Amyloid-β Receptors: The
Good, the Bad, and the Prion Protein. J Biol Chem. 2016 Feb 12;291(7):3174-83. doi:
10.1074/jbc.R115.702704. Epub 2015 Dec 30. PMID: 26719327; PMCID:
PMC4751366.
63. Andersen, Jens V et al. “Hippocampal disruptions of synaptic and astrocyte metabolism
are primary events of early amyloid pathology in the 5xFAD mouse model of
Alzheimer's disease.” Cell death & disease vol. 12,11 954. 16 Oct. 2021,
doi:10.1038/s41419-021-04237-y
64. Ellinger, James J et al. “Role of aminotransferases in glutamate metabolism of human
erythrocytes.” Journal of biomolecular NMR vol. 49,3-4 (2011): 221-9.
doi:10.1007/s10858-011-9481-9
65. Ren, Zhao-Xiang et al. “Dihydromyricetin protects neurons in an MPTP-induced model
of Parkinson's disease by suppressing glycogen synthase kinase-3 beta activity.” Acta
pharmacologica Sinica vol. 37,10 (2016): 1315-1324. doi:10.1038/aps.2016.42
66. Haga, Hidaka et al. “Enhancement of ATP production ameliorates motor and cognitive
impairments in a mouse model of MPTP-induced Parkinson's disease.” Neurochemistry
international vol. 129 (2019): 104492. doi:10.1016/j.neuint.2019.104492
67. Watanabe, Saki et al. “Social isolation induces succinate dehydrogenase dysfunction in
anxious mice.” Neurochemistry international vol. 161 (2022): 105434.
doi:10.1016/j.neuint.2022.105434
68. Augusto, Elisabete et al. “Increased ATP release and CD73-mediated adenosine A2A
receptor activation mediate convulsion-associated neuronal damage and hippocampal
dysfunction.” Neurobiology of disease vol. 157 (2021): 105441.
doi:10.1016/j.nbd.2021.105441



46
69. Gonçalves, Francisco Q et al. “Synaptic and memory dysfunction in a β-amyloid model
of early Alzheimer's disease depends on increased formation of ATP-derived
extracellular adenosine.” Neurobiology of disease vol. 132 (2019): 104570.
doi:10.1016/j.nbd.2019.104570
70. Woods, Lucas T et al. “Purinergic receptors as potential therapeutic targets in
Alzheimer's disease.” Neuropharmacology vol. 104 (2016): 169-79.
doi:10.1016/j.neuropharm.2015.10.031
71. Moro, Nobuhiro et al. “Massive efflux of adenosine triphosphate into the extracellular
space immediately after experimental traumatic brain injury.” Experimental and
therapeutic medicine vol. 21,6 (2021): 575. doi:10.3892/etm.2021.10007
72. Frenguelli, Bruno G et al. “Temporal and mechanistic dissociation of ATP and adenosine
release during ischaemia in the mammalian hippocampus.” Journal of neurochemistry
vol. 101,5 (2007): 1400-13. doi:10.1111/j.1471-4159.2006.04425.x
73. Cho, Woo-Hyun et al. “Hippocampal astrocytes modulate anxiety-like behavior.” Nature
communications vol. 13,1 6536. 7 Nov. 2022, doi:10.1038/s41467-022-34201-z
74. Lalo, Ulyana et al. “Exocytosis of ATP from astrocytes modulates phasic and tonic
inhibition in the neocortex.” PLoS biology vol. 12,1 (2014): e1001747.
doi:10.1371/journal.pbio.1001747
75. Huang, Qiang et al. “P2X7 Receptor: an Emerging Target in Alzheimer's Disease.”
Molecular neurobiology vol. 61,5 (2024): 2866-2880. doi:10.1007/s12035-023-03699-9
76. ibič, Lučka, and Leanne Stokes. “Revisiting the Idea That Amyloid-β Peptide Acts as an
Agonist for P2X7.” Frontiers in molecular neuroscience vol. 13 166. 17 Sep. 2020,
doi:10.3389/fnmol.2020.00166
77. Diaz-Hernandez, Juan Ignacio et al. “In vivo P2X7 inhibition reduces amyloid plaques in
Alzheimer's disease through GSK3β and secretases.” Neurobiology of aging vol. 33,8
(2012): 1816-28. doi:10.1016/j.neurobiolaging.2011.09.040
78. Delarasse, Cécile et al. “The purinergic receptor P2X7 triggers alpha-secretase-dependent
processing of the amyloid precursor protein.” The Journal of biological chemistry vol.
