SlideShare a Scribd company logo
1 of 53
Download to read offline
DISORDERS OF REGULATION: TOWARDS A MODEL OF SYSTEMIC BASED
INDIVIDUALIZED TREATMENT FOR AUTOIMMUNE DISEASE,
NEURODEGENRATIVE DISORDERS, AND CANCER
2
DISORDERS OF REGULATION: A SYSTEMS APPROACH
The brain and the body work as a cohesive system. However, most theoretical
understandings of disease processes do not take a systems-based approach to understanding the
way that complex behavior emerges from these physical structures. Applying systems thinking to
the problem diseases could yield novel models for disorders and treatment approaches. This
paper will outline a theoretical basis for a class of disorders identified here as disorders of
regulation, apply a systems-based approach to the modeling of these diseases and delineate some
principals that could be used to create viable treatments.
Some systems are capable of complex behavior that lasts for long periods of time. Some
systems are not and degenerate into a form of repetitive order (Shank et al., 1999). An example
of the latter in the brain is the kindling effects of grand mal seizure activity (Shousea & Ryan,
1984). In the kindling of a seizure the brain begins to synchronize its firing until an entire
hemisphere fires at once. During a seizure the brain system becomes highly structured and
organized, and the ability to produce complex human behavior is diminished. It is only within a
coherent range of systemic functioning that complex behavior can emerge and be maintained. In
other words, if the disruptions in the functioning of a system of the body or brain leads to a
reduction of complex behavior or a disruption in the functional balance between systems, it can
lead to disease and, in some cases, death.
A complex system has points in its state space in which it is highly vulnerable to
disintegrating into one of the aforementioned patterns of loss of systemic complexity (Prigogine
& Holte, 1993). The balanced oscillation between systems forms a coherent pattern of
relationship. If that coherent pattern of relationships is disrupted in minor ways, the system will
display small perturbations in functioning until it can right itself (Shank et al., 1999). If that
3
system exceeds its regulatory capacity at a key point for long enough or with sufficient intensity,
the entire system adapts, forming a new functional relationship between systems. The behavior
of many disease processes appears to be created and maintained by disruptions in the functional
relationship between systems.
At times it is the body’s own processes that are involved in the genesis and maintenance
of these syndromes (D. Kerr, personal communication, August 08, 2008). Some disorders that
appear to be effected by this type of disease process are autoimmune disorders, many types of
cancers, and neurodegenerative disorders. The causes for each of these disorders are highly
varied, as will be the treatments postulated; however, these disorders follow a similar pathway of
disruptions in the functioning of systems. This similar pathway can be a guide for the creation of
appropriate and effective clinical interventions.
Evidence that Dysregulation Leads to Pathology
There are many data points supporting the postulation that dysregulation leads to
pathological functioning in the body, brain and mind. Multiple studies have shown that over-
secretion of cortisol in both Cushing’s syndrome and depression leads to reduction in the cellular
density and dendritic connection in the hippocampus (Bourdeau et al., 2002; Bremner, Narayan,
Anderson, Staib, Miller, and Charney, 2000; Starkman and Schteingart, 1981; Dorn, Burgess,
Friedman, Dubbert, Gold, and Chrousos, 1997). Cortisol dysregulation in many leads to loss of
sleep. Loss of sleep in turn can lead to a reduction of neurogenesis in the hippocampus (Guzman-
Marin et al., 2005). Overproduction of insulin leads to symptoms including mood swings, weight
gain, hypoglycemia, increased facial hair in women, hair loss, bloating, and high blood pressure
(Norman, 2010). Underproduction of dopamine can lead to movement disorders, dementia,
alterations in working memory and symptoms of psychosis. Overproduction of Telomerase has
4
been associated with development of cancerous tumors (Harley and Villeponteau, 2002).
Overproduction of glutamate can lead to cell death; elevated levels of glutamate are found in
dementia, Alzheimer’s type (Choi, 2004; Ulas et al., 1994).
Telomerase: A Regulator of Cellular Life Span
Cells in the body are created, maintained for some time and then die. Different regions of
the body and brain require different rhythms of cellular lifespans. In the span of one week most
pancreatic cells are replaced by other cells; over of the span of several years all the cells in the
body are replaced by other cells. This rhythm of cell loss and replacement requires a consistent
cycle of cell death, cell birth and cell maintenance. If these key functions begin to occur either
too frequently or too rarely, pathology can develop (Lowe & Lin, 2000). This exemplifies the
nature of disorders of regulation. They are disorders where the balance between the functional
demands of a system is disrupted to the point that other systems relying on that system and the
body as a whole no longer function effectively.
Telomerase is one of the key regulators of cellular life span (Gorbunova, Seluanov, &
Pereira-Smith, 2002).Telomeres are short, compacted segments of DNA forming a cap at the end
of the chromosomes (Gorbunova, Seluanov, & Pereira-Smith, 2002). As the cell goes through
each cycle of mitosis the telomere is shortened (Blasco, 2005). These segments of the
chromosomal DNA (telomeres) function as regulators for cell division. As they shorten past a
threshold point the cell can no longer divide and produce other cells (Blasco, 2005). Many
tissues require more cell divisions than the original telomere length allows. In order to maintain
physical health and systemic regulation, an enzyme called telomerase is produced (Gorbunova,
Seluanov, & Pereira-Smith, 2002). This enzyme stimulates the rebuilding the telomere end cap
of the chromosomes. The end cap is rebuilt thus extending the ability of cells to divide and
5
replace cells at the end of their life span (Blasco, 2005).
Dysregulation of telomerase.
A hallmark of disorders of regulation is that there are many possible events that can cue
any single disruption in the functioning of a system. These disruptions lead to major systemic
functions occurring in dysynchronous manner. The dyssynchronous functioning of one system
can lead to large-scale dysfunctions in other systems. Dyssynchronous systemic functioning can
lead to what appears as symptoms of disease. There are multiple events and classes of events that
can lead to dysregulation of telomerase production, as there are multiple ways that telomerase
production can be altered (Epel et al., 2004). Some of these include genetic differences (e.g.,
disruptions in the Ras and Raf genes), autonomic stress, lifestyle choices, mental health and
chemical toxins, to name a few (Lua, Fua, and Mattson, 2001; von Zglinicki, 2002; Epel et al.,
2004). The effects of these events can act in isolation or in some cases synergistically to alter the
functioning of telomerase, if an individual has a genetic predeterminent for dysregulation of
telomerase that may or may not be sufficient to create major disruption in the tissues of the body.
If, however, that genetic determinent occurs along with other factors there could be a catalyzing
event leading to large scale disruptions in tissues and thus disease processes. In one study of a
telomerase deficient mouse, there were not global increases in symptoms of aging (Chang,
2004). However, the mice displayed reduced ability to repair injuries or recover from illnesses, a
shortened lifespan, and increased incidence of cancerous tumors (Serrano & Blasco, 2001). Thus
leaving them vulnerable to multiple disease processes.
Lifestyle and telomerase production.
A recent study implied that lifestyle changes such as alterations of diet, exercise, and
stress levels have a significant association with re-regulation in telomerase activity (Ornish et al.,
6
2008). The study notes that causation cannot be inferred and that a more comprehensive
randomized study is warranted from these results (Ornish et al., 2008).
Comprehensive lifestyle changes significantly increase
telomerase activity and consequently telomere maintenance
capacity in human immune-system cells. Given this finding and the
pilot nature of this study, we report these increases in telomerase
activity as a significant association rather than inferring causation.
(Ornish et al., 2008)
Stress and telomerase.
Telomerase has an interaction effect with the autonomic stress response (Choia, Faucea,
& Effros, 2007; Epel et al. 2004). High levels of autonomic stress and cortisol have been
associated with down-regulation of telomerase and shorter telomeres (Choia, Faucea, & Effros,
2007; Epel et al., 2004). Allostatic load is the term coined by Bruce McEwen to describe amount
of energy needed to return a system to homeostasis. Long-term exposure to stress hormones due
to prolonged stressors or a very large stress response leads to a higher allostatic load (McEwen,
2002). Telomerase production is one of the systems affected by overproduction of stress
hormones (Choia, Faucea, & Effros, 2007). Several studies have noted that changes in telomere
length occurred in response psychological stress and mood disorders. T lymphocytes exposed to
high levels of cortisol have displayed a significant reduction of telomerase activity during both
primary and secondary stimulation of cells (Choia, Faucea, & Effros, 2007; Epel et al., 2004).
Dysregulation of telomerase: neurodegenerative disorders.
The pattern of disorders of regulation, starting as a disruption in one system and leading
to another, can be seen in the effects of prolonged autonomic stress response (Epel et al., 2004;
7
McEwen, 2002). It is likely that cortisol levels produce a heightened risk environment that sets
the stage for other factors to catalyze into either a cancer or neurodegenerative disorder (R.
Sapolsky, personal communication, 2001). Recent studies have shown that TERT is present in
pre-differentiated neuronal blast cells but it precipitously drops off in adult neurons. Deficits of
TERT present in the cellular context have been shown to be predictive of the development of
neurodegenerative disorders. The presence of TERT is protective against the cell death due to
apoptotic factors and the onset of apoptotic cascade (Bermudez, Erasso, Johnson, Alfonso,
Lowell, & Kruk, 2006; Lua, Fua, & Mattson, 2001).
We found that expression of hTERT, the catalytic component of
telomerase, was sufficient and specific to reduce caspase-mediated
cellular apoptosis. Further, hTERT expression reduced activation
of caspases 3, 8, and 9, reduced expression of pro-apoptotic
mitochondrial proteins t-BID, BAD, and BAX and increased
expression of the anti-apoptotic mitochondrial protein, Bcl-2. The
ability of telomerase to suppress caspase-mediated apoptosis was
p-jnk dependent since abrogation of jnk expression with jip
abolished resistance to apoptosis. (Bermudez, Erasso, Johnson,
Alfonso, Lowell, & Kruk, 2006).
Increased cell death is a key aspect of multiple neurodegenerative disorders (Okouchi,
Ekshyyan, Maracine, & Yee, 2007). Down-regulation of telomerase is present in individuals with
Alzheimer’s dementia (AD), Parkinson’s dementia, Amyotrophic lateral sclerosis and Fronto-
temporal dementia, to name a few. In AD shortened telomere length was noted in T-
lymphocytes. This reduced telomere length correlated with scores on MMSE: “the
8
proinflammatory cytokine TNFα (a clinical marker of disease status), with the proportion of
CD8+ T cells lacking expression of the CD28 costimulatory molecule, and with apoptosis.
(Panossian et. al, 2003).” Another study finds similarly, “Thus, a simple count of chromosome
ends for the ‘presence/ absence’ of fluorescence (marking telomerase) may provide a valid
biomarker of dementia status” in individuals who meet the criteria for AD (Jenkins, 2008).
Alterations in the “SOD1 gene [and] deletions of the telomeric copy of the SMN gene”
were noted in individuals with motor neuron disease (Orrell & Figlewicz, 2001). An
upregulation of telomerase has been shown to be neuroprotective and to reduce the chance of an
apoptotic cascade (Mattson, 2000). There are many factors that increase or reduce the risk of
apoptosis (Mattson, 2000). Some recent studies have indicated that telomerase is important to
responses to insults to the brain as well as neural development (Mattson, 2000; Lua, Fua, and
Mattson, 2001). The regulation of apoptosis is a key role of the immune system (Feig & Peter,
2007). The disruption in immune functioning due to reduced telomere length is a key example of
how a disruption in one system’s functioning can lead to the disruptions in another (Rudolph,
Chang, Han-Woong, Blasco, Gottlieb, Greider, and DePinho, 1999). The under-regulation of
telomerase increases susceptibility to apoptosis and correlates with dementia ratings and
increased inflammation, possibly linking alterations in immune functioning and
neurodegenerative disorders (Lua, Fua, and Mattson, 2001).
Life in the balance: The effects of telomerase an oxidative stress on cellular senescence.
Oxidative stress has been shown to reduce the length of telomeres and is not repaired as
easily as damage to other areas of the DNA (von Zglinicki, 2002). Oxidative stress has been
shown to play a key role for enchained cellular senescence and antioxidants have been shown to
decelerate cellular senescence (von Zglinicki, 2002; Naka, Akira, Ikeda, & Motoyama, 2003).
9
There is an interaction between regulation of oxidative stress and the regulation of replicative
processes (von Zglinicki, 2002). One study has found that increased anti-oxidant protection plays
a key role in the ability of embryonic stem cells to remain pluripotent even after multiple mitotic
cycles (von Zglinicki, 2002; Naka, Akira, Ikeda, & Motoyama, 2003). This also points to the
interrelationships between systems. Upregulation of telomerase can reduce the effects of
oxidative stress and stop a cell from entering senescence, even in an oxidative upregulated
context (von Zglinicki, 2002; Naka, Akira, Ikeda, & Motoyama, 2003). Upregulation of
telomerase can also play a key role leading to immortalization of cells and the onset of a
cancerous replication cycle. One could easily imagine that a balance is established between
oxidative stress and telomerase that, if exceeded in either direction, could cause significant
disruptions to a single tissue/system or at a more global, body-wide level.
Systemic stress.
Systemic stress is a term this author uses to describe when the change to any one system
in the body enters into a state that exceeds its normal range of ability to return to a baseline of
functioning thus requiring increased energy or adaptations in other systems in order to return to
baseline. An excellent example of how certain types of systemic stress can be virtually
irreversible is the way cells can enter into stress-induced senescence. Senescence is the lack of
ability for a cell to continue to reproduce through mitosis. When cells are “exposed to sublethal
(systemic) stress” they will often enter what is known as stress-induced cellular senescence
(SIPS) (Naka, Akira, Ikeda, & Motoyama, 2003). Like with most disorders of regulation SIPS
can be triggered by multiple means: exposure to UV light, radiation, oxidative stress and other
external insults that damage the length of the telomere. These cells display key markers of
cellular senescence, such as flattening of the cell body, β-galactosidase activity, and a rapid
10
reduction in telomere length. Overproduction of reactive oxygen can induce cellular senescence
(Naka, Akira, Ikeda, & Motoyama, 2003). The cellular senescence brought on by oxidative stress
cannot be reversed with an upregulation of telomerase. Similar findings exist for damage due to
other factors such as radiation. This indicates that global DNA damage also can induce
premature cellular senescence (Naka, Akira, Ikeda, & Motoyama, 2003). The oncogenes Ras and
Raf also trigger what appears to be stress-induced cellular senescence, resulting in a permanent
arrest to the cycle of mitotic replication. This is known as ontogenetic stress-induced senescence
(Naka, Akira, Ikeda, & Motoyama, 2003).
Global and local effects.
Adding to the complexity are the possible effects of global systems that set the tone of
multiple systems or in some cases the entire body. When there is a global disruption it can lead
to many symptoms that appear unrelated and disconnected from a single cause. Some of these
global systems could include diurnal patterns of endocrine and neurotransmitter production,
sleep cycles, autonomic stress reactivity and so on. As stated above, disorders of regulation is
that any single system’s functioning can be affected by many different events. These events can
be localized to a system or an area of tissue, such as the shortening of telomeres when oxidative
stress is upregulated in a specific region, and these events can also be global, such as with broad
scale damage to the DNA structure from radiation that leads to a cell entering senescence early
and losing the ability to replicate (von Zglinicki, 2002; Naka, Akira, Ikeda, & Motoyama, 2003).
To exemplify this overproduction of insulin effects the entire body including mood swings,
weight gain, hypoglycemia, increased facial hair in women, hair loss, bloating, and high blood
pressure (Norman, 2010).
11
Complexity small changes big results.
As small differences happen in the replication process they can lead to large-scale
alterations in system functions (Briggs & Peat, 1989). This type of difficulty has a particular
sensitivity to initial conditions. Cellular senescence highlights this type of change. Multiple
replications of cells and the process of cell reproduction are ripe for dysregulation that mirrors
the dysregulations that are possible in the population growth equation.
Second Impact Syndrome
When individuals have a head injury there are profound alterations in the internal
working and chemical dynamics of the brain (Yoshinoa, Hovda, Kawamata, Katayama, &
Beckera, 1991). These shifts in metabolic and chemical functioning are intended to protect the
brain and allow it to heal after a concussive injury (Giza & Hovda, 2001). Medicine, even the
body’s own medicine in sufficient dosages, can cause damage. If certain aspects of the brain’s
functioning exceed its regulatory capacity, there can be a major loss of brain tissue and
functioning from what seems like insignificant insults to the brain (Giza & Hovda, 2001). This
process of an initial dysregulation of brain metabolism leading to vulnerable states from which
even a minor insult (e.g., small impact to the skull) leads to significant brain damage is an apt
example of the process of systemic dysregulation leading to a disease process.
The functional dysregulation model of disorders of regulation would hold that as a
system exceeds its regulatory capacity in one area, the entire system begins to adapt. It also holds
that there are key areas, times or states of vulnerability from which exceeding the regulatory
capacity would lead to fundamental alterations in the systemic function. These alterations occur
more frequently in two situations: 1. When the body’s defensive strategies put the system into
systemic stress (high allostatic load on the system) or 2. The dysregulation of one system
12
precludes the effective functioning of another, thus leading to that system no longer functioning
as a regulatory boundary for the dysregulated system and the non-functioning system no longer
being able to perform its vital function, leading to more systemic adaptations and so on. In
McEwen’s (2002) theory of allostatic load alterations in stress response, either in intensity or
duration, the context leads to systemic adaptation. In this model I would extend the idea of
allostatic load to any significant functional adaptation of a systemic relationship due to the
inability to return to a functional baseline of oscillatory patterns between systems.“It is during
the post-injury period, when cellular metabolism is stretched to its limits, that the cell (and the
brain) is most vulnerable to further insults (Giza & Hovda, 2001).”
Multiple Causes – Multiple Systems
There are multiple systems in the body. Each system has multiple contextual events that
maintain its functioning. Disruption to a system can come from any surrounding system. Some
systems have global reach and can affect the entire organism at once. This adds a significant
layer of complexity in understanding the antecedents of symptoms. To put this in more concrete
terms, a neuron will die under many conditions (Trump, Berezesky, Chang, & Phelps, 1997).
Some of these include increased metabolism, excessive glutamate production, apoptosis inducing
factors, genetic abnormalities, epigenetic mutations, exceeding its number of mitotic cycles,
oxidative stress, being attacked by t-cells and so on. To quote John Muir, "When we try to pick
out anything by itself, we find it hitched to everything else in the Universe.” In the case of these
disorders, this is true. However, the goal is to identify the corner being tugged on and stop the
tug. The aforementioned list contains some of the systems affecting a neuron’s life. There are
countless other systems.
13
Implications for treatment
The difficulty in addressing the symptoms of disorders of dysregulation stems from: a.
there being multiple possible causes for a single outcome, b. second order effects where the
primary cause is not the main cause for presenting symptoms, c. the symptoms are created by
adaptations in the functioning of the body’s own processes, d. there can often be additive effects,
and e. small disruptions can become larger over many iterations, such as through mitosis, across
a life span, thus making effect and causal events not apparently contiguous in time.
In this model creating a treatment is much more precise and therefore labor intensive than
in traditional treatments. The goal of treatment is not amelioration of symptoms but the re-
establishment of a coherent relationship between systems. This requires: a. identifying the areas
of dysregulation, b. identifying the main functions and regulators of these areas, c. in some cases
differentiating primary, secondary and tertiary symptoms, and d. interventions aimed at re-
establishing the body’s natural ability to return to a coherent allostatic range.
Current Treatments Using Systems Approaches to Treat Disorders of Regulation
There are many current treatments that utilize this type of reasoning to reestablish a
homeostatic pattern or to help a dysregulated system find a new stable pattern of systemic
oscillation. Some of these are: defibrillation as a treatment for certain classes of myocardial
infarction, deep brain electric stimulation for movement based Parkinsonian symptoms, motor
neuron atrophy due to encephalitis infection, cognitive behavioral therapy for depression, saline
trigger point injections for pain, and mindfulness-based stress reduction. Exploring existing
treatments and appling systems thinking to understanding patterns of dysregulation explanatory
model of how a treatment works to re-establish a coherent pattern of functioning could illuminate
14
this further.
Neural Adapted Sinibus Virus (NSV)
Neuroadapted Sindbis virus (NSV) in humans is a moderate lung infection; in mice it
leads to a severe sickness that if it infects the brains of animals will cause paralysis, a stripping
of dendritic connections, global excitotoxic nerve death and eventually the death of the animal.
One of the key findings of the study of this infection is that the viral infection is not the direct
cause of these terrible effects (D. Kerr, personal communication, 2008). The animal will become
quite sick but it is not the infection of the cells that leads to the cell death. The virus it self leads
to the death of only around 20% of the neurons while at the end of three weeks the animal
displays 95% neuron loss. Douglas Kerr (2008) and his research group found that it is
dysregulation of autoregulatory functions of the microglial and a metabolic protective defensive
strategy in the neurons that leads to the catastrophic loss of cells.
It is a dysregulation of the re-uptake of glutamate and an increased neuron signaling of
stress through the secretion of nNOS that leads paralysis and death. What the researchers did not
know at first was that this upregulation of nNOS and the dendritic sloughing was a protective
strategy that is vital for a cell near metabolic overload. The secretion of nNOS signals to the
neuron it is in danger of excytotoxicity and evokes a protective strategy which is to reduce input.
If the dendrites coming into the neuron continue to signal it to fire, it will enter a metabolic crisis
and die; thus, dropping the dendritic connections protects the neuron against over-excitation,
attempting to be a shutoff switch if the neuron in colloquial speech, “overheats”.
However, because this virus leads to global upregulating of nNOS (rather then local
upregulation more normative for axotomy injuries) and the sloughing of dendritic connections in
the entire motor cortex the system is primed another form of paralysis and of cell death induced
15
by isolation from other cells (D. Kerr, personal communication, 2008). Indeed when the
researchers down regulated nNOS and TNFa-alpha, what they found was that the dendrites
remained connected but the cells died due excytotoxitcity. The sloughing of dendrites is
protective function that if it happens on local level likely can be protective. If upregulation of
nNOS happens on a global level it exceed the brains capacity to regrow connections after the
need for the initial protective response has passed. Researchers working in systemic manor were
able to halt this process by providing other forms of protective interventions for the time when
the cells were in a vulnerable state.
From the systems perspective there are five key factors in this process. The first is that
there were alterations in the current system state due to a viral infection leading to priming
effects for a catastrophic cell loss. The second is that researchers identified the system areas
where the system entered a state space vulnerable to produce the results noted (e.g. finding
events that could cause dendritic sloughing). The third is that there were synergistic interactions
between multiple systems defensive responses that lead to disruptions in the ability of the state
space to return to homeostatic range after an allostatic protective response. Fourth is that the
team identified several systems and their defensive actions. In other words they identified the
key protective functions that are leading to the neuron entering into an allostatic response. Fifth
the team found ways to hold the system stable while time dependant defensive strategies could
complete and reduced the signaling for other defensive strategies that lead to the synergistic
catastrophic destabilizations of the spinal motor neuron functioning. Due to these contextually
relevant interventions during the critical period the normal and typically quite stable functioning
of the motor neuron system was allowed to reassert itself through re-establishing the regulatory
boundaries already present in the system (D. Kerr, personal communication, 2008).
16
Applying a systemic approach to understanding the formation of symptoms of Alzheimer’s
Dementia: Role of Systemic Dysregulation in Alzheimer’s Dementia
AD, or Dementia of the Alzheimer’s type, has multiple precipitating events that lead to
the development of this condition (Attix & Welsh-Bohmer, 2006). The multiple precipitating
events are often additive, leading to a synergistic risk for the neurodegenerative disorder. The
traditional theory of AD is that there are disruptions of gene expressions that can lead to
development of AD (Bullido et al., 1998). Some recent theories hold that the disruption of gene
expression needs catalyzing events to make the transition from genotype to phenotype (Bullido
et al., 1998). Little consideration is given to epigenetic alterations in both gene expression and
gene patterns (Becker, 2004; Wang, Oelze, & Schumacher, 2008). Theorists have indicated that
many instances of AD are not accounted for by the current theories of the pathophysiology of
AD (Becker, 2003). The central tenet of the thesis presented here is that while the genetic theory
is accurate, there is another relevant story about the role of systemic regulation and interactions
between brain, body and environment that could have implications for understanding and
treating neurodegenerative disorders (Wang, Oelze, & Schumacher, 2008).
The systems dysregulation paradigm starts out with the premise that there are many roads
to neurodegeneration and there are multiple events and classes of events that can synergistically
lead to the same type of dysregulation and similar patterns of dysfunction. To exemplify this,
disruptions in cortisol secretion can be brought about through: a. tumors in HPA axis, b.
17
depression, c. PTSD, and d. disruption in early attachment (Young, Abelsona, & Camerona,
2003; Yehuda, Teicherbc, Trestmana, Levengooda, & Sievera, 1996; Penza, Heim, & Nemeroff,
2003; Dorn, Burgess, Friedman, Dubbert, Gold, and Chrousos, 1997). Dysregulation of cortisol
has been associated with hippocampus shrinkage, loss of sleep patterns, disruption to the
dopamine system, disruptions in concentration, toxic cell death and anhadonia (R. Sapolsky,
personal communication, 2001). This is only one system. It is an important system for
autoregulation but not by any means the only.
From the systemic dysregulation paradigm, in order to for the genotype of AD to become
the phenotype of AD, there would need to be disruptions in the ability to form memories,
maintain hippocampus volume, rate of cell death, mitosis and maintain previously encoded
memories. These dysregulations could occur in multiple levels of the system. The interplay
between life events, environmental toxins, volitional behaviors and chemical contexts and the
functional relationship between anatomical structures are a few of the factors that could add to
disruptions in memory retention and formation. From a systems perspective this is not a surprise
because there are often several key areas of vulnerability in a system that lead to increased risk
for these patterns’ systemic dysregulation, not simply one.
Systemic Disruptions Present in Individuals with AD
In the paths leading to Alzheimer’s there are some major themes that emerge. These are:
a. changes in stress response, environmental stressors and systemic stress, b. changes in
subsystem functioning, c. functional shifts between systems, d. alterations in the metaplastic
environment, and e. changes in cellular senescence (Attix & Welsh, Bohmer; Chen, Kagan,
Hirakura, & Xie, 2000; Sorg et al., 2007; Peskind, Wilkinson, Petrie, Schellenberg, & Raskind,
2001). Mapping some of these key changes could illuminate possible places in the system where
18
one might create interventions to re-establish the neurobiological system’s functional capacity to
maintain itself and auto-regulate.
Alterations in Stress Response
Abnormal stress response has been noted in many individuals with AD (Popp et al.,
2009). High levels of CORT have been shown to be associated with loss of hippocampal tissue
in depression, PTSD, Cushing’s syndrome and AD (Young, Abelsona, & Camerona, 2003;
Yehuda, Teicherbc, Trestmana, Levengooda, & Sievera, 1996; Penza1, Heim, & Nemeroff,
2003; Dorn, Burgess, Friedman, Dubbert, Gold, & Chrousos, 1997). In one study of the effects
of reducing CORT levels in individuals with Cushing’s syndrome, it has been noted that the
hippocampal tissue regenerated significantly (Starkman, Giordani, Gebarskic, Berent, Schork, &
Schteingart, 1999). Upregulation of CORT is associated with anhadonia, psychomotor
retardation, poor memory encoding, lack of ability to concentrate, loss of interest in sexuality,
increased anxiety and aggression. PTSD is a risk factor for the development of AD. In both AD
and PTSD there is a decrease in heart rate variability (HRV) in AD a direct relationship was
noted between HRV and symptom severity (Zulli et al., 2005; Cohen et. al., 1998, Zulli et al.,
2005).
Alterations in CORT levels also lead to disruptions in the dopamine system,
norepinephrine system, the serotonin systems and the acytocholine system (Oswald et al., 2005;
Pacaka et al., 2002; Idoyaga-Vargas, Abulafia, & Calandria, 2001; Geracioti, et Al., 2001; Pacak,
Palkovits, Kopin, & Goldstein, 1995; Meshorer, & Soreq, 2008; Kirkwood, Rozas, Kirkwood,
Perez, & Bear, 1999). CORT also plays a role in the regulation of dopamine and dopamine loss
could lead to increased LTD and reduced LTP (Calabresi et al., 2000). Cortisol may end up
being a large system that, if dysregulated, can lead to many systemic changes (R. Sapolsky,
19
personal communication, 2001). Alterations in stress response due to life stress in younger
animals have been noted to create large-scale changes in multiple systems. “Compared to
controls, traumatized animals showed an increase in Ca2+ homeostatic proteins, dysregulated
signaling pathways and energy metabolism enzymes, cytoskeleton protein changes, a decrease in
neuroplasticity regulators…, and an increase in apoptotic initiator proteins (Uys, Hatting, Stein,
& Daniels, 2008).” CORT production decreases telomerase production throughout the body
(Choia, Faucea, & Effros, 2007; Epel et al., 2004). Telomerase down-regulation has been
implicated in AD and other neurodegenerative disorders (Mattson, 2000).
There are multiple determinants for the systemic functioning of CORT; genetics, diet,
environmental factors, and epigenetic factors. Some environmental factors are number of adverse
childhood events, PTSD, disruptions in infant child bonding, exercise, and social support (Shea,
Walsh, MacMillan, & Steinera, 2004). Genetics has been implicated in the creation of the
autonomic set point for CORT production and changes in set point were catalyzed in a gene by
environmental interaction (Adamafio, 2009; Wüst, Federenkoa, Hellhammera, & Kirschbaumb,
2000; Kirschbaum, Wust, Faig, & Hellhammer, 1992). Even short-term disruptions in parent-
child bonding or mild increases in parental aggression lead to significant life time alterations in
CORT response (Van Oersa, Kloetb, & Levinea, 1998). Number of adverse childhood
experiences correlates with increased depression, anxiety and self-destructive behaviors (Felitti,
Anda, Nordenberg, Williamson, Spitz, & Edwards, 1998).
Neuroplastic Dysregulation
The brain is a learning context. Each time we move, think or plan we are sculpting the
brain. In order for the brain to maintain optimal functioning it needs to balance its rate of change.
If it changes too quickly the brain becomes an unstable environment. If it does not change
20
quickly enough it will have difficulty learning to map experiences. There are currently seven
main processes that describe how the brain maintains its internal architecture at an optimal level
of flexibility: long-term potentiation, long-term depression, activity dependent changes,
metaplasticity, neurogenesis, kindling and salience effects (Burrell & Sahley, 2004; Eriksson et.
al., 1998; Ichise et. al., 2000; Kalivas, & O'Brien, 2008; Malenka, & Nicolla, 1999; Minabe &
Emori, 1992; Mueller, Pollock, Lieblich, Epp, Galea, & Mistlberger, 2007; Nitsche et. al., 2006;
Wickliffe & Bear, 1996). These are the learning-based regulators identified to date. Along with
these regulators there are also metabolic and structural regulators of plasticity. One of these is the
Mglur (Matabtropic glutamate receptors) that, when stimulated, reduce the number of AmpR
receptors attempting to control and regulate the level of excitatory potential possible. If the level
of excitatory potential is exceeded, then the neuron can die due to excytotoxicity (Choi, 1992).
Individuals with AD have multiple alterations in the rate of neuroplasticity (Arendt,
2003; Kimmo, Krystyna, Tomoaki, Daniel, & Stanley, 1999; Flood, & Coleman, 1990; Shankar
et. al., 1990). They also have been noted to display a decline in cell density in the hippocampus
that spreads over the course of the disease to other brain areas. One of the key areas of
dysregulation is in the actocholine systems which is a global regulator of the rate of plasticity
(McKay, Placzek, & Dani, 2007; Whitehouse, Martino, Antuono, Lowenstein, Coyle, Price, &
Kellar, 1986). Most current AD meications are acytocholinergic. Another effect on plasticity is
Abeta (often expressed in individuals with AD) in one study found to be a clear down-regulator
of LTP, possibly effecting neuronal pattern stability (Chen, Kagan, Hirakura, & Xie, 2000). In a
2010 study in a Prelisin 1 knock out mouse (a mouse model of some of the processes in AD)
there was noted an upregulation of LTD. These mice show an upregulation of LTP (early phase
LTP, late is phase similar to controls) early in life that drops off as they age. This produces a
21
high rate of quick changes that do not get stabilized into fully encoded neuronal patterns. This
upregulation thus leaves the brain vulnerable to destabilizing the patterns of connectivity and
catastrophic LTD. Another study found that chronic exposure to adrenergic stimulation (common
in those with AD) upregulated the LTD mediated by Alpha1AR (McElligott, & Winder, 2008;
Davis et. al., 1996; Popp et. al., 2009).
Endocannabinoids are another regulator of the rate of plasticity (Kyriakatos & Manira,
2007; Pazos, Núñez, Benito, & Tolón, 2004). They function as agents facilitating the increase of
excitatory synaptic plasticity, LTD and LTP (Pazos, Núñez, Benito, & Tolón, 2004). The
endogenous canibinoids are regulated by a calcium dependent mechanism. The mechanism
works like a switch priming cells to release eCBs but only coupled with a transient rise in Ca2+.
Another group of researchers found “the existence of profound changes in the location and
density of several elements of (the endocanibanoid) system in Alzheimer's disease tissue
samples, indicating that a non-neuronal endocannabinoid system is up-regulated in activated
glia” (Pazos, Núñez, Benito, & Tolón, 2004).
Multiple studies have shown changes in Serotonin in individuals with AD (Mintzera, et.
Al, 1997). Sertatonin has been noted to “regulate cell proliferation, migration and maturation in a
variety of cell types, including lung, kidney, endothelial cells, mast cells, neurons and astrocytes”
(Azmitia1, 2001). Alterations in the dopamine system (increasing D2/D3 receptors) have been
noted in individuals with AD (Reeves, Brown, Howard, & Grasby, 2009). Dopamine is a key
mediator of LTP and LTD firing for both salient (both positive and negative) and positive salient
events (O. Hikosaka, personal communication, May 7, 2009). If there is an action potential
previously associated with a dopamine response, that fires without dopamine modulation there is
a stimulation LTD. The acytocholine system and the dopamine system work in conjunction,
22
modulating the rate of plastic change. Upregulation or down regulation of dopamine could have
profound effects on rates of neuroplastic change (Shen, Flajolet, Greengard, & Surmeier, 2008).
Another class of neuroplastic regulation is the activity dependent class of regulation.
Activity has been shown to prime patterns of cellular connectivity for LTP (Antonov, Antonova,
Kandel, & Hawkins, 2003). Previous firing of a pattern of neurons also primes the neurons for
firing. This is could easily form a positive feedback loop of learning. LTD is upregulated by
excitatory activity that does not produce a full action potential (Stanton, 1995). If multiple cells
send positive signals for action potential to a cell and that cell does not fire, this increases the
likelihood of LTD (Calabresi, Maj, Pisani, Mercuri, & Bernardi, 1992).
The ratio of signal to noise is another activity-dependent regulator of plasticity (M. Bear,
personal communication, June 16, 2003). The brain as a learning context would more likely be
served by encoding accurate patterns than irrelevant patterns. The mind is capable of tracking
irrelevant patterns if this system is co-opted by other regulators; among these is salience effects
and another is signal noise. If the pattern recognized by the brain is highly noisy (e.g., not a
strong predictable relationship) this would likely indicate that it is not a pattern. For phylogenic
reasons the brain would have been unlikely to evolve if it tracked too many irrelevant patterns.
Increased signal noise is a better predictor of LTD than long-term disuse of the synapse. In a
study conducted by Marc Bear (2003) Cats who have an eye disrupted chemically so that no
signal is getting to the brain, have less LTD in the visual cortex than a cat who wears a single eye
patch. In the cat with the patched eye, the increased noise of the eye seeing the blackness alters
the patterns of the visual cortex more with more loss in connections then the cat with the nerve
signal chemically blocked. In other words higher signal noise ratio lead to more LTD (M. Bear,
personal communication, June 16, 2003). This fact has profound implications for the loss of
23
memory in AD: Earlier memories whose pattern is both more encoded and least likely to be
triggered by current context would be the most undisturbed by this process. It is possible that the
memory is evoked and the heightened plastic context, in conjunction with the increased noise-to
signal-ratio due to the flattening of salience indicators, leads to a destabilization of the cellular
pattern that marks the memory of the event. This destabilization in turn leads to a loss of
connectivity and eventually to cell tissue death.
In older adults there are several key possibilities increasing signal to noise and thus the
likelihood of a catastrophic loss of connectivity and apoptic cascade. These are: a. poor sensory
information due to changes in physical sensors (ear drum, hairs in the ear, hardening of the
cornea, slower reaction times, reduced sense of smell); b. inaccurate reading of the sensory
information by brain systems (changes in ear structure have not changed how the brain receives
the information from the ear); and c. internal events (stress, anxiety, pain, fear, other affect,
worry, thoughts creating internal distraction and increased signal noise) (Mahncke, Bronstone, &
Merzenich, 2006). This last class (internal events) is significant in older adults and has not been
discussed thoroughly as an impact on brain health in aging. Another final note on the activity
dependent regulators of plasticity is that in older adults, particularly in Western cultures, there
are alterations in lifestyle that can reduce the amount and quality of environmental stimulation. It
is interesting to note that increases in mobility, social stimulation and environmental novelty
have been noted to reduce symptoms of AD and be protective factors against developing AD
(Nithianantharajah & Hannan, 2006).
Major Types of Neuroplastic Change and Their Implication for Systemic Dysregulation
Regulation of Metaplasticity: Upregulation of long-term potential could in certain system
states lead to forming many patterns that are weakly associated, as in the blooming and pruning
24
cycles early in life. This cycle leads to the formation of many new but not entirely accurate
patterns of neuronal associations. Later these patterns are not supported by external events. In
essence, an upregulation of metaplasticity, increasing LTP past a certain point, could lead to an
upregulation of LTD due to increased signal–to-noise ratio, thus leading to receptor loss,
destabilization of balance between functional areas, axonal connections and eventually cell
death.
Down-regulation of LTP, telomerase activity and neurogenesis could lead to an inability
to replace cells and rebuild damaged synapses. This leads to a slow but steady attrition of
memory and abilities that eventually reaches a crisis point when the relationship between
external events has such dramatic increased noise that it precipitates a rapid rate of LTD.
A metaplastic decreased LTD could lead to many patterns being encoded and competing
for attention there by disrupting retrieval and overloading pattern recognition with relevant
possibilities. It is also possible that this leads to a single external event triggering in a context
irrelevant manor many patterns of neuronal relationships disrupting the synchronous patterns of
firing.
Regulation of Neurogenesis: Neurogenesis is the forming of new neurons in the brain.
Until recently it was believed that neurogenesis stopped as an individual reached adulthood. An
under-regulation of neurogenesis could lead to lack of replacement for damaged cells, reduced
memory formation and overtime destabilization of current patterns of neuronal connectivity.
Regulation of Salience Effects: If dopamine cells fire in conjunction with a pattern of
neuronal firming, this marks the pattern or an aspect of the pattern as highly salient for the
continued functioning of the organism. This increases the possibility of LTP forming a new
neuronal pattern. If a pattern fires without a dopamine signal. the pattern is more likely to
25
destabilize and enter the LTD cycle. To be precise it is currently thought that dopamine fires at
both salient (both positive and negatively relevant events) and positive events.
Acytocholine Cells: These cells increase the sustained attention on the external event and
thereby the accurate encoding of a neuronal map of that pattern (Himmelheber, Sarter, & Bruno,
2000). Increases of ACh lead to a metaplastic increase in the rate of neuroplasticity
(Jerusalinsky, Kornisiuk, & Izquierdo, 1997). Upregulation in ACh could lead to destabilized
patterns of relationship due to the formation of neural circuits that are only loosely reflected in
the external context. An under-regulation of ACh could lead to a lack of ability to form new
memories and reduced activity-dependent maintenance of current patterns of neuronal firing.
This dysregulation would likely not produce a boom and bust cycle but rather produce a slow
degradation of memory and memory formation with an exponential increase in loss toward later
parts of the disease process.
Brain Derived Neurotropic Factors (BDNF) Regulation: BDNF is a protein responsible
for the growth and maintenance of nerve cells. As well as its action in the brain, it also plays a
role in motor neurons, kidneys, prostate and is often present in saliva (Binderm & Scharfman,
2004; Huber, Hempstead, & Donovan, 1996; Pflug, Dionne, Kaplan, Lynch & Djakiew, 1995;
Mandel, Ozdener, & Utermohlen, 2009). BDNF supports the health of existing cells, the growth
of new cells and the building of new synapses. It is found in areas related to higher order