286,4 (2011): 2596-606. doi:10.1074/jbc.M110.200618
79. Ly, Philip T T et al. “Inhibition of GSK3β-mediated BACE1 expression reduces
Alzheimer-associated phenotypes.” The Journal of clinical investigation vol. 123,1
(2013): 224-35. doi:10.1172/JCI64516
80. Sáez-Orellana, F et al. “ATP leakage induces P2XR activation and contributes to acute
synaptic excitotoxicity induced by soluble oligomers of β-amyloid peptide in
hippocampal neurons.” Neuropharmacology vol. 100 (2016): 116-23.
doi:10.1016/j.neuropharm.2015.04.005
81. Gessi, Stefania et al. “A2A Adenosine Receptor as a Potential Biomarker and a Possible
Therapeutic Target in Alzheimer's Disease.” Cells vol. 10,9 2344. 7 Sep. 2021,
doi:10.3390/cells10092344
82. Temido-Ferreira, Mariana et al. “Age-related shift in LTD is dependent on neuronal
adenosine A2A receptors interplay with mGluR5 and NMDA receptors.” Molecular
psychiatry vol. 25,8 (2020): 1876-1900. doi:10.1038/s41380-018-0110-9



47
83. Merighi, Stefania et al. “Upregulation of Cortical A2A Adenosine Receptors Is Reflected
in Platelets of Patients with Alzheimer's Disease.” Journal of Alzheimer's disease : JAD
vol. 80,3 (2021): 1105-1117. doi:10.3233/JAD-201437
84. Gomez-Murcia, Victoria et al. “Neuronal A2A receptor exacerbates synapse loss and
memory deficits in APP/PS1 mice.” Brain : a journal of neurology vol. 147,8 (2024):
2691-2705. doi:10.1093/brain/awae113
85. Ji, Qi et al. “Blockade of adenosine A2A receptors reverses early spatial memory defects
in the APP/PS1 mouse model of Alzheimer's disease by promoting synaptic plasticity of
adult-born granule cells.” Alzheimer's research & therapy vol. 15,1 187. 30 Oct. 2023,
doi:10.1186/s13195-023-01337-z
86. Zhao, Z-A et al. “Adenosine A2A receptor inactivation alleviates early-onset cognitive
dysfunction after traumatic brain injury involving an inhibition of tau
hyperphosphorylation.” Translational psychiatry vol. 7,5 e1123. 9 May. 2017,
doi:10.1038/tp.2017.98
87. Canas, Paula M et al. “Neuronal Adenosine A2A Receptors Are Critical Mediators of
Neurodegeneration Triggered by Convulsions.” eNeuro vol. 5,6 ENEURO.0385-18.2018.
26 Dec. 2018, doi:10.1523/ENEURO.0385-18.2018
88. Nasrallah, Kaoutsar et al. “Retrograde adenosine/A2A receptor signaling facilitates
excitatory synaptic transmission and seizures.” Cell reports vol. 43,7 (2024): 114382.
doi:10.1016/j.celrep.2024.114382
89. Matos, Marco et al. “Deletion of adenosine A2A receptors from astrocytes disrupts
glutamate homeostasis leading to psychomotor and cognitive impairment: relevance to
schizophrenia.” Biological psychiatry vol. 78,11 (2015): 763-74.
doi:10.1016/j.biopsych.2015.02.026
90. Mouro, Francisco M et al. “Adenosine A2A receptors facilitate synaptic NMDA currents
in CA1 pyramidal neurons.” British journal of pharmacology vol. 175,23 (2018): 4386-
4397. doi:10.1111/bph.14497
91. Numakawa, Tadahiro, and Ryutaro Kajihara. “An Interaction between Brain-Derived
Neurotrophic Factor and Stress-Related Glucocorticoids in the Pathophysiology of
Alzheimer's Disease.” International journal of molecular sciences vol. 25,3 1596. 27 Jan.