learning, memory, and problem solving (e.g., hippocampus and cortex) (Hall, Thomas & Everitt,
2000). BDNF knock out mice have been shown to die at birth or have major neurological
difficulties, including sensory neuron loss (balance, hearing, and taste) and breathing problems
(Ernfors, Kucera, Lee, Loring, & Jaenisch, 1995). BDNF is implicated in multiple disease
processes including AD, depression, psychotic spectrum disorders, obsessive compulsive
26
disorder, dementia, anorexia, and bulimia. Under certain conditions it can increase cell death
instead of protect against it (secretion of p75NTR in the absence of Track A, B or C can lead to
cell death). BDNF plays a large role in neurogenesis (Bekinschtein et. al., 2008). It has a reward
salience effect in the ventral tegemental area. BDNF has been shown to be increased by exercise,
restricted calories, treatments for depression and intellectual stimulation (Gómez-Pinilla, Ying,
Roy, Molteni, & Edgerton, 2002). BDNF is implicated in the reversal of hippocampal damage
that occurs subsequent to depression treatment. Cortisol has been shown to reduce overall brain
levels of BDNF (Smith, Makino, Kvetnansky, & Post, 1995). Under or overregulation of BDNF
could have catastrophic effects on brain health. If BDNF were down-regulated in a high cortisol
context elevated it could be catastrophic.
Amygdala and Bed Nucleus: The amygdala is a well known area of the brain that acts like
a smoke detector for threats, stimulating the brain and body into a fight, flight or freeze defensive
response. The AMY plays a large role in two aspects of neuroplasticity: it controls cortisol
secretion and thus affects the metabolic ways a neuron can live or die, and it regulates salience
effects for threat. Hyper-sensitive, easily triggered stress response or high number of triggering
events coded by the AMY could lead to profound cell damage due to being in a metabolic
endangered state such that minor insults could kill them. The bed nucleus is connected to the
same areas of the brain as the AMY. It mediates long-term anxiety. Thus it is highly implicated
in worry or psychological stress and may be responsible for the slow attrition of neurons seen in
individuals who are low in a dominance hierarchy or have poor emotional regulation skills.
Upregulation of these areas could lead to increased excitoxic cell death and Under-regulation of
these areas could lead to reduced salience effects and negative consequences on multiple systems
(Sopolsky, 2005).
27
Activity Dependent - Previous Firing of Cells: Recent previous firing predisposes the area
to fire again under similar conditions (Antonov, Antonova, Kandel, & Hawkins, 2003). The cells
are thus primed to highlight certain relationships in external contexts by firing more easily in the
internal context. In a highly upregulated context of firing there is a loss of differentiation
between firing patterns. If firing becomes indiscriminant and not related to situational events it
could increased LTP, the encoding of inaccurate patterns of events and subsequently a
catastrophic destabilization of neural context. Under-regulation of firing rate could lead to
increased LTD due to inability for one part of a pattern of firing to trigger an action potential in
related systems.
Switching Neurological Sets: Another key aspect of the brain physiology that could
enhance the development of AD is the reduction of the ability to switch between functional
systems. Recent studies have found that there are major neurological organizational systems for
broad classes of types of life tasks (Taylor, Seminowicz, & Davis, 2009). Purely cognitive tasks
require different neurological demands than physical tasks or emotional learning tasks. A study
of individuals with early AD has found that there is a reduction of the ability to shift between
these larger scale functional systems. The insula, which often plays a gating function between
cognitive and emotional systems, no longer activates freely in those in early stage AD. A key
impact could be that mental states and memories that require the ability to shift between
functional systems could be difficult to access. This could lead a destabilization of neuronal
memory constellations and learned helplessness in accessing memories.
Attention and Signal Noise: Working memory has a limited amount of space for
processing information. Pain, psychological stress, and internal sensations could all co-opt
significant portions of working memory. This could lead to a feedback loop of increased focus
28
on the pain or stressor, causing more pain and stress, and so on. If the pain and stress are
occupying significant amounts of working memory space, concentration goes down, the
metabolic effects of stress go up, and the ability to attend to and learn from the environment goes
down. This could lead to lack of neurogenesis, loss of nerve cells and reduced dendritic density
through increased signal noise ratio.
Metabolic Effects
Apoptotic Regulatory Factors
Multiple alterations in indicators of apoptosis are seen in individuals with AD. Amiloid B
protein, thought to be a major factor in the formation of AD, has been shown to be neurotoxic
(Cotman & Anderson, 1995). Alterations in reactive oxygen have been noted in individuals with
AD. Reactive oxygen is a major chemical used by the immune system to induce apoptosis in the
body. T-lymphocytes, the cells responsible for this immune function, are down-regulated in
aging but upregulated in individuals with AD. This study finds “elevation of [Ca2+]i appears to
be a prerequisite for apoptosis, which is suggested to be involved in the neuronal death occurring
in AD. An increased [Ca2+]i in AD is consistent with processes leading to neurodegeneration in
AD” (Sulgera, Dumais-Hubera, Zerfassa, Henna & Aldenhoffb, 1998). Alterations in caspase-8
have been noted, “[a] role for caspase-8 and the receptor-mediated apoptotic pathway as a
mechanism leading to the activation of caspase-3 within neurons of the AD brain” (Rohna,
Headb, Nessea, Cotmanb and Cribbs, 2001). Alterations in the mitochondrial-produced AIF
(apoptic inducing factor) have been noted in individuals with AD.
Apoptic Catastrophe
There are multiple major paths that can lead to a catastrophic apoptic cascade in the
subsystems of the brain. One path leads to apoptic cascade through a metabolic upregulation in
29
cellular energy production (Choi, 2004). The second is through destabilization of cellular
connectivity through alterations in the homeostatic range of neuroplastic change. In this process
there are transformations in the brain’s potential to learn. This dysregulation could lead to
destabilization of a functional system or brain-wide, leading to loss of synapses, reduction of
neurogenesis and cell death. The third is through dysregulation in the process of cellular
reproduction, transcription, translation and the life cycle of a cell. The fourth is systemic
dysregulation of major homeostatic processes. In this process the destabilization of the
neurological environment is established by lack of vital chemicals, disruptions in autoregulatory
process (i.e., blood sugar, sleeping, metabolism, oxygenation, excretion, illness). The fifth is
dysregulation of the relationships between major psychoneurological functional areas, including
dysregulation of switching between functional systems, dissociation between functional areas in
a system, over-coupling between functional systems and increased irrelevant cueing for a
psychobiological functional area. The path to apoptic cascade includes multiple types of causal
events, including predispositions, synergistic events and non-linear dynamics.
There is an interplay in the brain between genetic determinants and contextual events that
sculpt its development (Thompson et. al., 2001; Caspi & Moffitt, 2006). The context of learning
is created by an algorithmic pattern of associative events that allows for the mapping of the
external realities, internal aptitudes and behavioral patterns in the biological relationship between
cells. This process is likely in part mapped genetically and determined by a self-organizing
process defined by the biological possibilities interacting with external events. Of note children
who face trauma, are raised under high stress or experience disruptions in parenting and have
differently organized cortical and sub-cortical areas (Schore, 2001).
Life-span Dependent Psychoneurochemical Changes
30
In older adults there is often a confluence of difficulties that could increase the likelihood
of apoptic cascade due to either metabolic factors or learning-based factors. As adults age there
is an change in the production of GABA (one of the brain’s main inhibitory neurotransmitters)
and a decrease of the regulatory inhibition of gabanergic neurons by adenosine triphosphate
(ATP) (Marczynski, 1998). The increase of inhibitory neurotransmitters could increase the
likelihood of raising the resting state of the brain to a point well below the threshold for action
potential creating significant difficulty for the encoding of a new patterns.
Reduction in sleep and sleep disturbances are common among elders (Huanga, Liua,
Wangb, van Somerenc, Xub, & Zhou, 2000). Lack of sleep in younger adults has been associated
with precipitous drop-offs in neurogenesis and thus a slower ability to replace cells when they
die (Mueller, Pollock, Lieblich, Epp, Galae, & Mistlberger, 2008). REM sleep is the time of the
most major daily increase in acytocholine, a chemical involved in the regulation of attention,
encoding memory and the rate of plasticity. REM sleep is significantly reduced as modern
Westerners age (Ehlersa & Kupfe, 1989).
Another key time period of susceptibility for women is when entering menopause
(Solertea, Fioravantia, Racchib, Trabucchic, Zanettib, & Govonid, 1999). Reduction in estrogen
has been associated with the increased chance of development of neurodegenerative disorders
and low levels of estrogen replacement were found to be a mild protective factor against AD in
initial research (Vegetoa, Benedusia, & Maggi, 2008; Solertea et. al., 1999).
Another key time of vulnerability is in infancy. During this time even seemingly minor
increases in stress can lead to marked changes patterns of stress reactivity and regulation apoptic
factors in the hippocampus throughout a lifetime (Liu et. al., 1997; Anand & Scalzo, 2000;
Weaver, Grant, & Meaney, 2002). Other likely times of disruptions are in times of rapid
31
neurological development, such as myolenation of the frontal lobe in late teens or major life
transitions.
Often older adults live in an impoverished learning context due to several reasons: a.
significant over-learning about relevant environmental events and thus a lack of novelty, b. lack
of novel environmental cues and reduction of mobility, c. diminished perceptual strength due to
changes in sensory input, d. poor exercise, and e. poor social support due to ageism in Western
societies. An impoverished learning context has been associated with loss of synaptic density and
cell loss in neural structures (Sandeman & Sandeman, 2000; Turnera & Greenough, 1985; van
Praag, Shubert, Zhao, & Gage, 2005). Assessing the individual for disruptions that happen at key
points of developmental transition could lead to effective interventions.
Systems Based Approach: Implications for the modeling of brain aging and treatment of AD
As we have seen before there are multiple ways that any system can become unstable.
Alzheimer’s disorder has many different antecedent events (Attix & Welsh, Bohmer; Chen,
Kagan, Hirakura, & Xie, 2000; Sorg et. al., 2007; Peskind, Wilkinson, Petrie, Schellenberg, &
Raskind, 2001). As one prominent researcher notes pithily, when you have seen one case of
Alzheimer’s, you have seen one case of Alzheimer’s (Whitehouse & George, 2008). In the
systems approach it is assumed that there are many possible ways that the brain and body could
produce what looks like Alzheimer’s disorder. Each of these ways of creating symptoms called
Alzheimer’s Dementia will have a specific conglomeration of factors. The role of this section
will be to outline how to use this approach in identifying key dysfunctions in the brain system
and to provide means to reregulate these processes.
The first step in this process would be to identify on a case-by-case basis which major
systems are dysregulated. This would require identifying possible disruptions that could lead to
32
the symptoms at hand. Some key areas of systemic disruptions we have identified are
dysregulation of CORT, disruptions to oscillation between parasympathetic and sympathetic
nervous system functioning, disruptions in plasticity, dysregulation of cellular senescence, and
dysregulation of dopamine or acytocholine systems. These factors could be exacerbated by
natural aging effects on the neural systemic contexts. Events such as changes in GABA
production and the phase transition from pre-menopausal to post-menopausal neurochemistry
could create synergistic effects with the above systems. These disruptions could be identified by
multiple types of tests such as diurnal cortisol tests, measuring for overall telomerase production,
assessing glutamate production, and assessing if GABA is presented in age-appropriate levels.
These and other findings could be assigned a statistical range of likely contribution to the disease
process (identified through studies and the existing literature), the additive effects, synergistic
effects and catalyzation points for phase change in the brain could be also be quantified. A good
multivariate analysis could assess the impacts, interactions and synergies and find threshold
levels for phase changes in the brain.
The second step would be to assess if there are global or local effects of the
dysregulation. This could be vital information for how to assess which systems are more likely to
be vulnerable and which systems are main drivers for the dysregulation.
The third step is to identify typically occurring protective defensive responses in these
systems and the steps and stages of return to systemic coherence after an insult. Of particular
interest are areas vulnerable to feedback loops. Often when a given system is taxed or is injured
it will enter an allostatic response as it attempts to mitigate the damage as it occurs and repair
damage that is existing. The ability of the tissue to instigate this pattern and then return to
baseline once the threat is gone are both vital to the health of the organism. As Dr. Kurr noted in
33
his work with the neural adapted sinibus virus, sometimes these defensive responses can
themselves trigger other protective responses and enter into a deadly feedback loop.
The fourth step is to identify what resources the system needs to meet the demands of
completing the protective response. Sapolsky, in a 2001 talk on Alzheimer’s disorder and over-
activation of cortisol, describes that cells, when stimulated by cortisol, can enter into an energy
crisis. The demands for energy in the hippocampal tissue exceed the tissue’s ability to meet those
demands. The dendritic connections are sloughed off and many protective measures are taken to
avoid the metabolic crisis that occurs if the cell is over-stimulated. Sapolsky (2001) goes onto to
state that if the cells are given an easy to digest form of glucose, thus providing enough energy to
complete the defensive measures and meet the demands of the cell, the crisis is averted and the
cells will not die.
The fifth step is to identify the secondary, tertiary, etc., systemic effects and note if any of
these effects will not re-regulate once the regulatory boundary is re-established for the system of
primary dysregulation (e.g., hippocampal tissue loss will be stopped and reversed through
reregulation of CORT; on the other hand, in another system the reregulation of telomerase will
not lead to reduction of stress-induced premature cellular senescence). The effects of a primary
dysregulation on other systems could be that situational stress leading to increased CORT
production is leading to: a. increasing the rate of cellular senescence; b. reduced cognitive
functioning that synergistically compounds these effects through enhancing poor eating habits, c.
increased anhadonia through disruption in the dopamine systems, leading to lack of motivation
and impoverished social environments; and d. increased aggression disrupting the ability the
primary social support system to provide regulation for stress reactivity.
34
The sixth step would be to identify means to work with the system to re-establish a
functional regulatory range. A key assessment in this is to identify when to use “global” or top
down interventions vs. when to use focused bottom up interventions. Bottom up interventions
address the regulation of the whole system by changing a single system, function, protein or
chemical. Top down interventions change the regulation in subsystems by re-regulating the
entire context of the body (e.g. increasing sleep and re-establishing sleep architecture can change
metabolism, neurogenesis, BDNF levels and work functioning etc.). In the case in which
dysregulation of CORT is the major driver for dysregulation building in the ability for the
nervous system to smoothly down-regulate through establishment of a parasympathetic braking
of the sympathetic system could be vital. This could be accomplished through multiple means.
Increased sleep and re-establishing healthy sleep architecture could lead to down-regulation of
the stress system. Increase in number and duration of pleasurable events could increase the
parasympathetic tone. Social engagement also increases parasympathetic tone. Many stress
reduction techniques found in CBT, MBSR and other protocols could reduce the stress response
(Koszyckia, Bengera, Shlika, & Bradwejna, 2007; Praissman, 2008). This could then leave the
system more able to form new memories and regrow the hippocampal tissue.
The seventh step is to assess the functioning of affected systems post re-regulation of the
primary dysregulated system. Any systems that do not re-establish their functional range may
need to have a process that addresses their functional dysregulation. Assessing in this case for
dysregulation and factors reducing the system’s ability to return to functional range will likely
allow the clinician to help this system return to functional capacity. It may in fact turn out that
the stress reactivity set in motion alterations that have made a stable phase shift. Finding means
to hold the regulatory boundaries for that system so that it can shift to the previous functional
35
relationship could then reduce the dysregulation in that system. As an example, if the CORT
secretion has reduced telomerase production to a level where oxidative stress can lead classes of
cells to premature senescence, reduction of CORT with likely not re-regulate this area (Serrano
& Blasco, 2002). Increases in factors leading neurogenesis, re-regulation of telomerase and
increased antioxidants could begin the process of interrupting the cascade of cell death that was
established by the dysregulation of cellular senescence. Increased REM sleep could help to
increase neurogenesis and re-regulate any disruptions to the acytocholine system established by
the elevations of CORT (Mirescu & Gould, 2006; Cameron & McKay, 1999; Mueller, Pollock,
Lieblich, Epp, Galae, & Mistlberger, 2008; Mohapela, Giampiero, Kokaiaa, & Lindvalla, 2005;
Vazquez & Baghdoyan, 2001).
It is important to note that there are many ways to affect any given system and the many
effects that any given system can have. This implies that the above formulation is only a guide
for possible interventions and modeling. If the main dysregulation was in neuroplasticity and not
CORT there would be multiple other types of interventions and the systems that are subsequently
affected would differ. Taking a systems approach to treatment and working with the health in the
system, it may be possible to reverse some of the loss of functioning and extend the health and
mental life for individuals with and without cognitive impairment till much closer to the end of
their life. This could enhance the quality of life across the lifespan.
This description of Alzheimer’s disorder is meant as an example of a broader class of
disorders of regulation and ways to map cases of AD that have other causal events leading to the
main dysregulation. Disorders of regulation are marked by changes in auto-regulation that can
lead to disruptions in the functioning of multiple systems in the body, brain and mind.
Alzheimer’s disorder, like other disorders of regulation, can have multiple events that lead to its
36
onset. These events lead to disruption in functional dynamics and destabilization of the system.
A complex functioning system like the human brain and body also has many processes that help
establish and maintain healthy systemic functioning. Just as this class of disorders can be created
by the body’s own mechanisms working out of synchronization, they likely can be healed
through re-establishment of the functional regulators of the biological system.
37
References
Adamafio, N. A. (2009). The hormone-emotion-behavioral gene-neuromessenger labyrinth:
Pertinent questions. African Journal of Biochemistry Research, 3(10), 326-331.
Adamec, R., & Young, B. (2000). Neuroplasticity in specific limbic system circuits may mediate
specific kindling induced changes in animal affect—implications for understanding
anxiety associated with epilepsy. Neuroscience Biobehaviral Review. 24(7), 705-23.
doi:10.1016/S0149-7634(00)00032-4.
Artola, A. (2008). Diabetes, stress, and ageing-related changes in synaptic plasticity in
hippocampus and neocortex - The same metaplastic process? European Journal of
Pharmacology. 585(1),153-62. doi:10.1016/j.ejphar.2007.11.084.
Attix, D., & Welsh-Bohmer, K. (2006) Geriatric Neuropsychology. New York, NY: Guilford
Press.
Azmitia1, E. C. (2001). Modern views on an ancient chemical: serotonin effects on cell
proliferation, maturation, and apoptosis. Brain Research Bulletin, 56(5), 413-424.
doi:10.1016/S0361-9230(01)00614-1
Anand, K. J. S., & Scalzo, F. M. (2000). Can Adverse Neonatal Experiences Alter Brain
Development and Subsequent Behavior? Biological Neonatology Fetal and Neonatal
Research, 77, 69-82. DOI: 10.1159/000014197
Bear, M. (2003). The changing brain, MIT WORLD http://mitworld.mit.edu/video/152
Beaumont, D., Lehmann A. B., & James, O. F. W. (1989). Protein Turnover in Malnourished
Elderly Subjects: The Effects of Refeeding. Oxford Journal of Medicine, 18(4), 235-240.
Becker, K. (2003). The common variants/multiple disease hypothesis of common complex
genetic disorders. Medical Hypothesis, 62(2), 309-317. ISSN: 0306-9877.
38
Bermudez, Y., Erasso, D., Johnson, N., Alfonso, M., Lowell, M., and Kruk, P. (2006).
Telomerase confers resistance to caspase-mediated apoptosis. Clinical Intervention
Aging, 1(2), 155–167. DOI 10.2147/CIA.S.
Berkeley, California, LINK to PDF:
http://docs.google.com/viewer?a=v&q=cache:Xccxp4gQcEQJ:www.er.doe.gov/bes/repor
ts/files/CS_rpt.pdf+Coherence+in+complex+systems&hl=en&gl=us&pid=bl&srcid=AD
GEESiDxzbmZac3kdV_hc4UmFDZXuZAK5kbR8Fl-
Wr5q89rZtuO4SAjxZvf7d4XlONL05ZpQsHE1uCo__2r_3fGBxBu2aRXTtedkFPCTv6q
YdAip-xjHZATR8qaiFPwHncKGYJYf9oM&sig=AHIEtbT-PLhHUBxrmx3-
pyZM2GOE5xsZFw
Blasco, M. A. (2005). Telomeres and human disease: aging, cancer, and beyond. Nature Review
Genetics, 6(8), 611-622. PMID 16136653.
Brett, M., Johnsrude, I. S., & Owen, A. M. (2002). Opinion: The problem of functional
localization in the human brain. Nature Reviews Neuroscience 3, 243-249.
doi:10.1038/nrn756.
Bremner, D., Narayan, M., Anderson, E., Staib, L., Miller, H., and Charney D., (2000).
Hippocampal Volume Reduction in Major Depression. American Journal of Psychiatry,
157, 115-118. PMID: 10618023.
Bruzz, P., Green, S., Byar. D., Brinton, L., & Schairer, C. (1985). Estimating the population
attributable risk factors using case-control data. American Journal of Epidemiology,
122(5), 904-914. PMID: 4050778.
Bourdeau, I., Bard, C., Noël, B., Leclerc, I., Cordeau, M., Bélair, M., Lesage, J., Lafontaine, L.,
and Lacroix, A. (2002). Loss of Brain Volume in Endogenous Cushing’s Syndrome and
39
Its Reversibility after Correction of Hypercortisolism. The Journal of Clinical
Endocrinology & Metabolism, 87(5), 1949-1954. PMID: 11994323.
Bullido, M. J., Artiga, M. J., María Recuero, M., Migue, I., García, A., Aldudo, J., Lendon, C.,
Han, S., Morris, M., Frank, A., Vázquez, J., Goate, A. & Valdivieso, F. (1998). A
polymorphism in the regulatory region of APOE associated with risk for Alzheimer's
dementia. Nature Genetics, 18, 69–71. doi:10.1038/ng0198-69.
Calabresi, P., Gubellini, P., Centonze, D., Picconi, B., Bernardi, G., Chergui, K., Svenningsson,
P., Fienberg, A., & Greengard, P. (2000). Dopamine and cAMP-Regulated
Phosphoprotein 32 kDa Controls Both Striatal Long-Term Depression and Long-Term
Potentiation, Opposing Forms of Synaptic Plasticity. The Journal of Neuroscience,
20(22), 8443-8451. PMID: 11069952.
Cameron, H. A., McKay, R. D. (1999). Restoring production of hippocampal neurons in old age.
Nature Neuroscience 2, 894-897. doi:10.1038/13197.
Casparya, D. M., Milbrandta, J. C. & Helfert, R. H. (1995). Central auditory aging: GABA
changes in the inferior colliculus. Experimental Gerontology, 30(3-4), 349-360.
doi:10.1016/0531-5565(94)00052-5.
Chang, S. (2004). Modeling aging and cancer in the telomerase knockout mouse. Mutation
Research/Fundamental and Molecular Mechanisms of Mutagenesis. 576(1-2), 39-53.
Chen, QS, Kagan, BL, Hirakura, Y, Xie, C. (2000). Impairment of hippocampal long-term
potentiation by Alzheimer amyloid beta-peptides. Journal of Neuroscience Research,
60(1), 65-72. PMID: 10723069.
40
Choia, J., Faucea S., & Effros, R. (2007). Reduced telomerase activity in human T lymphocytes
exposed to cortisol. Brain, Behavior, and Immunity, 22(4), 600–605. doi:
10.1016/j.bbi.2007.12.004.
Choi, D. (2004). Excitotoxic cell death. Journal of Neurobiology, 23(9), 1261–1276. PMID:
1361523.
Cohen, S., Graham, M., Lovrecz, G., Bache, N., Robinson, P., & Reddel, R. (2007). Protein
composition of catalytically active human telomerase from immortal cells. Science
315(5820), 1850–3. doi:10.1126/science.1138596.
Cohen, H., Kotlera, M., Matara, M., Kaplana, Z., Loewenthala, U., Miodownika, H., & Cassuto,
Y. (1998). Analysis of heart rate variability in posttraumatic stress disorder patients in
response to a trauma-related reminder. Biological Psychiatry, 44(10), 1054-1059. PMID:
9821570.
Cotman, C., and Anderson, A. (1995). A potential role for apoptosis in neurodegeneration and
Alzheimer's disease. Molecular Neurobiology, 10, 19-45. doi:10.1007/BF02740836.
Coyle, J. T., & Puttfarcken, P. (1993). Oxidative stress, glutamate, and neurodegenerative
disorders. Science, 262(5134), 689-695. doi:10.1126/science.7901908.
Davies, M. J., & Pomerance, A. (1972). Pathology of atrial fibrillation in man. British Heart
Journal, 34, 520-525. doi:10.1136/hrt.34.5.520.
de Diego-Otero, Y., Romero-Zerbo, Y., el Bekay, R., Decara, J., Sanchez, L., Rodriguez-de
Fonseca, F., & del Arco-Herrera, I. (2009). Alpha-tocopherol protects against oxidative
stress in the fragile X knockout mouse: an experimental therapeutic approach for the
Fmr1 deficiency. Neuropsychopharmacology, 34(4), 1011–26.
doi:10.1038/npp.2008.152.
41
Dorn, L., Burgess, E., Friedman, T., Dubbert, B., Gold, P., and Chrousos, G. (1997). The
Longitudinal Course of Psychopathology in Cushing’s Syndrome after Correction of
Hypercortisolism. Journal of Clinical Endocrinology, 82, 912-919. PMID: 9062506.
Epel, E., Blackburn, E., Lin, J., Dhabhar, F., Adler, N., Morrow, J., and Cawthon, R. (2004).
Accelerated telomere shortening in response to life stress. PNAS, 101(50). 17323-17324.
doi:10.1073/pnas.0408041101.
Felitti, V.J., Anda, R.F., Nordenberg, D., Williamson, D.F., Spitz, A.M., & Edwards, V. (1998).
The relationship of adult health status to childhood abuse and household dysfunction.
American Journal of Preventive Medicine, 14, 245-258. PMID: 9635069.
Falduto, M., Young, A., and Hickson, C. (1992). Exercise inhibits glucocorticoid-induced
glutamine synthetase expression in red skeletal muscles. AJP-Cell Physiology, 262(1),
C214-C220. PMID: 1346351.
Feig, C., Peter, M. (2007). How apoptosis got the immune system in shape. European Journal of
Immunology, 37(S1), S61 - S70. PMID: 17972347.
Geracioti, T., Baker, D., Ekhator, N., West, S., Hill, K., Bruce, A., Schmidt, D., Rounds-Kugler,
B., Yehuda, R., Keck, P., and Kasckow, J. (2001). CSF Norepinephrine Concentrations in
Posttraumatic Stress Disorder. American Journal of Psychiatry 158, 1227-1230.
Gerlach. J., Lublin, H., and Peacock, L. (1996). Extrapyramidal Symptoms during Long-Term
Treatment with Antipsychotics: Special Focus on Clozapine and D1 and D2 Dopamine
Antagonists. Neuropsychopharmacology, (14), 35S-39S. doi:10.1016/0893-
133X(95)00203-P.
Gorbunova, V., Seluanov, A., & Pereira-Smith, O. (2002). Expression of Human Telomerase
(hTERT) Does Not Prevent Stress-induced Senescence in Normal Human Fibroblasts but
42
Protects the Cells from Stress-induced Apoptosis and Necrosis. Journal of Biological
Chemestry. 277(41), 38540-9. PMID: 12140282.
Grenier, J., Tomkiewicz, C., Troussona, A., Rajkowskia, K., Schumachera, M., and Massaad, C.
(2004). Identification by microarray analysis of aspartate aminotransferase and glutamine
synthetase as glucocorticoid target genes in a mouse Schwann cell line. The Journal of
steroid biochemistry and molecular biology, 97(4), 342-52, PMID: 16182522.
Guzman-Marin, R., Suntsova, N., Methippara, M., Greiffenstein R., Szymusiak R., and McGinty
D. (2005). Sleep deprivation suppresses neurogenesis in the adult hippocampus of rats.
European Journal of Neuroscience, 22(8), 2111-2116. doi:10.1111/j.1460-
9568.2005.04376.x.
Harley, C., and Villeponteau, B., (1995). Telomeres and telomerase in aging and cancer: Geron
Corporation, 200 Constitution Drive. Current Opinion in Genetics & Development, 5(2),
249-255. doi:10.1016/0959-437X(95)80016-6.
Harley, C. B. (1996). Telomerase and cancer, Nature Reviews Cancer, 8(3), 167-179.
Hurley, M., Masha, D., & Jenner, P. (2001). Dopamine D1 receptor expression in human basal
ganglia and changes in Parkinson’s disease. Molecular Brain Research, 87(2), 271-279.
Hurley, M., Stubbs, C., Jenner, P. & Marsden, D. (1996). D3 receptor expression within the basal
ganglia is not affected by Parkinson's disease. Neuroscience Letters, 214(2-3), 23.5-78.
PMID: 8878087.
Idoyaga-Vargas, V., Abulafia, D., & Calandria, J. (2001). Correlation between cortisol level and
serotonin uptake in patients with chronic stress and depression. Cognitive, Affective, &
Behavioral Neuroscience, 1, 388-393. doi: 10.3758/CABN.1.4.388.
43
Janicki, S., and Monteiro, M. (1997). Increased Apoptosis Arising from Increased Expression of
the Alzheimer's Disease–associated Presenilin-2 Mutation (N141I). The Journal of Cell
Biology, 139(2), 485-495. PMID: 9334350.
Kannana, K., & Jain, S. (2000). Oxidative stress and apoptosis. Pathophysiology. 7(3), 153-163.
PMID: 10996508.
Kiel, D., Elliott. E. (1996). Chaos theory in the social sciences: foundations and applications.
University of Michegan Press.
Kirkwood, A., Rozas, C., Kirkwood, J., Perez, F., & Bear, M. (1999). Modulation of Long-Term
Synaptic Depression in Visual Cortex by Acetylcholine and Norepinephrine. The Journal
of Neuroscience, 19(5), 1599-1609. PMID: 10024347.
Kirschbaum, C., Wust, S., Faig, H. G., & Hellhammer, D. H. (1992). Heritability of cortisol
responses to human corticotropin-releasing hormone, ergometry, and psychological stress
in humans. Journal of Clinical Endocrinology & Metabolism, 75, 1526-1530. ISSN
0021-972X.
Koszyckia, D., Bengera, M., Shlika, J., & Bradwejna, J. (2007). Randomized trial of a
meditation-based stress reduction program and cognitive behavior therapy in generalized
social anxiety disorder. Behavior Research and Therapy, 45(10), 2518-2526.
doi:10.1016/j.brat.2007.04.011.
Whitehouse, P. J., & George, D., (2008). The Myth of Alzheimer's: What You Aren't Being Told
About Today's Most Dreaded Diagnosis. St. Martin's Press.
Lawrence, M., Redmond, D. E. Jr., Elsworth, J. D., Taylor, J. R., & Roth, R. H. (1991). The D1
receptor antagonist, SCH 23390, induces signs of parkinsonism in African green
monkeys. Life Sciences, 49(25), PL229-PL234. doi:10.1016/0024-3205(91)90299-Q.
44
Le Ray, D., De Sevilla, D. F., Porto, A. B., Fuenzalida, M., & Bun˜o, W. (2004). Heterosynaptic
Metaplastic Regulation of Synaptic Efffcacy in CA1 Pyramidal Neurons of Rat
Hippocampus. Published online in Wiley InterScience (www.interscience.wiley.com).
DOI 10.1002/hipo.20021.
Lidow, M. S., & Goldman-Rakic, P. S. (1994). A common action of clozapine, haloperidol, and
remoxipride on D1- and D2-dopaminergic receptors in the primate cerebral cortex.
PNAS, 91(10), 4353-4356. PMID: 8183912.
Linga, L. L., Hughes, L. F., & Caspary, D. M. (2005). Age-related loss of the GABA synthetic
enzyme glutamic acid decarboxylase in rat primary auditory cortex. Neuroscience,
132(4), 1103-1113. doi:10.1016/j.neuroscience.2004.12.043
Lisman, J., (2001). Three Ca2+ levels affect plasticity differently: the LTP zone, the LTD zone
and no man's land. Journal of Phisology, 532(2), 285.
Liu, D., Diorio, J., Tannenbaum, B., Caldji, C., Francis, D., Freedman, A., Sharma, S., Pearson,
D., Plotsky, P. M., Meaney, M. J. (1997). Maternal Care, Hippocampal Glucocorticoid
Receptors, and Hypothalamic-Pituitary-Adrenal Responses to Stress. Science, 277(5332),
1659–1662. doi:10.1126/science.277.5332.1659.
Lowe, S., and Lin, A. (2000). Apoptosis in cancer. Carcinogenesis, 21(3), 485-495. PMID:
10688869.
Lua, C., Fua, W., and Mattson, M. (2001). Telomerase protects developing neurons against DNA
damage-induced cell death. Developmental Brain Research, 131(1-2), 167-171.
doi:10.1016/S0165-3806(01)00237-1.
45
Lennon, S. V., Martin, S. J., Cotter, T. G. (1991). Dose-dependent induction of apoptosis in
human tumour cell lines by widely diverging stimuli. Cell Proliferation. 24(2), 203–14.
PMID: 2009322 .
Mattson, M. (2000). Apoptosis in neurodegenerative disorders. Nature reviews. Molecular cell
biology, 120(1), 120-129. PMID: 11253364.
Meshorer, E., and Soreq, H. (2008). mRNA Modulations in Stress and Aging: Handbook of
Neurochemistry and Molecular Neurobiology. New York, NY: Springer Science
DOI10.1007/978-0-387-32671-9.
Mirescu, C., & Gould, E., (2006) Stress and adult neurogenesis. Hippocampus 16, 233-238.
PMID: 16411244.
Mohapela, P., Giampiero, L., Kokaiaa, M., Lindvalla. O. (2005). Forebrain acetylcholine
regulates adult hippocampal neurogenesis and learning. Neurobiology of Aging, 26(6),
939-946. doi:10.1016/j.neurobiolaging.2004.07.015.
Mueller, A. D., Pollock, M. S., Lieblich, S. E., R. Epp, J. R., Galea, L. A. M., & Mistlberger, R.
E., (2007). Sleep deprivation can inhibit adult hippocampal neurogenesis independent of
adrenal stress hormones. American Journal of Physiology, Regulatory Integrative and
Comparative Physiology. 294(5), R1693-703.
Muir, J. (1911). My First Summer in the Sierra (Boston: Houghton Mifflin, 1911), on page 110
of the Sierra Club Books 1988 edition.
Nattel, S. (2002). Review article: new ideas about atrial fibrillation 50 years on. Nature 415, 219-
226. doi:10.1038/415219a
46
Naka, K., Akira, T., Ikeda, I., Motoyama, I. (2003). Stress-induced Premature Senescence in
hTERT-expressing Ataxia Telangiectasia Fibroblasts. The Journal of Biological
Chemistry, 279, 2030-2037. doi: 10.1074/jbc.M309457200.
Nichol, G., Hallstrom, A. P., Kerber, R., Moss, A. J., Ornato, J. P., Palmer, D., Riegel, B., Smith,
S., Weisfeldt, M. L. (1998). American Heart Association Report on the Second Public
Access Defibrillation Conference, April 17–19, 1997. Special Reports. American Heart
Association, Inc. 97:1309-1314.
Nilsson, J., von Euler, A., & Dalsgaard, C., (1985). Stimulation of connective tissue cell growth
by substance P and substance K, Nature 315, 61 – 63. doi:10.1038/315061a0.
Norman, J., (2010). The signs and symptoms which suggest the presence of high blood sugar and
diabetes. Pulled from the web on 3.21.10 from:
http://www.endocrineweb.com/diabetes/hyperglycemia.html
Okouchi, M., Ekshyyan, O., Maracine, M., & Yee, T. (2007). Apoptosis in Neurodegeneration.
Antioxidants & Redox Signaling. 9(8), 1059-1096. doi:10.1089/ars.2007.1511.
Ornish, D., Lin, J., Daubenmier, J., Weidner, Y., Epel, E., Kemp, C., Jesus, M., Magbanua, M.,
Marlin, R., Yglecias, L., Carroll, P., & Blackburn, E. (2008). Increased telomerase
activity and comprehensive lifestyle changes: a pilot study. The Lancet Oncology, 9(11).
1048-1057. PMID: 18799354.
Oswald, L., Wong, D., McCaul, M., Zhou, Y., Kuwabara, H., Choi, L., Brasic, J., & Wand, G.
(2005). Relationships Among Ventral Striatal Dopamine Release, Cortisol Secretion, and
Subjective Responses to Amphetamine. Neuropsychopharmacology 30, 821–832.
doi:10.1038/sj.npp.1300667.
47
Pacak, K., Palkovits, M., Kopin, I., & Goldstein, D. (1995) Stress-Induced Norepinephrine
Release in the Hypothalamic Paraventricular Nucleus and Pituitary-Adrenocortical and
Sympathoadrenal Activity: In Vivo Microdialysis Studies. Frontiers in
Neuroendocrinology. 16(2), 89-150. PMID: 7621982.
Pacaka, P., Tjurminab, O., Palkovitsc, M., Goldsteind, D., Kocha, C., Hofff, T., Chrousosa, G.
(2002). Chronic Hypercortisolemia Inhibits Dopamine Synthesis and Turnover in the
Nucleus accumbens: An in vivo Microdialysis Study Vol. 76, No. 3.Pacaka, P.,
Tjurminab, O., Palkovitsc, M., Goldsteind, D., Kocha, C., Hofff, T., Chrousosa, G.
(2002). Chronic Hypercortisolemia Inhibits Dopamine Synthesis and Turnover in the
Nucleus accumbens: An in vivo Microdialysis Study, Neuroendocrinology , 76(3), 148-
157. doi: 10.1159/000064522.
Panossiana, L., Porterb, V., Valenzuelaa, H., Zhua, X., Rebackb, E., Mastermanb, D.,
Cummingsb, J., Effrosac, R, (2003). Telomere shortening in T cells correlates with
Alzheimer’s disease status. Neurobiology of Ageing, 24(1), 77-84. PMID: 12493553.
Penza, K., Heim, C., and Nemeroff, C. (2003). Neurobiological effects of childhood abuse:
implications for the pathophysiology of depression and anxiety. Archives of Women's
Mental Health. 6(1), 15 - 22. doi:10.1007/s00737-002-0159-x
Peskind, E. R., Wilkinson, C. W., Petrie, E. C., Schellenberg, G.D., & Raskind, M. A.(2001).
Increased CSF cortisol in AD is a function of APOE genotype. Neurology. 56(8), 1094-8.
PMID: 11320185.
Popp, J., Schaper, K., Kölsch,H., Cvetanovska, G., Rommel, F., Klingmüller, D., Dodel, R.,
Wüllner, U., & Jessen, F. (2009). CSF cortisol in Alzheimer's disease and mild cognitive
impairment. Neurobiology of Aging, 30(3), 498-500. PMID: 17716786.
48
Praissman, S. (2008). Mindfulness-based stress reduction: A literature review and clinician’s
guide. Journal of the American Academy of Nurse Practitioners, 20(4), 212–216. DOI:
10.1111/j.1745-7599.2008.00306.x
Prigogine, I., Holte, J.,(1993). Chaos: the new science. University Press of America, Inc.
Lanham: Maryland.
Pryor W. A. (2000). Vitamin E and heart disease: basic science to clinical intervention trials.
Free Radical Biological Medicine, 28(1), 141–64. doi:10.1016/S0891-5849(99)00224-5
Rohn, Head, Nesse, Cotman, & Cribbs, (2001). Activation of Caspase-8 in the Alzheimer's
Disease Brain. Neurobiology of l Disease, 8(6), 1006-1016. PMID: 11741396.
Rudolph, K., Chang, S., Han-Woong, L., Blasco, M., Gottlieb, G., Greider, C., & DePinho, R.
(1999). Longevity, Stress Response, and Cancer in Aging Telomerase-Deficient Mice.
Cell Press, 96, 701–712. PMID: 10089885.
Schafer, F.Q., Buettner, G. R. (2001). Redox environment of the cell as viewed through the
redox state of the glutathione disulfide/glutathione couple. Free Radical Biological
Medicine, 30(11), 1191–212. doi:10.1016/S0891-5849(01)00480-4
Schaafsma, J. D., Balash, Y., Gurevich, T., Bartels, A. L., Hausdorff, J. M., & Giladi, N. (2003).
Characterization of freezing of gait subtypes and the response of each to levodopa in
Parkinson's disease. European Journal of Neurology, 10(4), 391 – 398.
doi:10.1046/j.1468-1331.2003.00611.x.
Schore, A. (2001). The effects of early relational trauma on right brain development, affect
regulation, and infant mental health. Infant Mental Health Journal, 22(1-2), 201–269.
doi:10.1002/1097-0355(200101/04)22:1<201::AID.
49
Serrano, M., & Blasco, M. (2001). Putting the stress on senescence. Current Opinion in Cell
Biology, 13(6-1), 748-753. PMID: 11698192.
Seuberta, W., Henninga, H. V., Schonera, W., & L'agea, M. (1968). Effects of cortisol on the
levels of metabolites and enzymes controlling glucose production from pyruvate.
Advances in Enzyme Regulation, 6, 153-187. doi:10.1016/0065-2571(68)90012-5.
Shank, C., Awschalom, D., Murphy, D., Bawendi, M., Stupp, S., Fréchet, J., Wolynes, P. (1999).
A Workshop on Complex Systems: Lawrence Berkeley National Laboratory
Shea, A., Walsh, C., MacMillan, H., and Steiner, M. (2004). Child maltreatment and HPA axis
dysregulation: relationship to major depressive disorder and post traumatic stress disorder
in females. Psychoneuroendocrinology, 30(2), 162-78. PMID: 15471614.
Shousea, M. & Ryan, W. (1984). Thalamic kindling: Electrical stimulation of the lateral
geniculate nucleus produces photosensitive grand mal seizures. Experimental Neurology,
86(1), 18-32. PMID: 6434340.
Starkman, M., Giordani, B., Gebarskic, S., Berent, S., Schork, A., Schteingart, D. (1999).
Decrease in cortisol reverses human hippocampal atrophy following treatment of
Cushing’s disease. Brain Research Reviews, 46(12), 1595-1602. ISSN:0006-3223.
Starkman, M., and Schteingart, D. (1981). Neuropsychiatric Manifestations of Patients With
Cushing's Syndrome: Relationship to Cortisol and Adrenocorticotropic Hormone Levels.
Archive of Internal Medicine 141, 215 - 219. PMID: 6257194.
Sohal, R. S. (2002). Role of oxidative stress and protein oxidation in the aging process. Free
Radical Biology Medicine, 33(1), 37–44. doi:10.1016/S0891-5849(02)00856-0.
50
Solertea, S., Fioravantia, M., Racchib, M., Trabucchic, M., Zanettib, O., & Govonid, S. (1999).
Menopause and estrogen deficiency as a risk factor in dementing illness: hypothesis on
the biological basis. Maturitas, 31(2), 95-101. ISSN:0378-5122.
Sorg, C., Riedl, V., Mühlau, M., Calhoun, V., Eichele, T., Läer, L., Drzezga, L., Förstl, H.,
Kurtz, A., Zimmer, C., & Wohlschläger, A. (2007). Selective changes of resting-state
networks in individuals at risk for Alzheimer's disease. PNAS, 104(47), 18760-18765.
doi: 10.1073/pnas.0708803104.
Sousaa, N., Cerqueira, J., & Osborne, A. (2008). Corticosteroid receptors and neuroplasticity.
Brain Research Reviews, 57(2), 561-570. PMID: 17692926.
Tang, Y., Yinghua Ma, Y., Wang, J., Fan, Y., Feng, S., Lu, Q., Yu, Q., Sui, D., Rothbart, M. K.,
Fan, M., & Posner. M. (2007). Short-term meditation training improves attention and
self-regulation. PNAS, 104(43), 17152-17156. doi: 10.1073/pnas.0707678104
Toussaint, O., Royer, V., Salmon, M., & Remacle, J. (2002). Stress-induced premature
senescence and tissue. Biochemical Pharmacology, 64(5-6), 1007-1009.
doi:10.1016/S0006-2952(02)01170-X.
Trump, B., Berezesky, I., Chang, S., Phelps, P. (1997). USA The Pathways of Cell Death:
Oncosis, Apoptosis, and Necrosis. Toxicologic Pathology, 25(1), 82-88. doi:
10.1177/019262339702500116.
Ulas, J., Weihmuller, L., Brunner, L., Joyce, J., Marshall, J., and Cotman, C. (1994). Selective
increase of NMDA-sensitive glutamate binding in the striatum of Parkinson's disease,
Alzheimer's disease, and mixed Parkinson's disease/Alzheimer's disease patients: an
autoradiographic study. Journal of Neuroscience, 14, 6317-6324. ISSN:0270-6474
51
Valko, M., Morris, H., & Cronin, M. T. (May 2005). Metals, toxicity and oxidative stress.
Current Medical Chemestry, 12 (10), 1161–208. doi:10.2174/0929867053764635. PMID
15892631.
Van Oersa, H., Kloetb, R., & Levinea, S. (1998). Early vs. late maternal deprivation
differentially alters the endocrine and hypothalamic responses to stress. Developmental
Brain Research 111(2), 245-252. doi:10.1016/S0165-3806(98)00143-6.
Vazquez, J. & Baghdoyan, H. A. (2001). Basal forebrain acetylcholine release during REM sleep
is significantly greater than during waking. American Journal of Physiological
Regulatory Integrative and Comparitive Physiology. 280, R598-R601. PMID:11208592.
Vegetoa, E., Benedusia, V., & Maggi, A. (2008). Estrogen anti-inflammatory activity in brain: A
therapeutic opportunity for menopause and neurodegenerative diseases. Frontiers in
Neuroendocrinology, 29(4), 507-519. doi:10.1016/j.yfrne.2008.04.001.
von Zglinicki, T., (2002). Oxidative stress shortens telomeres. Trends Biochememical Science,
27(7), 339-44. PMID:12114022.
Wall, J. T., Xu, J. & Wang. X. (2002). Human brain plasticity: an emerging view of the multiple
substrates and mechanisms that cause cortical changes and related sensory dysfunctions
after injuries of sensory inputs from the body. Brain Research Reviews, 39(2-3), 181-215.
doi:10.1016/S0165-0173(02)00192-3.
Wang, S., Oelze, B., & Axel Schumacher, A. (2008). Age-Specific Epigenetic Drift in Late-
Onset Alzheimer's Disease. PLoS ONE, 3(7). doi: 10.1371/journal.pone.0002698.
Weaver, I. C. G., Grant, R. J., & Meaney, M. J. (2002). Maternal behavior regulates long-term
hippocampal expression of BAX and apoptosis in the offspring. Journal of
Neurochemistry, 82(4), 998–1002. doi:10.1046/j.1471-4159.2002.01054.x
52
Weng, N., Levine, B., June, C., and Hodes, R. (1996). Regulated expression of telomerase
activity in human T lymphocyte development and activation. Journal of Experimental
Medicine, 183, 2471-2479. PMCID: PMC2192611.
Wüst, S., Federenkoa, I., Hellhammera, D., & Kirschbaumb, C. (2000). Genetic factors,
perceived chronic stress, and the free cortisol response to awakening.
Psychoneuroendocrinology, 25(7), 707-720. doi:10.1016/S0306-4530(00)00021-4.
Yehuda, R., Teicher, M. H., Seckl, J., Grossman, R., Morris, A., Bierer, L. (2007). Parental
Posttraumatic Stress Disorder as a Vulnerability Factor for Low Cortisol Trait in
Offspring of Holocaust Survivors. Archives of General Psychiatry, 64(9), 1040-1048.
PMID: 17768269.
Yehuda, R., Teicherbc, M., Trestmana, R., Levengooda, R., Sievera, L., (1996). Cortisol
regulation in posttraumatic stress disorder and major depression: A chronobiological
analysis. Biological Psychiatry, 40(2), 79-88. PMID: 8793040.
Yoshinoa, A., Hovda, D., Kawamata, T., Katayama, Y., & Beckera, D. (1991). Dynamic changes
in local cerebral glucose utilization following cerebral concussion in rats: evidence of a
hyper- and subsequent hypometabolic state. Brain Research, 561(1), 106-119. PMID:
1797338.
Young, E., Abelsona, J., Camerona, O. (2003). Effect of comorbid anxiety disorders on the
Hypothalamic-Pituitary-Adrenal axis response to a social stressor in major depression.
Biological Psychiatry, 56(2), 113-20. PMID: 15231443.
Zulli, R., Nicosia, F., Borroni, B., Agosti, C., Prometti, P., Donati, P., Vecchi, M., Romanelli,
G., Grassi, V., Padovani, A. (2005). Dispersion and Heart Rate Variability Abnormalities
53
in Alzheimer's Disease and in Mild Cognitive Impairment. Journal of the American
Geriatrics Society, 53(12), 2135 – 2139. doi:10.1111/j.1532-5415.2005.00508.x.