2024, doi:10.3390/ijms25031596
92. Jerónimo-Santos, André et al. “Dysregulation of TrkB Receptors and BDNF Function by
Amyloid-β Peptide is Mediated by Calpain.” Cerebral cortex (New York, N.Y. : 1991)
vol. 25,9 (2015): 3107-21. doi:10.1093/cercor/bhu105
93. Jerónimo-Santos, André et al. “Brain-derived neurotrophic factor mediates
neuroprotection against Aβ-induced toxicity through a mechanism independent on
adenosine 2A receptor activation.” Growth factors (Chur, Switzerland) vol. 33,4 (2015):
298-308. doi:10.3109/08977194.2015.1080696
94. Gomes, João R et al. “Excitotoxicity downregulates TrkB.FL signaling and upregulates
the neuroprotective truncated TrkB receptors in cultured hippocampal and striatal
neurons.” The Journal of neuroscience : the official journal of the Society for
Neuroscience vol. 32,13 (2012): 4610-22. doi:10.1523/JNEUROSCI.0374-12.2012
95. Dromard, Yann et al. “Loss of glucocorticoid receptor phosphorylation contributes to
cognitive and neurocentric damages of the amyloid-β pathway.” Acta neuropathologica
communications vol. 10,1 91. 22 Jun. 2022, doi:10.1186/s40478-022-01396-7



48
96. Barfield, Elizabeth T et al. “Regulation of actions and habits by ventral hippocampal trkB
and adolescent corticosteroid exposure.” PLoS biology vol. 15,11 e2003000. 29 Nov.
2017, doi:10.1371/journal.pbio.2003000
97. Arango-Lievano, Margarita et al. “Persistence of learning-induced synapses depends on
neurotrophic priming of glucocorticoid receptors.” Proceedings of the National Academy
of Sciences of the United States of America vol. 116,26 (2019): 13097-13106.
doi:10.1073/pnas.1903203116
98. Batalha, Vânia L et al. “The caffeine-binding adenosine A2A receptor induces age-like
HPA-axis dysfunction by targeting glucocorticoid receptor function.” Scientific reports
vol. 6 31493. 11 Aug. 2016, doi:10.1038/srep31493
99. Madeira, Daniela et al. “Adenosine A2A receptors blockade attenuates dexamethasoneinduced alterations in cultured astrocytes.” Purinergic signalling vol. 18,2 (2022): 199-
204. doi:10.1007/s11302-022-09864-4
100. Xia, Qingqing et al. “Excessive corticosterone induces excitotoxicity of
hippocampal neurons and sensitivity of potassium channels via insulin-signaling
pathway.” Metabolic brain disease vol. 34,1 (2019): 119-128. doi:10.1007/s11011-018-
0326-z
101. Tsai, Tsung-Chih et al. “A dorsal CA2 to ventral CA1 circuit contributes to
oxytocinergic modulation of long-term social recognition memory.” Journal of
biomedical science vol. 29,1 50. 10 Jul. 2022, doi:10.1186/s12929-022-00834-x
102. Lussier, A.L.; Romay-Tallón, R.; Caruncho, H.J.; Kalynchuk, L.E. (2013).
Altered GABAergic and glutamatergic activity within the rat hippocampus and amygdala
in rats subjected to repeated corticosterone administration but not restraint stress.