More Related Content

What's hot

HotStart poster. Presentation
HotStart poster. PresentationHotStart poster. Presentation
HotStart poster. Presentation
Jake Brown
 
THE FIRST SYSTEM OF REFERENCE FOR THE MEDICAL PRACTICE OF HOMEOPATHY IN FRANCE
THE FIRST SYSTEM OF REFERENCE FOR THE MEDICAL PRACTICE OF HOMEOPATHY IN FRANCETHE FIRST SYSTEM OF REFERENCE FOR THE MEDICAL PRACTICE OF HOMEOPATHY IN FRANCE
THE FIRST SYSTEM OF REFERENCE FOR THE MEDICAL PRACTICE OF HOMEOPATHY IN FRANCE
home
 

What's hot (17)

Dissertation
Dissertation Dissertation
Dissertation
 
Body Burden of Aluminum in Chronic Fatigue Syndrome
Body Burden of Aluminum in Chronic Fatigue Syndrome Body Burden of Aluminum in Chronic Fatigue Syndrome
Body Burden of Aluminum in Chronic Fatigue Syndrome
 
Amyloid precursor protein regulation of male sexual behaviour
Amyloid precursor protein regulation of male sexual behaviour Amyloid precursor protein regulation of male sexual behaviour
Amyloid precursor protein regulation of male sexual behaviour
 
The Science of Telomere / Telomeres in Anti Ageing
The Science of Telomere / Telomeres in Anti AgeingThe Science of Telomere / Telomeres in Anti Ageing
The Science of Telomere / Telomeres in Anti Ageing
 
Biological Basis of Aging
Biological Basis of AgingBiological Basis of Aging
Biological Basis of Aging
 
Aging- the biology of senescence
Aging- the biology of senescenceAging- the biology of senescence
Aging- the biology of senescence
 
Aging
Aging Aging
Aging
 
ASN Neuro-2016-Nikolakopoulou-
ASN Neuro-2016-Nikolakopoulou-ASN Neuro-2016-Nikolakopoulou-
ASN Neuro-2016-Nikolakopoulou-
 
Schematic diagram of transmission of acupuncture signals
Schematic diagram of transmission of acupuncture signalsSchematic diagram of transmission of acupuncture signals
Schematic diagram of transmission of acupuncture signals
 
HotStart poster. Presentation
HotStart poster. PresentationHotStart poster. Presentation
HotStart poster. Presentation
 
THE FIRST SYSTEM OF REFERENCE FOR THE MEDICAL PRACTICE OF HOMEOPATHY IN FRANCE
THE FIRST SYSTEM OF REFERENCE FOR THE MEDICAL PRACTICE OF HOMEOPATHY IN FRANCETHE FIRST SYSTEM OF REFERENCE FOR THE MEDICAL PRACTICE OF HOMEOPATHY IN FRANCE
THE FIRST SYSTEM OF REFERENCE FOR THE MEDICAL PRACTICE OF HOMEOPATHY IN FRANCE
 
Aging and model of aging
Aging and model of agingAging and model of aging
Aging and model of aging
 
biology of ageing
biology of ageing biology of ageing
biology of ageing
 
Aging and animal biotechnology
Aging and animal biotechnologyAging and animal biotechnology
Aging and animal biotechnology
 
Mechanism of action of the Body Wave
Mechanism of action of the Body WaveMechanism of action of the Body Wave
Mechanism of action of the Body Wave
 
Aging
AgingAging
Aging
 
Adam2017
Adam2017Adam2017
Adam2017
 

Similar to Dissorders of regulation towards a systemic based treatment of alzheimers disease

Hanipsych, stress
Hanipsych, stressHanipsych, stress
Hanipsych, stress
Hani Hamed
 
Pimentel_Research_Article_Sample
Pimentel_Research_Article_SamplePimentel_Research_Article_Sample
Pimentel_Research_Article_Sample
Alexander Pimentel
 
NEWS ABOUT TELOMERES
NEWS ABOUT TELOMERESNEWS ABOUT TELOMERES
NEWS ABOUT TELOMERES
jjc10
 
Psyc 395 Research Paper
Psyc 395 Research PaperPsyc 395 Research Paper
Psyc 395 Research Paper
Lexi Brenza
 
Theories of Aging - Part 1 in medicine and health sciences
Theories of Aging - Part 1 in medicine and health sciencesTheories of Aging - Part 1 in medicine and health sciences
Theories of Aging - Part 1 in medicine and health sciences
JackTan88
 
Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicine
Springer
 

Similar to Dissorders of regulation towards a systemic based treatment of alzheimers disease (20)

The Theory Of Aging - Ayu
The Theory Of Aging - AyuThe Theory Of Aging - Ayu
The Theory Of Aging - Ayu
 
Telomere length a 21st century biomarker
Telomere length  a 21st century biomarkerTelomere length  a 21st century biomarker
Telomere length a 21st century biomarker
 
Hanipsych, stress
Hanipsych, stressHanipsych, stress
Hanipsych, stress
 
Aging and the telomere connection dr. Jerry w. Shay - april 2012
Aging and the telomere connection   dr. Jerry w. Shay - april 2012Aging and the telomere connection   dr. Jerry w. Shay - april 2012
Aging and the telomere connection dr. Jerry w. Shay - april 2012
 
Biological theories of aging
Biological theories of agingBiological theories of aging
Biological theories of aging
 
Biology of ageing
Biology of ageingBiology of ageing
Biology of ageing
 
Pimentel_Research_Article_Sample
Pimentel_Research_Article_SamplePimentel_Research_Article_Sample
Pimentel_Research_Article_Sample
 
Psychological stress and ageing
Psychological stress and ageingPsychological stress and ageing
Psychological stress and ageing
 
NEWS ABOUT TELOMERES
NEWS ABOUT TELOMERESNEWS ABOUT TELOMERES
NEWS ABOUT TELOMERES
 
Psyc 395 Research Paper
Psyc 395 Research PaperPsyc 395 Research Paper
Psyc 395 Research Paper
 
From Selves to Cells: The New Biology of Cells, Illness and Depression
From Selves to Cells: The New Biology of Cells, Illness and DepressionFrom Selves to Cells: The New Biology of Cells, Illness and Depression
From Selves to Cells: The New Biology of Cells, Illness and Depression
 
ageing final.pptx
ageing final.pptxageing final.pptx
ageing final.pptx
 
Aging Medicine-1.pptx
Aging Medicine-1.pptxAging Medicine-1.pptx
Aging Medicine-1.pptx
 
Theories of Aging - Part 1 in medicine and health sciences
Theories of Aging - Part 1 in medicine and health sciencesTheories of Aging - Part 1 in medicine and health sciences
Theories of Aging - Part 1 in medicine and health sciences
 
Stress and Health
Stress and HealthStress and Health
Stress and Health
 
Cell Injury and Inflammation.pptx
Cell Injury and Inflammation.pptxCell Injury and Inflammation.pptx
Cell Injury and Inflammation.pptx
 
Basic of geriatrics and internal medicine for physiotherapist
Basic of geriatrics and internal medicine for physiotherapistBasic of geriatrics and internal medicine for physiotherapist
Basic of geriatrics and internal medicine for physiotherapist
 
Group 2 - cell cycle and chemotherapy
Group 2 - cell cycle and chemotherapyGroup 2 - cell cycle and chemotherapy
Group 2 - cell cycle and chemotherapy
 
Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicine
 
Als ppt conditions
Als ppt conditionsAls ppt conditions
Als ppt conditions
 

More from Michael Changaris

Primary Care and Behavioral Health Integration – Leveraging psychologists’ ro...
Primary Care and Behavioral Health Integration – Leveraging psychologists’ ro...Primary Care and Behavioral Health Integration – Leveraging psychologists’ ro...
Primary Care and Behavioral Health Integration – Leveraging psychologists’ ro...
Michael Changaris
 

More from Michael Changaris (20)

Primary Care and Behavioral Health Integration – Leveraging psychologists’ ro...
Primary Care and Behavioral Health Integration – Leveraging psychologists’ ro...Primary Care and Behavioral Health Integration – Leveraging psychologists’ ro...
Primary Care and Behavioral Health Integration – Leveraging psychologists’ ro...
 
Geriatric Pharmacotherapy Addressing SDOH and Reducing Disparities.pdf
Geriatric Pharmacotherapy Addressing SDOH and Reducing Disparities.pdfGeriatric Pharmacotherapy Addressing SDOH and Reducing Disparities.pdf
Geriatric Pharmacotherapy Addressing SDOH and Reducing Disparities.pdf
 
Health Psychology: Transforming social determinants of health to build health...
Health Psychology: Transforming social determinants of health to build health...Health Psychology: Transforming social determinants of health to build health...
Health Psychology: Transforming social determinants of health to build health...
 
Motivational Interviewing - interrupting Sustain Talk with Change Talk moving...
Motivational Interviewing - interrupting Sustain Talk with Change Talk moving...Motivational Interviewing - interrupting Sustain Talk with Change Talk moving...
Motivational Interviewing - interrupting Sustain Talk with Change Talk moving...
 
Motivational Interviewing: Change Talk moving to authentic wholeness (Lecture...
Motivational Interviewing: Change Talk moving to authentic wholeness (Lecture...Motivational Interviewing: Change Talk moving to authentic wholeness (Lecture...
Motivational Interviewing: Change Talk moving to authentic wholeness (Lecture...
 
Motivational Interviewing: Foundational Relationships for Building Change (Le...
Motivational Interviewing: Foundational Relationships for Building Change (Le...Motivational Interviewing: Foundational Relationships for Building Change (Le...
Motivational Interviewing: Foundational Relationships for Building Change (Le...
 
Motivational Interviewing: Introduction to Motivational Interviewing (Lecture...
Motivational Interviewing: Introduction to Motivational Interviewing (Lecture...Motivational Interviewing: Introduction to Motivational Interviewing (Lecture...
Motivational Interviewing: Introduction to Motivational Interviewing (Lecture...
 
Motivational Interviewing: Introduction to Motivational Interviewing (Lecture...
Motivational Interviewing: Introduction to Motivational Interviewing (Lecture...Motivational Interviewing: Introduction to Motivational Interviewing (Lecture...
Motivational Interviewing: Introduction to Motivational Interviewing (Lecture...
 
Motivational Interviewing: Engaging the Stages of Change (Lecture 8).pptx
Motivational Interviewing: Engaging the Stages of Change (Lecture  8).pptxMotivational Interviewing: Engaging the Stages of Change (Lecture  8).pptx
Motivational Interviewing: Engaging the Stages of Change (Lecture 8).pptx
 
Core CBT Skills for Self Evaluation
Core CBT Skills for Self EvaluationCore CBT Skills for Self Evaluation
Core CBT Skills for Self Evaluation
 
Grief, Loss and Transformation - Skills and Materials
Grief, Loss and Transformation - Skills and Materials Grief, Loss and Transformation - Skills and Materials
Grief, Loss and Transformation - Skills and Materials
 
Grief Group Slides Up Till May
Grief Group Slides Up Till MayGrief Group Slides Up Till May
Grief Group Slides Up Till May
 
Team Based Care for Hypertension Management a biopsychosocial approach
Team Based Care for Hypertension Management a biopsychosocial approachTeam Based Care for Hypertension Management a biopsychosocial approach
Team Based Care for Hypertension Management a biopsychosocial approach
 
Somatic Experiencing - Academic and Research References
Somatic Experiencing - Academic and Research References Somatic Experiencing - Academic and Research References
Somatic Experiencing - Academic and Research References
 
Slides for Living Well with Difficult Emotions Online Group
Slides for Living Well with Difficult Emotions Online GroupSlides for Living Well with Difficult Emotions Online Group
Slides for Living Well with Difficult Emotions Online Group
 
Understanding Bipolar Disorder: Biopsychosocial Approaches to Mind Body Health
Understanding Bipolar Disorder: Biopsychosocial Approaches to Mind Body HealthUnderstanding Bipolar Disorder: Biopsychosocial Approaches to Mind Body Health
Understanding Bipolar Disorder: Biopsychosocial Approaches to Mind Body Health
 
Lecture Today - Psychosis Introduction Neuropharm
Lecture Today - Psychosis Introduction NeuropharmLecture Today - Psychosis Introduction Neuropharm
Lecture Today - Psychosis Introduction Neuropharm
 
Seeking Safety Zoom Group Materials - Post-Traumatic Growth COVID-19
Seeking Safety Zoom Group Materials - Post-Traumatic Growth COVID-19Seeking Safety Zoom Group Materials - Post-Traumatic Growth COVID-19
Seeking Safety Zoom Group Materials - Post-Traumatic Growth COVID-19
 
PTSD Systems Treatment Pathway in Public Health Nursing and Primary Care
PTSD Systems Treatment Pathway in Public Health Nursing and Primary CarePTSD Systems Treatment Pathway in Public Health Nursing and Primary Care
PTSD Systems Treatment Pathway in Public Health Nursing and Primary Care
 
Integrated Primary Care Assessment SBIRT (Substance Use) and Mental and Refer...
Integrated Primary Care Assessment SBIRT (Substance Use) and Mental and Refer...Integrated Primary Care Assessment SBIRT (Substance Use) and Mental and Refer...
Integrated Primary Care Assessment SBIRT (Substance Use) and Mental and Refer...
 

Recently uploaded

Electrocardiogram (ECG) physiological basis .pdf
Electrocardiogram (ECG) physiological basis .pdfElectrocardiogram (ECG) physiological basis .pdf
Electrocardiogram (ECG) physiological basis .pdf
MedicoseAcademics
 
Premium Call Girls Nagpur {9xx000xx09} ❤️VVIP POOJA Call Girls in Nagpur Maha...
Premium Call Girls Nagpur {9xx000xx09} ❤️VVIP POOJA Call Girls in Nagpur Maha...Premium Call Girls Nagpur {9xx000xx09} ❤️VVIP POOJA Call Girls in Nagpur Maha...
Premium Call Girls Nagpur {9xx000xx09} ❤️VVIP POOJA Call Girls in Nagpur Maha...
Sheetaleventcompany
 
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan 087776558899
 
❤️Amritsar Escorts Service☎️9815674956☎️ Call Girl service in Amritsar☎️ Amri...
❤️Amritsar Escorts Service☎️9815674956☎️ Call Girl service in Amritsar☎️ Amri...❤️Amritsar Escorts Service☎️9815674956☎️ Call Girl service in Amritsar☎️ Amri...
❤️Amritsar Escorts Service☎️9815674956☎️ Call Girl service in Amritsar☎️ Amri...
Sheetaleventcompany
 
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan CytotecJual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
jualobat34
 
Premium Call Girls Dehradun {8854095900} ❤️VVIP ANJU Call Girls in Dehradun U...
Premium Call Girls Dehradun {8854095900} ❤️VVIP ANJU Call Girls in Dehradun U...Premium Call Girls Dehradun {8854095900} ❤️VVIP ANJU Call Girls in Dehradun U...
Premium Call Girls Dehradun {8854095900} ❤️VVIP ANJU Call Girls in Dehradun U...
Sheetaleventcompany
 
Call Girl In Indore 📞9235973566📞 Just📲 Call Inaaya Indore Call Girls Service ...
Call Girl In Indore 📞9235973566📞 Just📲 Call Inaaya Indore Call Girls Service ...Call Girl In Indore 📞9235973566📞 Just📲 Call Inaaya Indore Call Girls Service ...
Call Girl In Indore 📞9235973566📞 Just📲 Call Inaaya Indore Call Girls Service ...
Sheetaleventcompany
 
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
rajnisinghkjn
 
💚Call Girls In Amritsar 💯Anvi 📲🔝8725944379🔝Amritsar Call Girl No💰Advance Cash...
💚Call Girls In Amritsar 💯Anvi 📲🔝8725944379🔝Amritsar Call Girl No💰Advance Cash...💚Call Girls In Amritsar 💯Anvi 📲🔝8725944379🔝Amritsar Call Girl No💰Advance Cash...
💚Call Girls In Amritsar 💯Anvi 📲🔝8725944379🔝Amritsar Call Girl No💰Advance Cash...
Sheetaleventcompany
 
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Sheetaleventcompany
 
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
Sheetaleventcompany
 
Whitefield { Call Girl in Bangalore ₹7.5k Pick Up & Drop With Cash Payment 63...
Whitefield { Call Girl in Bangalore ₹7.5k Pick Up & Drop With Cash Payment 63...Whitefield { Call Girl in Bangalore ₹7.5k Pick Up & Drop With Cash Payment 63...
Whitefield { Call Girl in Bangalore ₹7.5k Pick Up & Drop With Cash Payment 63...
dishamehta3332
 

Recently uploaded (20)

Electrocardiogram (ECG) physiological basis .pdf
Electrocardiogram (ECG) physiological basis .pdfElectrocardiogram (ECG) physiological basis .pdf
Electrocardiogram (ECG) physiological basis .pdf
 
Premium Call Girls Nagpur {9xx000xx09} ❤️VVIP POOJA Call Girls in Nagpur Maha...
Premium Call Girls Nagpur {9xx000xx09} ❤️VVIP POOJA Call Girls in Nagpur Maha...Premium Call Girls Nagpur {9xx000xx09} ❤️VVIP POOJA Call Girls in Nagpur Maha...
Premium Call Girls Nagpur {9xx000xx09} ❤️VVIP POOJA Call Girls in Nagpur Maha...
 
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
 
Call 8250092165 Patna Call Girls ₹4.5k Cash Payment With Room Delivery
Call 8250092165 Patna Call Girls ₹4.5k Cash Payment With Room DeliveryCall 8250092165 Patna Call Girls ₹4.5k Cash Payment With Room Delivery
Call 8250092165 Patna Call Girls ₹4.5k Cash Payment With Room Delivery
 
Kolkata Call Girls Shobhabazar 💯Call Us 🔝 8005736733 🔝 💃 Top Class Call Gir...
Kolkata Call Girls Shobhabazar  💯Call Us 🔝 8005736733 🔝 💃  Top Class Call Gir...Kolkata Call Girls Shobhabazar  💯Call Us 🔝 8005736733 🔝 💃  Top Class Call Gir...
Kolkata Call Girls Shobhabazar 💯Call Us 🔝 8005736733 🔝 💃 Top Class Call Gir...
 
💰Call Girl In Bangalore☎️63788-78445💰 Call Girl service in Bangalore☎️Bangalo...
💰Call Girl In Bangalore☎️63788-78445💰 Call Girl service in Bangalore☎️Bangalo...💰Call Girl In Bangalore☎️63788-78445💰 Call Girl service in Bangalore☎️Bangalo...
💰Call Girl In Bangalore☎️63788-78445💰 Call Girl service in Bangalore☎️Bangalo...
 
ANATOMY AND PHYSIOLOGY OF REPRODUCTIVE SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF REPRODUCTIVE SYSTEM.pptxANATOMY AND PHYSIOLOGY OF REPRODUCTIVE SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF REPRODUCTIVE SYSTEM.pptx
 
❤️Amritsar Escorts Service☎️9815674956☎️ Call Girl service in Amritsar☎️ Amri...
❤️Amritsar Escorts Service☎️9815674956☎️ Call Girl service in Amritsar☎️ Amri...❤️Amritsar Escorts Service☎️9815674956☎️ Call Girl service in Amritsar☎️ Amri...
❤️Amritsar Escorts Service☎️9815674956☎️ Call Girl service in Amritsar☎️ Amri...
 
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan CytotecJual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
 
Premium Call Girls Dehradun {8854095900} ❤️VVIP ANJU Call Girls in Dehradun U...
Premium Call Girls Dehradun {8854095900} ❤️VVIP ANJU Call Girls in Dehradun U...Premium Call Girls Dehradun {8854095900} ❤️VVIP ANJU Call Girls in Dehradun U...
Premium Call Girls Dehradun {8854095900} ❤️VVIP ANJU Call Girls in Dehradun U...
 
Call Girl In Indore 📞9235973566📞 Just📲 Call Inaaya Indore Call Girls Service ...
Call Girl In Indore 📞9235973566📞 Just📲 Call Inaaya Indore Call Girls Service ...Call Girl In Indore 📞9235973566📞 Just📲 Call Inaaya Indore Call Girls Service ...
Call Girl In Indore 📞9235973566📞 Just📲 Call Inaaya Indore Call Girls Service ...
 
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
 
💚Call Girls In Amritsar 💯Anvi 📲🔝8725944379🔝Amritsar Call Girl No💰Advance Cash...
💚Call Girls In Amritsar 💯Anvi 📲🔝8725944379🔝Amritsar Call Girl No💰Advance Cash...💚Call Girls In Amritsar 💯Anvi 📲🔝8725944379🔝Amritsar Call Girl No💰Advance Cash...
💚Call Girls In Amritsar 💯Anvi 📲🔝8725944379🔝Amritsar Call Girl No💰Advance Cash...
 
Chandigarh Call Girls Service ❤️🍑 9809698092 👄🫦Independent Escort Service Cha...
Chandigarh Call Girls Service ❤️🍑 9809698092 👄🫦Independent Escort Service Cha...Chandigarh Call Girls Service ❤️🍑 9809698092 👄🫦Independent Escort Service Cha...
Chandigarh Call Girls Service ❤️🍑 9809698092 👄🫦Independent Escort Service Cha...
 
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
 
Kolkata Call Girls Naktala 💯Call Us 🔝 8005736733 🔝 💃 Top Class Call Girl Se...
Kolkata Call Girls Naktala  💯Call Us 🔝 8005736733 🔝 💃  Top Class Call Girl Se...Kolkata Call Girls Naktala  💯Call Us 🔝 8005736733 🔝 💃  Top Class Call Girl Se...
Kolkata Call Girls Naktala 💯Call Us 🔝 8005736733 🔝 💃 Top Class Call Girl Se...
 
tongue disease lecture Dr Assadawy legacy
tongue disease lecture Dr Assadawy legacytongue disease lecture Dr Assadawy legacy
tongue disease lecture Dr Assadawy legacy
 
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
 
❤️Call Girl Service In Chandigarh☎️9814379184☎️ Call Girl in Chandigarh☎️ Cha...
❤️Call Girl Service In Chandigarh☎️9814379184☎️ Call Girl in Chandigarh☎️ Cha...❤️Call Girl Service In Chandigarh☎️9814379184☎️ Call Girl in Chandigarh☎️ Cha...
❤️Call Girl Service In Chandigarh☎️9814379184☎️ Call Girl in Chandigarh☎️ Cha...
 
Whitefield { Call Girl in Bangalore ₹7.5k Pick Up & Drop With Cash Payment 63...
Whitefield { Call Girl in Bangalore ₹7.5k Pick Up & Drop With Cash Payment 63...Whitefield { Call Girl in Bangalore ₹7.5k Pick Up & Drop With Cash Payment 63...
Whitefield { Call Girl in Bangalore ₹7.5k Pick Up & Drop With Cash Payment 63...
 