Neuroscience, 231(), 38–48. doi:10.1016/j.neuroscience.2012.11.037
103. Patel, Rucha et al. “Glucocorticoids regulate the metabolic hormone FGF21 in a
feed-forward loop.” Molecular endocrinology (Baltimore, Md.) vol. 29,2 (2015): 213-23.
doi:10.1210/me.2014-1259
104. Matsui, S., Sasaki, T., Kohno, D. et al. Neuronal SIRT1 regulates macronutrientbased diet selection through FGF21 and oxytocin signalling in mice. Nat Commun 9,
4604 (2018). https://doi.org/10.1038/s41467-018-07033-z
105. Dong, Hong-Wei, and Larry W Swanson. “Projections from bed nuclei of the stria
terminalis, dorsomedial nucleus: implications for cerebral hemisphere integration of
neuroendocrine, autonomic, and drinking responses.” The Journal of comparative
neurology vol. 494,1 (2006): 75-107. doi:10.1002/cne.20790
106. Hong-Wei Dong; Larry W. Swanson (2004). Organization of axonal projections
from the anterolateral area of the bed nuclei of the stria terminalis. , 468(2), 277–298.
doi:10.1002/cne.10949
107. Jiang, Zhiying et al. “CRF signaling between neurons in the paraventricular
nucleus of the hypothalamus (PVN) coordinates stress responses.” Neurobiology of stress
vol. 11 100192. 10 Aug. 2019, doi:10.1016/j.ynstr.2019.100192
108. Forray, María Inés, and Katia Gysling. “Role of noradrenergic projections to the
bed nucleus of the stria terminalis in the regulation of the hypothalamic-pituitary-adrenal
axis.” Brain research. Brain research reviews vol. 47,1-3 (2004): 145-60.
doi:10.1016/j.brainresrev.2004.07.011



49
109. Radley, Jason J, and Paul E Sawchenko. “A common substrate for prefrontal and
hippocampal inhibition of the neuroendocrine stress response.” The Journal of
neuroscience : the official journal of the Society for Neuroscience vol. 31,26 (2011):
9683-95. doi:10.1523/JNEUROSCI.6040-10.2011
110. Hammack, Sayamwong E et al. “The response of neurons in the bed nucleus of
the stria terminalis to serotonin: implications for anxiety.” Progress in neuropsychopharmacology & biological psychiatry vol. 33,8 (2009): 1309-20.
doi:10.1016/j.pnpbp.2009.05.013
111. Hong-Wei Dong; Larry W. Swanson (2004). Projections from bed nuclei of the
stria terminalis, posterior division: Implications for cerebral hemisphere regulation of
defensive and reproductive behaviors. , 471(4), 396–433. doi:10.1002/cne.20002
112. Bota, Mihail et al. “Neuroinformatics analysis of molecular expression patterns
and neuron populations in gray matter regions: the rat BST as a rich exemplar.” Brain
research vol. 1450 (2012): 174-93. doi:10.1016/j.brainres.2012.02.034
113. Van Pett, K et al. “Distribution of mRNAs encoding CRF receptors in brain and
pituitary of rat and mouse.” The Journal of comparative neurology vol. 428,2 (2000):
191-212. doi:10.1002/1096-9861(20001211)428:2<191::aid-cne1>3.0.co;2-u
114. Land, Benjamin B et al. “The dysphoric component of stress is encoded by
activation of the dynorphin kappa-opioid system.” The Journal of neuroscience : the
official journal of the Society for Neuroscience vol. 28,2 (2008): 407-14.
doi:10.1523/JNEUROSCI.4458-07.2008
115. Haun, Harold L et al. “Dynorphin/Kappa Opioid Receptor Activity Within the
Extended Amygdala Contributes to Stress-Enhanced Alcohol Drinking in Mice.”
Biological psychiatry vol. 91,12 (2022): 1019-1028. doi:10.1016/j.biopsych.2022.01.002
116. Erikson, Chloe M et al. “Maladaptive behavioral regulation in alcohol
dependence: Role of kappa-opioid receptors in the bed nucleus of the stria terminalis.”
Neuropharmacology vol. 140 (2018): 162-173. doi:10.1016/j.neuropharm.2018.07.034
117. Ingallinesi, M et al. “CRF2 receptor-deficiency eliminates opiate withdrawal
distress without impairing stress coping.” Molecular psychiatry vol. 17,12 (2012): 1283-
94. doi:10.1038/mp.2011.119
118. Elharrar, Einat; Warhaftig, Gal; Issler, Orna; Sztainberg, Yehezkel; Dikshtein,
Yahav; Zahut, Roy; Redlus, Lior; Chen, Alon; Yadid, Gal (2013). Overexpression of
Corticotropin-Releasing Factor Receptor Type 2 in the Bed Nucleus of Stria Terminalis
Improves Posttraumatic Stress Disorder-like Symptoms in a Model of Incubation of Fear.
Biological Psychiatry, 74(11), 827–836. doi:10.1016/j.biopsych.2013.05.03
119. Henckens, M., Printz, Y., Shamgar, U. et al. CRF receptor type 2 neurons in the
posterior bed nucleus of the stria terminalis critically contribute to stress recovery. Mol
Psychiatry 22, 1691–1700 (2017). https://doi.org/10.1038/mp.2016.133
120. Hong-Wei Dong; Gorica D. Petrovich; Alan G. Watts; Larry W. Swanson (2001).
Basic organization of projections from the oval and fusiform nuclei of the bed nuclei of
the stria terminalis in adult rat brain. , 436(4), 430–455. doi:10.1002/cne.1079
121. Park, Jinwoo et al. “Opposing catecholamine changes in the bed nucleus of the
stria terminalis during intracranial self-stimulation and its extinction.” Biological
psychiatry vol. 74,1 (2013): 69-76. doi:10.1016/j.biopsych.2012.11.008



50
122. Kash, Thomas L et al. “Dopamine enhances fast excitatory synaptic transmission
in the extended amygdala by a CRF-R1-dependent process.” The Journal of neuroscience
: the official journal of the Society for Neuroscience vol. 28,51 (2008): 13856-65.
doi:10.1523/JNEUROSCI.4715-08.2008
123. Jaferi, A, and V M Pickel. “Mu-opioid and corticotropin-releasing-factor
receptors show largely postsynaptic co-expression, and separate presynaptic distributions,
in the mouse central amygdala and bed nucleus of the stria terminalis.” Neuroscience vol.
159,2 (2009): 526-39. doi:10.1016/j.neuroscience.2008.12.061
124. Valentino, R.J., Volkow, N. Untangling the complexity of opioid receptor
function. Neuropsychopharmacol 43, 2514–2520 (2018). https://doi.org/10.1038/s41386-
018-0225-3
125. Li, Chia et al. “Presynaptic inhibition of gamma-aminobutyric acid release in the
bed nucleus of the stria terminalis by kappa opioid receptor signaling.” Biological
psychiatry vol. 71,8 (2012): 725-32. doi:10.1016/j.biopsych.2011.11.015
126. Borrego, Marissa B et al. “Central nucleus of the amygdala projections onto the
nucleus accumbens core regulate binge-like alcohol drinking in a CRF-dependent
manner.” Neuropharmacology vol. 203 (2022): 108874.
doi:10.1016/j.neuropharm.2021.108874
127. Shin, Jung-Won et al. “Inputs to the ventrolateral bed nucleus of the stria
terminalis.” The Journal of comparative neurology vol. 511,5 (2008): 628-57.
doi:10.1002/cne.21870
128. Bienkowski, Michael S, and Linda Rinaman. “Common and distinct neural inputs
to the medial central nucleus of the amygdala and anterior ventrolateral bed nucleus of
stria terminalis in rats.” Brain structure & function vol. 218,1 (2013): 187-208.
doi:10.1007/s00429-012-0393-6
129. Zhu, Zhaoming et al. “GABAergic neurons in nucleus accumbens are correlated
to resilience and vulnerability to chronic stress for major depression.” Oncotarget vol.