Dissorders of regulation towards a systemic based treatment of alzheimers disease

  • 1. DISORDERS OF REGULATION: TOWARDS A MODEL OF SYSTEMIC BASED INDIVIDUALIZED TREATMENT FOR AUTOIMMUNE DISEASE, NEURODEGENRATIVE DISORDERS, AND CANCER
  • 2. 2 DISORDERS OF REGULATION: A SYSTEMS APPROACH The brain and the body work as a cohesive system. However, most theoretical understandings of disease processes do not take a systems-based approach to understanding the way that complex behavior emerges from these physical structures. Applying systems thinking to the problem diseases could yield novel models for disorders and treatment approaches. This paper will outline a theoretical basis for a class of disorders identified here as disorders of regulation, apply a systems-based approach to the modeling of these diseases and delineate some principals that could be used to create viable treatments. Some systems are capable of complex behavior that lasts for long periods of time. Some systems are not and degenerate into a form of repetitive order (Shank et al., 1999). An example of the latter in the brain is the kindling effects of grand mal seizure activity (Shousea & Ryan, 1984). In the kindling of a seizure the brain begins to synchronize its firing until an entire hemisphere fires at once. During a seizure the brain system becomes highly structured and organized, and the ability to produce complex human behavior is diminished. It is only within a coherent range of systemic functioning that complex behavior can emerge and be maintained. In other words, if the disruptions in the functioning of a system of the body or brain leads to a reduction of complex behavior or a disruption in the functional balance between systems, it can lead to disease and, in some cases, death. A complex system has points in its state space in which it is highly vulnerable to disintegrating into one of the aforementioned patterns of loss of systemic complexity (Prigogine & Holte, 1993). The balanced oscillation between systems forms a coherent pattern of relationship. If that coherent pattern of relationships is disrupted in minor ways, the system will display small perturbations in functioning until it can right itself (Shank et al., 1999). If that
  • 3. 3 system exceeds its regulatory capacity at a key point for long enough or with sufficient intensity, the entire system adapts, forming a new functional relationship between systems. The behavior of many disease processes appears to be created and maintained by disruptions in the functional relationship between systems. At times it is the body’s own processes that are involved in the genesis and maintenance of these syndromes (D. Kerr, personal communication, August 08, 2008). Some disorders that appear to be effected by this type of disease process are autoimmune disorders, many types of cancers, and neurodegenerative disorders. The causes for each of these disorders are highly varied, as will be the treatments postulated; however, these disorders follow a similar pathway of disruptions in the functioning of systems. This similar pathway can be a guide for the creation of appropriate and effective clinical interventions. Evidence that Dysregulation Leads to Pathology There are many data points supporting the postulation that dysregulation leads to pathological functioning in the body, brain and mind. Multiple studies have shown that over- secretion of cortisol in both Cushing’s syndrome and depression leads to reduction in the cellular density and dendritic connection in the hippocampus (Bourdeau et al., 2002; Bremner, Narayan, Anderson, Staib, Miller, and Charney, 2000; Starkman and Schteingart, 1981; Dorn, Burgess, Friedman, Dubbert, Gold, and Chrousos, 1997). Cortisol dysregulation in many leads to loss of sleep. Loss of sleep in turn can lead to a reduction of neurogenesis in the hippocampus (Guzman- Marin et al., 2005). Overproduction of insulin leads to symptoms including mood swings, weight gain, hypoglycemia, increased facial hair in women, hair loss, bloating, and high blood pressure (Norman, 2010). Underproduction of dopamine can lead to movement disorders, dementia, alterations in working memory and symptoms of psychosis. Overproduction of Telomerase has
  • 4. 4 been associated with development of cancerous tumors (Harley and Villeponteau, 2002). Overproduction of glutamate can lead to cell death; elevated levels of glutamate are found in dementia, Alzheimer’s type (Choi, 2004; Ulas et al., 1994). Telomerase: A Regulator of Cellular Life Span Cells in the body are created, maintained for some time and then die. Different regions of the body and brain require different rhythms of cellular lifespans. In the span of one week most pancreatic cells are replaced by other cells; over of the span of several years all the cells in the body are replaced by other cells. This rhythm of cell loss and replacement requires a consistent cycle of cell death, cell birth and cell maintenance. If these key functions begin to occur either too frequently or too rarely, pathology can develop (Lowe & Lin, 2000). This exemplifies the nature of disorders of regulation. They are disorders where the balance between the functional demands of a system is disrupted to the point that other systems relying on that system and the body as a whole no longer function effectively. Telomerase is one of the key regulators of cellular life span (Gorbunova, Seluanov, & Pereira-Smith, 2002).Telomeres are short, compacted segments of DNA forming a cap at the end of the chromosomes (Gorbunova, Seluanov, & Pereira-Smith, 2002). As the cell goes through each cycle of mitosis the telomere is shortened (Blasco, 2005). These segments of the chromosomal DNA (telomeres) function as regulators for cell division. As they shorten past a threshold point the cell can no longer divide and produce other cells (Blasco, 2005). Many tissues require more cell divisions than the original telomere length allows. In order to maintain physical health and systemic regulation, an enzyme called telomerase is produced (Gorbunova, Seluanov, & Pereira-Smith, 2002). This enzyme stimulates the rebuilding the telomere end cap of the chromosomes. The end cap is rebuilt thus extending the ability of cells to divide and
  • 5. 5 replace cells at the end of their life span (Blasco, 2005). Dysregulation of telomerase. A hallmark of disorders of regulation is that there are many possible events that can cue any single disruption in the functioning of a system. These disruptions lead to major systemic functions occurring in dysynchronous manner. The dyssynchronous functioning of one system can lead to large-scale dysfunctions in other systems. Dyssynchronous systemic functioning can lead to what appears as symptoms of disease. There are multiple events and classes of events that can lead to dysregulation of telomerase production, as there are multiple ways that telomerase production can be altered (Epel et al., 2004). Some of these include genetic differences (e.g., disruptions in the Ras and Raf genes), autonomic stress, lifestyle choices, mental health and chemical toxins, to name a few (Lua, Fua, and Mattson, 2001; von Zglinicki, 2002; Epel et al., 2004). The effects of these events can act in isolation or in some cases synergistically to alter the functioning of telomerase, if an individual has a genetic predeterminent for dysregulation of telomerase that may or may not be sufficient to create major disruption in the tissues of the body. If, however, that genetic determinent occurs along with other factors there could be a catalyzing event leading to large scale disruptions in tissues and thus disease processes. In one study of a telomerase deficient mouse, there were not global increases in symptoms of aging (Chang, 2004). However, the mice displayed reduced ability to repair injuries or recover from illnesses, a shortened lifespan, and increased incidence of cancerous tumors (Serrano & Blasco, 2001). Thus leaving them vulnerable to multiple disease processes. Lifestyle and telomerase production. A recent study implied that lifestyle changes such as alterations of diet, exercise, and stress levels have a significant association with re-regulation in telomerase activity (Ornish et al.,
  • 6. 6 2008). The study notes that causation cannot be inferred and that a more comprehensive randomized study is warranted from these results (Ornish et al., 2008). Comprehensive lifestyle changes significantly increase telomerase activity and consequently telomere maintenance capacity in human immune-system cells. Given this finding and the pilot nature of this study, we report these increases in telomerase activity as a significant association rather than inferring causation. (Ornish et al., 2008) Stress and telomerase. Telomerase has an interaction effect with the autonomic stress response (Choia, Faucea, & Effros, 2007; Epel et al. 2004). High levels of autonomic stress and cortisol have been associated with down-regulation of telomerase and shorter telomeres (Choia, Faucea, & Effros, 2007; Epel et al., 2004). Allostatic load is the term coined by Bruce McEwen to describe amount of energy needed to return a system to homeostasis. Long-term exposure to stress hormones due to prolonged stressors or a very large stress response leads to a higher allostatic load (McEwen, 2002). Telomerase production is one of the systems affected by overproduction of stress hormones (Choia, Faucea, & Effros, 2007). Several studies have noted that changes in telomere length occurred in response psychological stress and mood disorders. T lymphocytes exposed to high levels of cortisol have displayed a significant reduction of telomerase activity during both primary and secondary stimulation of cells (Choia, Faucea, & Effros, 2007; Epel et al., 2004). Dysregulation of telomerase: neurodegenerative disorders. The pattern of disorders of regulation, starting as a disruption in one system and leading to another, can be seen in the effects of prolonged autonomic stress response (Epel et al., 2004;
  • 7. 7 McEwen, 2002). It is likely that cortisol levels produce a heightened risk environment that sets the stage for other factors to catalyze into either a cancer or neurodegenerative disorder (R. Sapolsky, personal communication, 2001). Recent studies have shown that TERT is present in pre-differentiated neuronal blast cells but it precipitously drops off in adult neurons. Deficits of TERT present in the cellular context have been shown to be predictive of the development of neurodegenerative disorders. The presence of TERT is protective against the cell death due to apoptotic factors and the onset of apoptotic cascade (Bermudez, Erasso, Johnson, Alfonso, Lowell, & Kruk, 2006; Lua, Fua, & Mattson, 2001). We found that expression of hTERT, the catalytic component of telomerase, was sufficient and specific to reduce caspase-mediated cellular apoptosis. Further, hTERT expression reduced activation of caspases 3, 8, and 9, reduced expression of pro-apoptotic mitochondrial proteins t-BID, BAD, and BAX and increased expression of the anti-apoptotic mitochondrial protein, Bcl-2. The ability of telomerase to suppress caspase-mediated apoptosis was p-jnk dependent since abrogation of jnk expression with jip abolished resistance to apoptosis. (Bermudez, Erasso, Johnson, Alfonso, Lowell, & Kruk, 2006). Increased cell death is a key aspect of multiple neurodegenerative disorders (Okouchi, Ekshyyan, Maracine, & Yee, 2007). Down-regulation of telomerase is present in individuals with Alzheimer’s dementia (AD), Parkinson’s dementia, Amyotrophic lateral sclerosis and Fronto- temporal dementia, to name a few. In AD shortened telomere length was noted in T- lymphocytes. This reduced telomere length correlated with scores on MMSE: “the
  • 8. 8 proinflammatory cytokine TNFα (a clinical marker of disease status), with the proportion of CD8+ T cells lacking expression of the CD28 costimulatory molecule, and with apoptosis. (Panossian et. al, 2003).” Another study finds similarly, “Thus, a simple count of chromosome ends for the ‘presence/ absence’ of fluorescence (marking telomerase) may provide a valid biomarker of dementia status” in individuals who meet the criteria for AD (Jenkins, 2008). Alterations in the “SOD1 gene [and] deletions of the telomeric copy of the SMN gene” were noted in individuals with motor neuron disease (Orrell & Figlewicz, 2001). An upregulation of telomerase has been shown to be neuroprotective and to reduce the chance of an apoptotic cascade (Mattson, 2000). There are many factors that increase or reduce the risk of apoptosis (Mattson, 2000). Some recent studies have indicated that telomerase is important to responses to insults to the brain as well as neural development (Mattson, 2000; Lua, Fua, and Mattson, 2001). The regulation of apoptosis is a key role of the immune system (Feig & Peter, 2007). The disruption in immune functioning due to reduced telomere length is a key example of how a disruption in one system’s functioning can lead to the disruptions in another (Rudolph, Chang, Han-Woong, Blasco, Gottlieb, Greider, and DePinho, 1999). The under-regulation of telomerase increases susceptibility to apoptosis and correlates with dementia ratings and increased inflammation, possibly linking alterations in immune functioning and neurodegenerative disorders (Lua, Fua, and Mattson, 2001). Life in the balance: The effects of telomerase an oxidative stress on cellular senescence. Oxidative stress has been shown to reduce the length of telomeres and is not repaired as easily as damage to other areas of the DNA (von Zglinicki, 2002). Oxidative stress has been shown to play a key role for enchained cellular senescence and antioxidants have been shown to decelerate cellular senescence (von Zglinicki, 2002; Naka, Akira, Ikeda, & Motoyama, 2003).
  • 9. 9 There is an interaction between regulation of oxidative stress and the regulation of replicative processes (von Zglinicki, 2002). One study has found that increased anti-oxidant protection plays a key role in the ability of embryonic stem cells to remain pluripotent even after multiple mitotic cycles (von Zglinicki, 2002; Naka, Akira, Ikeda, & Motoyama, 2003). This also points to the interrelationships between systems. Upregulation of telomerase can reduce the effects of oxidative stress and stop a cell from entering senescence, even in an oxidative upregulated context (von Zglinicki, 2002; Naka, Akira, Ikeda, & Motoyama, 2003). Upregulation of telomerase can also play a key role leading to immortalization of cells and the onset of a cancerous replication cycle. One could easily imagine that a balance is established between oxidative stress and telomerase that, if exceeded in either direction, could cause significant disruptions to a single tissue/system or at a more global, body-wide level. Systemic stress. Systemic stress is a term this author uses to describe when the change to any one system in the body enters into a state that exceeds its normal range of ability to return to a baseline of functioning thus requiring increased energy or adaptations in other systems in order to return to baseline. An excellent example of how certain types of systemic stress can be virtually irreversible is the way cells can enter into stress-induced senescence. Senescence is the lack of ability for a cell to continue to reproduce through mitosis. When cells are “exposed to sublethal (systemic) stress” they will often enter what is known as stress-induced cellular senescence (SIPS) (Naka, Akira, Ikeda, & Motoyama, 2003). Like with most disorders of regulation SIPS can be triggered by multiple means: exposure to UV light, radiation, oxidative stress and other external insults that damage the length of the telomere. These cells display key markers of cellular senescence, such as flattening of the cell body, β-galactosidase activity, and a rapid
  • 10. 10 reduction in telomere length. Overproduction of reactive oxygen can induce cellular senescence (Naka, Akira, Ikeda, & Motoyama, 2003). The cellular senescence brought on by oxidative stress cannot be reversed with an upregulation of telomerase. Similar findings exist for damage due to other factors such as radiation. This indicates that global DNA damage also can induce premature cellular senescence (Naka, Akira, Ikeda, & Motoyama, 2003). The oncogenes Ras and Raf also trigger what appears to be stress-induced cellular senescence, resulting in a permanent arrest to the cycle of mitotic replication. This is known as ontogenetic stress-induced senescence (Naka, Akira, Ikeda, & Motoyama, 2003). Global and local effects. Adding to the complexity are the possible effects of global systems that set the tone of multiple systems or in some cases the entire body. When there is a global disruption it can lead to many symptoms that appear unrelated and disconnected from a single cause. Some of these global systems could include diurnal patterns of endocrine and neurotransmitter production, sleep cycles, autonomic stress reactivity and so on. As stated above, disorders of regulation is that any single system’s functioning can be affected by many different events. These events can be localized to a system or an area of tissue, such as the shortening of telomeres when oxidative stress is upregulated in a specific region, and these events can also be global, such as with broad scale damage to the DNA structure from radiation that leads to a cell entering senescence early and losing the ability to replicate (von Zglinicki, 2002; Naka, Akira, Ikeda, & Motoyama, 2003). To exemplify this overproduction of insulin effects the entire body including mood swings, weight gain, hypoglycemia, increased facial hair in women, hair loss, bloating, and high blood pressure (Norman, 2010).
  • 11. 11 Complexity small changes big results. As small differences happen in the replication process they can lead to large-scale alterations in system functions (Briggs & Peat, 1989). This type of difficulty has a particular sensitivity to initial conditions. Cellular senescence highlights this type of change. Multiple replications of cells and the process of cell reproduction are ripe for dysregulation that mirrors the dysregulations that are possible in the population growth equation. Second Impact Syndrome When individuals have a head injury there are profound alterations in the internal working and chemical dynamics of the brain (Yoshinoa, Hovda, Kawamata, Katayama, & Beckera, 1991). These shifts in metabolic and chemical functioning are intended to protect the brain and allow it to heal after a concussive injury (Giza & Hovda, 2001). Medicine, even the body’s own medicine in sufficient dosages, can cause damage. If certain aspects of the brain’s functioning exceed its regulatory capacity, there can be a major loss of brain tissue and functioning from what seems like insignificant insults to the brain (Giza & Hovda, 2001). This process of an initial dysregulation of brain metabolism leading to vulnerable states from which even a minor insult (e.g., small impact to the skull) leads to significant brain damage is an apt example of the process of systemic dysregulation leading to a disease process. The functional dysregulation model of disorders of regulation would hold that as a system exceeds its regulatory capacity in one area, the entire system begins to adapt. It also holds that there are key areas, times or states of vulnerability from which exceeding the regulatory capacity would lead to fundamental alterations in the systemic function. These alterations occur more frequently in two situations: 1. When the body’s defensive strategies put the system into systemic stress (high allostatic load on the system) or 2. The dysregulation of one system
  • 12. 12 precludes the effective functioning of another, thus leading to that system no longer functioning as a regulatory boundary for the dysregulated system and the non-functioning system no longer being able to perform its vital function, leading to more systemic adaptations and so on. In McEwen’s (2002) theory of allostatic load alterations in stress response, either in intensity or duration, the context leads to systemic adaptation. In this model I would extend the idea of allostatic load to any significant functional adaptation of a systemic relationship due to the inability to return to a functional baseline of oscillatory patterns between systems.“It is during the post-injury period, when cellular metabolism is stretched to its limits, that the cell (and the brain) is most vulnerable to further insults (Giza & Hovda, 2001).” Multiple Causes – Multiple Systems There are multiple systems in the body. Each system has multiple contextual events that maintain its functioning. Disruption to a system can come from any surrounding system. Some systems have global reach and can affect the entire organism at once. This adds a significant layer of complexity in understanding the antecedents of symptoms. To put this in more concrete terms, a neuron will die under many conditions (Trump, Berezesky, Chang, & Phelps, 1997). Some of these include increased metabolism, excessive glutamate production, apoptosis inducing factors, genetic abnormalities, epigenetic mutations, exceeding its number of mitotic cycles, oxidative stress, being attacked by t-cells and so on. To quote John Muir, "When we try to pick out anything by itself, we find it hitched to everything else in the Universe.” In the case of these disorders, this is true. However, the goal is to identify the corner being tugged on and stop the tug. The aforementioned list contains some of the systems affecting a neuron’s life. There are countless other systems.
  • 13. 13 Implications for treatment The difficulty in addressing the symptoms of disorders of dysregulation stems from: a. there being multiple possible causes for a single outcome, b. second order effects where the primary cause is not the main cause for presenting symptoms, c. the symptoms are created by adaptations in the functioning of the body’s own processes, d. there can often be additive effects, and e. small disruptions can become larger over many iterations, such as through mitosis, across a life span, thus making effect and causal events not apparently contiguous in time. In this model creating a treatment is much more precise and therefore labor intensive than in traditional treatments. The goal of treatment is not amelioration of symptoms but the re- establishment of a coherent relationship between systems. This requires: a. identifying the areas of dysregulation, b. identifying the main functions and regulators of these areas, c. in some cases differentiating primary, secondary and tertiary symptoms, and d. interventions aimed at re- establishing the body’s natural ability to return to a coherent allostatic range. Current Treatments Using Systems Approaches to Treat Disorders of Regulation There are many current treatments that utilize this type of reasoning to reestablish a homeostatic pattern or to help a dysregulated system find a new stable pattern of systemic oscillation. Some of these are: defibrillation as a treatment for certain classes of myocardial infarction, deep brain electric stimulation for movement based Parkinsonian symptoms, motor neuron atrophy due to encephalitis infection, cognitive behavioral therapy for depression, saline trigger point injections for pain, and mindfulness-based stress reduction. Exploring existing treatments and appling systems thinking to understanding patterns of dysregulation explanatory model of how a treatment works to re-establish a coherent pattern of functioning could illuminate