8,22 (2017): 35933-35945. doi:10.18632/oncotarget.16411
130. Williams, Courtney L et al. “CRF-R2 and the heterosynaptic regulation of VTA
glutamate during reinstatement of cocaine seeking.” The Journal of neuroscience : the
official journal of the Society for Neuroscience vol. 34,31 (2014): 10402-14.
doi:10.1523/JNEUROSCI.0911-13.2014
131. Gould, Timothy et al. “GABA(B) receptor-mediated activation of astrocytes by
gamma-hydroxybutyric acid.” Philosophical transactions of the Royal Society of London.
Series B, Biological sciences vol. 369,1654 (2014): 20130607.
doi:10.1098/rstb.2013.0607
132. Sharpe, Amanda L et al. “Repeated cocaine or methamphetamine treatment alters
astrocytic CRF2 and GLAST expression in the ventral midbrain.” Addiction biology vol.
27,2 (2022): e13120. doi:10.1111/adb.13120
133. Novoa, Javier et al. “Social isolation of adolescent male rats increases anxiety and
K+ -induced dopamine release in the nucleus accumbens: Role of CRF-R1.” The
European journal of neuroscience vol. 54,3 (2021): 4888-4905. doi:10.1111/ejn.15345
134. Pati, Dipanwita et al. “Chronic intermittent ethanol exposure dysregulates a
GABAergic microcircuit in the bed nucleus of the stria terminalis.” Neuropharmacology
vol. 168 (2020): 107759. doi:10.1016/j.neuropharm.2019.107759



51
135. Takach, Oliver et al. “Modulation of insulin signaling rescues BDNF transport
defects independent of tau in amyloid-β oligomer-treated hippocampal neurons.”
Neurobiology of aging vol. 36,3 (2015): 1378-82.
doi:10.1016/j.neurobiolaging.2014.11.018
136. Gupta, Smriti et al. “siRNA Mediated GSK3β Knockdown Targets Insulin
Signaling Pathway and Rescues Alzheimer's Disease Pathology: Evidence from In Vitro
and In Vivo Studies.” ACS applied materials & interfaces vol. 14,1 (2022): 69-93.
doi:10.1021/acsami.1c15305
137. Ma, Qiu-Lan et al. “Beta-amyloid oligomers induce phosphorylation of tau and
inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling:
suppression by omega-3 fatty acids and curcumin.” The Journal of neuroscience : the
official journal of the Society for Neuroscience vol. 29,28 (2009): 9078-89.
doi:10.1523/JNEUROSCI.1071-09.2009
138. Liberman, Ziva, and Hagit Eldar-Finkelman. “Serine 332 phosphorylation of
insulin receptor substrate-1 by glycogen synthase kinase-3 attenuates insulin signaling.”
The Journal of biological chemistry vol. 280,6 (2005): 4422-8.
doi:10.1074/jbc.M410610200
139. Huang, Rong; Tian, Sai; Zhang, Haoqiang; Zhu, Wenwen; Wang, Shaohua
(2020). Chronic hyperglycemia induces tau hyperphosphorylation by downregulating
OGT-involved O-GlcNAcylation in vivo and in vitro. Brain Research Bulletin, 156(), 76–
85. doi:10.1016/j.brainresbull.2020.01.006
140. Lv, Hui et al. “Intranasal insulin administration may be highly effective in
improving cognitive function in mice with cognitive dysfunction by reversing brain
insulin resistance.” Cognitive neurodynamics vol. 14,3 (2020): 323-338.
doi:10.1007/s11571-020-09571-z
141. Marciniak, Elodie et al. “Tau deletion promotes brain insulin resistance.” The
Journal of experimental medicine vol. 214,8 (2017): 2257-2269.
doi:10.1084/jem.20161731
142. Jiang, J.; Li, K.; Xiao, Y.; Zhong, A.; Tang, J.; Duan, Y.; Li, Z.
Limosilactobacillus reuteri Regulating Intestinal Function: A Review. Fermentation
2023, 9, 19. https://doi.org/10.3390/fermentation9010019
143. Jiao, Y.; Yang, H.; Shigwedha, N.; Zhang, S.; Liu, F.; Zhang, L. Probiotic Effects
and Metabolic Products of Enterococcus faecalis LD33 with Respiration Capacity. Foods
2022, 11, 606. https://doi.org/10.3390/foods11040606
144. Engen, Phillip A et al. “The Gastrointestinal Microbiome: Alcohol Effects on the
Composition of Intestinal Microbiota.” Alcohol research : current reviews vol. 37,2
(2015): 223-36.
145. Pohl, Keith et al. “Alcohol's Impact on the Gut and Liver.” Nutrients vol. 13,9
3170. 11 Sep. 2021, doi:10.3390/nu13093170
146. Flippo, Kyle H et al. “FGF21 suppresses alcohol consumption through an
amygdalo-striatal circuit.” Cell metabolism vol. 34,2 (2022): 317-328.e6.
doi:10.1016/j.cmet.2021.12.024
147. Zhang, Xi et al. “Fibroblast growth factor 21 alleviates diabetes-induced cognitive
decline.” Cerebral cortex (New York, N.Y. : 1991) vol. 34,2 (2024): bhad502.
doi:10.1093/cercor/bhad502 
Abstract (if available)
Abstract Neurodegenerative diseases such as Alzheimer’s and Parkinson’s are complex disorders that are poorly understood. They incorporate a broad range of dysfunctions, including the formation of Tau and Amyloid aggregates in the hippocampus. This results in changes in behavior and impairment to cognitive function. However, given the complexity of these disorders, it is difficult to characterize the active mechanisms and how they contribute to disease progression. This thesis aims to provide a new model that can better describe the cellular and neurological changes associated with neurodegenerative disorders. This includes an investigation into the molecular mechanisms that contribute to impaired neuronal function. In particular, this thesis proposes a neural pathway that connects multiple regions of the brain in order to characterize changes in behavior. This thesis integrates current research into neurodegeneration in order to synthesize a model that can guide future research. 
Linked assets
University of Southern California Dissertations and Theses
doctype icon
University of Southern California Dissertations and Theses 
Action button
Conceptually similar
Integration of KNIME and molecular docking for evaluation of tau fibril inhibitors
PDF
Integration of KNIME and molecular docking for evaluation of tau fibril inhibitors 
Exploration and expansion of novel therapeutic strategies targeting brain metabolism in neurodegenerative diseases
PDF
Exploration and expansion of novel therapeutic strategies targeting brain metabolism in neurodegenerative diseases 
Inflammatory responses during infection: implications for neurodegeneration
PDF
Inflammatory responses during infection: implications for neurodegeneration 
DHM reduces cognitive impairment induced by social isolation and ethanol consumption
PDF
DHM reduces cognitive impairment induced by social isolation and ethanol consumption 
Sodium butyrate prevents antibiotic-induced increase in ethanol drinking in C57BL/6J mice by modulating neuroinflammatory response
PDF
Sodium butyrate prevents antibiotic-induced increase in ethanol drinking in C57BL/6J mice by modulating neuroinflammatory response 
Dihydromyricetin improves social isolation anxiety induced synaptic loss and astrogliosis in mice hippocampus
PDF
Dihydromyricetin improves social isolation anxiety induced synaptic loss and astrogliosis in mice hippocampus 
Memory abnormalities in Alzheimer's disease and anxiety models
PDF
Memory abnormalities in Alzheimer's disease and anxiety models 
Role of purinergic P2X7 receptors in inflammatory responses in the brain and liver: a study using a mouse model of chronic ethanol and high-fat diet exposure
PDF
Role of purinergic P2X7 receptors in inflammatory responses in the brain and liver: a study using a mouse model of chronic ethanol and high-fat diet exposure 
Dihydromyricetin as a potential therapeutic for Alzheimer's disease-like changes induced by social isolation
PDF
Dihydromyricetin as a potential therapeutic for Alzheimer's disease-like changes induced by social isolation 
Alterations in renin-angiotensin system signaling in SARS-CoV2 infection
PDF
Alterations in renin-angiotensin system signaling in SARS-CoV2 infection 
Investigation into the role of the intestinal microbiome on ethanol consumption behaviors
PDF
Investigation into the role of the intestinal microbiome on ethanol consumption behaviors 
Evaluation of P2X4 receptor modulation as a novel approach for treating Parkinson’s disease
PDF
Evaluation of P2X4 receptor modulation as a novel approach for treating Parkinson’s disease 
Age dependent modulation of synaptic plasticity and insulin mimetic effect of lipoic acid on a 3xTg-AD mouse model of Alzheimer's disease: implications as a therapeutic/nutraceutical agent
PDF
Age dependent modulation of synaptic plasticity and insulin mimetic effect of lipoic acid on a 3xTg-AD mouse model of Alzheimer's disease: implications as a therapeutic/nutraceutical agent 
Investigating sodium butyrate as a potential treatment for alcohol liver disease through the gut-liver axis
PDF
Investigating sodium butyrate as a potential treatment for alcohol liver disease through the gut-liver axis 
Role of translocator protein (18 kDa) in cell function and metabolism
PDF
Role of translocator protein (18 kDa) in cell function and metabolism 
Elucidating structural and functional aspects of biomolecules using classical and quantum mechanics
PDF
Elucidating structural and functional aspects of biomolecules using classical and quantum mechanics 
Neuroinflammation and ApoE4 genotype in at-risk female aging: implications for Alzheimer's disease
PDF
Neuroinflammation and ApoE4 genotype in at-risk female aging: implications for Alzheimer's disease 
Averting dementia: renin-angiotensin system and angiogenic cells in cognitive decline
PDF
Averting dementia: renin-angiotensin system and angiogenic cells in cognitive decline 
The effects of a fasting mimicking diet (FMD) on mouse models of Alzheimer's and Parkinson's disease
PDF
The effects of a fasting mimicking diet (FMD) on mouse models of Alzheimer's and Parkinson's disease 
Insulin sensitivity in cognition, Alzheimer's disease and brain aging
PDF
Insulin sensitivity in cognition, Alzheimer's disease and brain aging 
Action button
Asset Metadata
Creator Talamas, Robin (author) 
Core Title A new model of neurodegeneration: integrating molecular, electrochemical, and neuronal behavior into a novel paradigm 
Contributor Electronically uploaded by the author (provenance) 
School School of Pharmacy 
Degree Master of Science 
Degree Program Pharmaceutical Sciences 
Degree Conferral Date 2024-12 
Publication Date 01/15/2025 
Defense Date 01/14/2025 
Publisher Los Angeles, California (original), University of Southern California (original), University of Southern California. Libraries (digital) 
Tag Addiction,Alzheimer's,bed nucleus of the stria terminalis,hippocampus,neurodegeneration,OAI-PMH Harvest 
Format theses (aat) 
Language English
Advisor Davies, Daryl (committee chair), Asatryan, Liana (committee member), Seidler, Paul (committee member) 
Creator Email talamas@usc.edu 
Unique identifier UC11399FAX8 
Identifier etd-TalamasRob-13761.pdf (filename) 
Legacy Identifier etd-TalamasRob-13761 
Document Type Thesis 
Format theses (aat) 
Rights Talamas, Robin 
Internet Media Type application/pdf 
Type texts
Source 20250115-usctheses-batch-1235 (batch), University of Southern California (contributing entity), University of Southern California Dissertations and Theses (collection) 
Access Conditions The author retains rights to his/her dissertation, thesis or other graduate work according to U.S. copyright law.  Electronic access is being provided by the USC Libraries in agreement with the author, as the original true and official version of the work, but does not grant the reader permission to use the work if the desired use is covered by copyright.  It is the author, as rights holder, who must provide use permission if such use is covered by copyright. 
Repository Name University of Southern California Digital Library
Repository Location USC Digital Library, University of Southern California, University Park Campus MC 2810, 3434 South Grand Avenue, 2nd Floor, Los Angeles, California 90089-2810, USA
Repository Email cisadmin@lib.usc.edu
Tags
Alzheimer's
bed nucleus of the stria terminalis
hippocampus
neurodegeneration