  • 14. 14 this further. Neural Adapted Sinibus Virus (NSV) Neuroadapted Sindbis virus (NSV) in humans is a moderate lung infection; in mice it leads to a severe sickness that if it infects the brains of animals will cause paralysis, a stripping of dendritic connections, global excitotoxic nerve death and eventually the death of the animal. One of the key findings of the study of this infection is that the viral infection is not the direct cause of these terrible effects (D. Kerr, personal communication, 2008). The animal will become quite sick but it is not the infection of the cells that leads to the cell death. The virus it self leads to the death of only around 20% of the neurons while at the end of three weeks the animal displays 95% neuron loss. Douglas Kerr (2008) and his research group found that it is dysregulation of autoregulatory functions of the microglial and a metabolic protective defensive strategy in the neurons that leads to the catastrophic loss of cells. It is a dysregulation of the re-uptake of glutamate and an increased neuron signaling of stress through the secretion of nNOS that leads paralysis and death. What the researchers did not know at first was that this upregulation of nNOS and the dendritic sloughing was a protective strategy that is vital for a cell near metabolic overload. The secretion of nNOS signals to the neuron it is in danger of excytotoxicity and evokes a protective strategy which is to reduce input. If the dendrites coming into the neuron continue to signal it to fire, it will enter a metabolic crisis and die; thus, dropping the dendritic connections protects the neuron against over-excitation, attempting to be a shutoff switch if the neuron in colloquial speech, “overheats”. However, because this virus leads to global upregulating of nNOS (rather then local upregulation more normative for axotomy injuries) and the sloughing of dendritic connections in the entire motor cortex the system is primed another form of paralysis and of cell death induced
  • 15. 15 by isolation from other cells (D. Kerr, personal communication, 2008). Indeed when the researchers down regulated nNOS and TNFa-alpha, what they found was that the dendrites remained connected but the cells died due excytotoxitcity. The sloughing of dendrites is protective function that if it happens on local level likely can be protective. If upregulation of nNOS happens on a global level it exceed the brains capacity to regrow connections after the need for the initial protective response has passed. Researchers working in systemic manor were able to halt this process by providing other forms of protective interventions for the time when the cells were in a vulnerable state. From the systems perspective there are five key factors in this process. The first is that there were alterations in the current system state due to a viral infection leading to priming effects for a catastrophic cell loss. The second is that researchers identified the system areas where the system entered a state space vulnerable to produce the results noted (e.g. finding events that could cause dendritic sloughing). The third is that there were synergistic interactions between multiple systems defensive responses that lead to disruptions in the ability of the state space to return to homeostatic range after an allostatic protective response. Fourth is that the team identified several systems and their defensive actions. In other words they identified the key protective functions that are leading to the neuron entering into an allostatic response. Fifth the team found ways to hold the system stable while time dependant defensive strategies could complete and reduced the signaling for other defensive strategies that lead to the synergistic catastrophic destabilizations of the spinal motor neuron functioning. Due to these contextually relevant interventions during the critical period the normal and typically quite stable functioning of the motor neuron system was allowed to reassert itself through re-establishing the regulatory boundaries already present in the system (D. Kerr, personal communication, 2008).
  • 16. 16 Applying a systemic approach to understanding the formation of symptoms of Alzheimer’s Dementia: Role of Systemic Dysregulation in Alzheimer’s Dementia AD, or Dementia of the Alzheimer’s type, has multiple precipitating events that lead to the development of this condition (Attix & Welsh-Bohmer, 2006). The multiple precipitating events are often additive, leading to a synergistic risk for the neurodegenerative disorder. The traditional theory of AD is that there are disruptions of gene expressions that can lead to development of AD (Bullido et al., 1998). Some recent theories hold that the disruption of gene expression needs catalyzing events to make the transition from genotype to phenotype (Bullido et al., 1998). Little consideration is given to epigenetic alterations in both gene expression and gene patterns (Becker, 2004; Wang, Oelze, & Schumacher, 2008). Theorists have indicated that many instances of AD are not accounted for by the current theories of the pathophysiology of AD (Becker, 2003). The central tenet of the thesis presented here is that while the genetic theory is accurate, there is another relevant story about the role of systemic regulation and interactions between brain, body and environment that could have implications for understanding and treating neurodegenerative disorders (Wang, Oelze, & Schumacher, 2008). The systems dysregulation paradigm starts out with the premise that there are many roads to neurodegeneration and there are multiple events and classes of events that can synergistically lead to the same type of dysregulation and similar patterns of dysfunction. To exemplify this, disruptions in cortisol secretion can be brought about through: a. tumors in HPA axis, b.
  • 17. 17 depression, c. PTSD, and d. disruption in early attachment (Young, Abelsona, & Camerona, 2003; Yehuda, Teicherbc, Trestmana, Levengooda, & Sievera, 1996; Penza, Heim, & Nemeroff, 2003; Dorn, Burgess, Friedman, Dubbert, Gold, and Chrousos, 1997). Dysregulation of cortisol has been associated with hippocampus shrinkage, loss of sleep patterns, disruption to the dopamine system, disruptions in concentration, toxic cell death and anhadonia (R. Sapolsky, personal communication, 2001). This is only one system. It is an important system for autoregulation but not by any means the only. From the systemic dysregulation paradigm, in order to for the genotype of AD to become the phenotype of AD, there would need to be disruptions in the ability to form memories, maintain hippocampus volume, rate of cell death, mitosis and maintain previously encoded memories. These dysregulations could occur in multiple levels of the system. The interplay between life events, environmental toxins, volitional behaviors and chemical contexts and the functional relationship between anatomical structures are a few of the factors that could add to disruptions in memory retention and formation. From a systems perspective this is not a surprise because there are often several key areas of vulnerability in a system that lead to increased risk for these patterns’ systemic dysregulation, not simply one. Systemic Disruptions Present in Individuals with AD In the paths leading to Alzheimer’s there are some major themes that emerge. These are: a. changes in stress response, environmental stressors and systemic stress, b. changes in subsystem functioning, c. functional shifts between systems, d. alterations in the metaplastic environment, and e. changes in cellular senescence (Attix & Welsh, Bohmer; Chen, Kagan, Hirakura, & Xie, 2000; Sorg et al., 2007; Peskind, Wilkinson, Petrie, Schellenberg, & Raskind, 2001). Mapping some of these key changes could illuminate possible places in the system where
  • 18. 18 one might create interventions to re-establish the neurobiological system’s functional capacity to maintain itself and auto-regulate. Alterations in Stress Response Abnormal stress response has been noted in many individuals with AD (Popp et al., 2009). High levels of CORT have been shown to be associated with loss of hippocampal tissue in depression, PTSD, Cushing’s syndrome and AD (Young, Abelsona, & Camerona, 2003; Yehuda, Teicherbc, Trestmana, Levengooda, & Sievera, 1996; Penza1, Heim, & Nemeroff, 2003; Dorn, Burgess, Friedman, Dubbert, Gold, & Chrousos, 1997). In one study of the effects of reducing CORT levels in individuals with Cushing’s syndrome, it has been noted that the hippocampal tissue regenerated significantly (Starkman, Giordani, Gebarskic, Berent, Schork, & Schteingart, 1999). Upregulation of CORT is associated with anhadonia, psychomotor retardation, poor memory encoding, lack of ability to concentrate, loss of interest in sexuality, increased anxiety and aggression. PTSD is a risk factor for the development of AD. In both AD and PTSD there is a decrease in heart rate variability (HRV) in AD a direct relationship was noted between HRV and symptom severity (Zulli et al., 2005; Cohen et. al., 1998, Zulli et al., 2005). Alterations in CORT levels also lead to disruptions in the dopamine system, norepinephrine system, the serotonin systems and the acytocholine system (Oswald et al., 2005; Pacaka et al., 2002; Idoyaga-Vargas, Abulafia, & Calandria, 2001; Geracioti, et Al., 2001; Pacak, Palkovits, Kopin, & Goldstein, 1995; Meshorer, & Soreq, 2008; Kirkwood, Rozas, Kirkwood, Perez, & Bear, 1999). CORT also plays a role in the regulation of dopamine and dopamine loss could lead to increased LTD and reduced LTP (Calabresi et al., 2000). Cortisol may end up being a large system that, if dysregulated, can lead to many systemic changes (R. Sapolsky,
  • 19. 19 personal communication, 2001). Alterations in stress response due to life stress in younger animals have been noted to create large-scale changes in multiple systems. “Compared to controls, traumatized animals showed an increase in Ca2+ homeostatic proteins, dysregulated signaling pathways and energy metabolism enzymes, cytoskeleton protein changes, a decrease in neuroplasticity regulators…, and an increase in apoptotic initiator proteins (Uys, Hatting, Stein, & Daniels, 2008).” CORT production decreases telomerase production throughout the body (Choia, Faucea, & Effros, 2007; Epel et al., 2004). Telomerase down-regulation has been implicated in AD and other neurodegenerative disorders (Mattson, 2000). There are multiple determinants for the systemic functioning of CORT; genetics, diet, environmental factors, and epigenetic factors. Some environmental factors are number of adverse childhood events, PTSD, disruptions in infant child bonding, exercise, and social support (Shea, Walsh, MacMillan, & Steinera, 2004). Genetics has been implicated in the creation of the autonomic set point for CORT production and changes in set point were catalyzed in a gene by environmental interaction (Adamafio, 2009; Wüst, Federenkoa, Hellhammera, & Kirschbaumb, 2000; Kirschbaum, Wust, Faig, & Hellhammer, 1992). Even short-term disruptions in parent- child bonding or mild increases in parental aggression lead to significant life time alterations in CORT response (Van Oersa, Kloetb, & Levinea, 1998). Number of adverse childhood experiences correlates with increased depression, anxiety and self-destructive behaviors (Felitti, Anda, Nordenberg, Williamson, Spitz, & Edwards, 1998). Neuroplastic Dysregulation The brain is a learning context. Each time we move, think or plan we are sculpting the brain. In order for the brain to maintain optimal functioning it needs to balance its rate of change. If it changes too quickly the brain becomes an unstable environment. If it does not change
  • 20. 20 quickly enough it will have difficulty learning to map experiences. There are currently seven main processes that describe how the brain maintains its internal architecture at an optimal level of flexibility: long-term potentiation, long-term depression, activity dependent changes, metaplasticity, neurogenesis, kindling and salience effects (Burrell & Sahley, 2004; Eriksson et. al., 1998; Ichise et. al., 2000; Kalivas, & O'Brien, 2008; Malenka, & Nicolla, 1999; Minabe & Emori, 1992; Mueller, Pollock, Lieblich, Epp, Galea, & Mistlberger, 2007; Nitsche et. al., 2006; Wickliffe & Bear, 1996). These are the learning-based regulators identified to date. Along with these regulators there are also metabolic and structural regulators of plasticity. One of these is the Mglur (Matabtropic glutamate receptors) that, when stimulated, reduce the number of AmpR receptors attempting to control and regulate the level of excitatory potential possible. If the level of excitatory potential is exceeded, then the neuron can die due to excytotoxicity (Choi, 1992). Individuals with AD have multiple alterations in the rate of neuroplasticity (Arendt, 2003; Kimmo, Krystyna, Tomoaki, Daniel, & Stanley, 1999; Flood, & Coleman, 1990; Shankar et. al., 1990). They also have been noted to display a decline in cell density in the hippocampus that spreads over the course of the disease to other brain areas. One of the key areas of dysregulation is in the actocholine systems which is a global regulator of the rate of plasticity (McKay, Placzek, & Dani, 2007; Whitehouse, Martino, Antuono, Lowenstein, Coyle, Price, & Kellar, 1986). Most current AD meications are acytocholinergic. Another effect on plasticity is Abeta (often expressed in individuals with AD) in one study found to be a clear down-regulator of LTP, possibly effecting neuronal pattern stability (Chen, Kagan, Hirakura, & Xie, 2000). In a 2010 study in a Prelisin 1 knock out mouse (a mouse model of some of the processes in AD) there was noted an upregulation of LTD. These mice show an upregulation of LTP (early phase LTP, late is phase similar to controls) early in life that drops off as they age. This produces a
  • 21. 21 high rate of quick changes that do not get stabilized into fully encoded neuronal patterns. This upregulation thus leaves the brain vulnerable to destabilizing the patterns of connectivity and catastrophic LTD. Another study found that chronic exposure to adrenergic stimulation (common in those with AD) upregulated the LTD mediated by Alpha1AR (McElligott, & Winder, 2008; Davis et. al., 1996; Popp et. al., 2009). Endocannabinoids are another regulator of the rate of plasticity (Kyriakatos & Manira, 2007; Pazos, Núñez, Benito, & Tolón, 2004). They function as agents facilitating the increase of excitatory synaptic plasticity, LTD and LTP (Pazos, Núñez, Benito, & Tolón, 2004). The endogenous canibinoids are regulated by a calcium dependent mechanism. The mechanism works like a switch priming cells to release eCBs but only coupled with a transient rise in Ca2+. Another group of researchers found “the existence of profound changes in the location and density of several elements of (the endocanibanoid) system in Alzheimer's disease tissue samples, indicating that a non-neuronal endocannabinoid system is up-regulated in activated glia” (Pazos, Núñez, Benito, & Tolón, 2004). Multiple studies have shown changes in Serotonin in individuals with AD (Mintzera, et. Al, 1997). Sertatonin has been noted to “regulate cell proliferation, migration and maturation in a variety of cell types, including lung, kidney, endothelial cells, mast cells, neurons and astrocytes” (Azmitia1, 2001). Alterations in the dopamine system (increasing D2/D3 receptors) have been noted in individuals with AD (Reeves, Brown, Howard, & Grasby, 2009). Dopamine is a key mediator of LTP and LTD firing for both salient (both positive and negative) and positive salient events (O. Hikosaka, personal communication, May 7, 2009). If there is an action potential previously associated with a dopamine response, that fires without dopamine modulation there is a stimulation LTD. The acytocholine system and the dopamine system work in conjunction,
  • 22. 22 modulating the rate of plastic change. Upregulation or down regulation of dopamine could have profound effects on rates of neuroplastic change (Shen, Flajolet, Greengard, & Surmeier, 2008). Another class of neuroplastic regulation is the activity dependent class of regulation. Activity has been shown to prime patterns of cellular connectivity for LTP (Antonov, Antonova, Kandel, & Hawkins, 2003). Previous firing of a pattern of neurons also primes the neurons for firing. This is could easily form a positive feedback loop of learning. LTD is upregulated by excitatory activity that does not produce a full action potential (Stanton, 1995). If multiple cells send positive signals for action potential to a cell and that cell does not fire, this increases the likelihood of LTD (Calabresi, Maj, Pisani, Mercuri, & Bernardi, 1992). The ratio of signal to noise is another activity-dependent regulator of plasticity (M. Bear, personal communication, June 16, 2003). The brain as a learning context would more likely be served by encoding accurate patterns than irrelevant patterns. The mind is capable of tracking irrelevant patterns if this system is co-opted by other regulators; among these is salience effects and another is signal noise. If the pattern recognized by the brain is highly noisy (e.g., not a strong predictable relationship) this would likely indicate that it is not a pattern. For phylogenic reasons the brain would have been unlikely to evolve if it tracked too many irrelevant patterns. Increased signal noise is a better predictor of LTD than long-term disuse of the synapse. In a study conducted by Marc Bear (2003) Cats who have an eye disrupted chemically so that no signal is getting to the brain, have less LTD in the visual cortex than a cat who wears a single eye patch. In the cat with the patched eye, the increased noise of the eye seeing the blackness alters the patterns of the visual cortex more with more loss in connections then the cat with the nerve signal chemically blocked. In other words higher signal noise ratio lead to more LTD (M. Bear, personal communication, June 16, 2003). This fact has profound implications for the loss of
  • 23. 23 memory in AD: Earlier memories whose pattern is both more encoded and least likely to be triggered by current context would be the most undisturbed by this process. It is possible that the memory is evoked and the heightened plastic context, in conjunction with the increased noise-to signal-ratio due to the flattening of salience indicators, leads to a destabilization of the cellular pattern that marks the memory of the event. This destabilization in turn leads to a loss of connectivity and eventually to cell tissue death. In older adults there are several key possibilities increasing signal to noise and thus the likelihood of a catastrophic loss of connectivity and apoptic cascade. These are: a. poor sensory information due to changes in physical sensors (ear drum, hairs in the ear, hardening of the cornea, slower reaction times, reduced sense of smell); b. inaccurate reading of the sensory information by brain systems (changes in ear structure have not changed how the brain receives the information from the ear); and c. internal events (stress, anxiety, pain, fear, other affect, worry, thoughts creating internal distraction and increased signal noise) (Mahncke, Bronstone, & Merzenich, 2006). This last class (internal events) is significant in older adults and has not been discussed thoroughly as an impact on brain health in aging. Another final note on the activity dependent regulators of plasticity is that in older adults, particularly in Western cultures, there are alterations in lifestyle that can reduce the amount and quality of environmental stimulation. It is interesting to note that increases in mobility, social stimulation and environmental novelty have been noted to reduce symptoms of AD and be protective factors against developing AD (Nithianantharajah & Hannan, 2006). Major Types of Neuroplastic Change and Their Implication for Systemic Dysregulation Regulation of Metaplasticity: Upregulation of long-term potential could in certain system states lead to forming many patterns that are weakly associated, as in the blooming and pruning
  • 24. 24 cycles early in life. This cycle leads to the formation of many new but not entirely accurate patterns of neuronal associations. Later these patterns are not supported by external events. In essence, an upregulation of metaplasticity, increasing LTP past a certain point, could lead to an upregulation of LTD due to increased signal–to-noise ratio, thus leading to receptor loss, destabilization of balance between functional areas, axonal connections and eventually cell death. Down-regulation of LTP, telomerase activity and neurogenesis could lead to an inability to replace cells and rebuild damaged synapses. This leads to a slow but steady attrition of memory and abilities that eventually reaches a crisis point when the relationship between external events has such dramatic increased noise that it precipitates a rapid rate of LTD. A metaplastic decreased LTD could lead to many patterns being encoded and competing for attention there by disrupting retrieval and overloading pattern recognition with relevant possibilities. It is also possible that this leads to a single external event triggering in a context irrelevant manor many patterns of neuronal relationships disrupting the synchronous patterns of firing. Regulation of Neurogenesis: Neurogenesis is the forming of new neurons in the brain. Until recently it was believed that neurogenesis stopped as an individual reached adulthood. An under-regulation of neurogenesis could lead to lack of replacement for damaged cells, reduced memory formation and overtime destabilization of current patterns of neuronal connectivity. Regulation of Salience Effects: If dopamine cells fire in conjunction with a pattern of neuronal firming, this marks the pattern or an aspect of the pattern as highly salient for the continued functioning of the organism. This increases the possibility of LTP forming a new neuronal pattern. If a pattern fires without a dopamine signal. the pattern is more likely to
  • 25. 25 destabilize and enter the LTD cycle. To be precise it is currently thought that dopamine fires at both salient (both positive and negatively relevant events) and positive events. Acytocholine Cells: These cells increase the sustained attention on the external event and thereby the accurate encoding of a neuronal map of that pattern (Himmelheber, Sarter, & Bruno, 2000). Increases of ACh lead to a metaplastic increase in the rate of neuroplasticity (Jerusalinsky, Kornisiuk, & Izquierdo, 1997). Upregulation in ACh could lead to destabilized patterns of relationship due to the formation of neural circuits that are only loosely reflected in the external context. An under-regulation of ACh could lead to a lack of ability to form new memories and reduced activity-dependent maintenance of current patterns of neuronal firing. This dysregulation would likely not produce a boom and bust cycle but rather produce a slow degradation of memory and memory formation with an exponential increase in loss toward later parts of the disease process. Brain Derived Neurotropic Factors (BDNF) Regulation: BDNF is a protein responsible for the growth and maintenance of nerve cells. As well as its action in the brain, it also plays a role in motor neurons, kidneys, prostate and is often present in saliva (Binderm & Scharfman, 2004; Huber, Hempstead, & Donovan, 1996; Pflug, Dionne, Kaplan, Lynch & Djakiew, 1995; Mandel, Ozdener, & Utermohlen, 2009). BDNF supports the health of existing cells, the growth of new cells and the building of new synapses. It is found in areas related to higher order learning, memory, and problem solving (e.g., hippocampus and cortex) (Hall, Thomas & Everitt, 2000). BDNF knock out mice have been shown to die at birth or have major neurological difficulties, including sensory neuron loss (balance, hearing, and taste) and breathing problems (Ernfors, Kucera, Lee, Loring, & Jaenisch, 1995). BDNF is implicated in multiple disease processes including AD, depression, psychotic spectrum disorders, obsessive compulsive
  • 26. 26 disorder, dementia, anorexia, and bulimia. Under certain conditions it can increase cell death instead of protect against it (secretion of p75NTR in the absence of Track A, B or C can lead to cell death). BDNF plays a large role in neurogenesis (Bekinschtein et. al., 2008). It has a reward salience effect in the ventral tegemental area. BDNF has been shown to be increased by exercise, restricted calories, treatments for depression and intellectual stimulation (Gómez-Pinilla, Ying, Roy, Molteni, & Edgerton, 2002). BDNF is implicated in the reversal of hippocampal damage that occurs subsequent to depression treatment. Cortisol has been shown to reduce overall brain levels of BDNF (Smith, Makino, Kvetnansky, & Post, 1995). Under or overregulation of BDNF could have catastrophic effects on brain health. If BDNF were down-regulated in a high cortisol context elevated it could be catastrophic. Amygdala and Bed Nucleus: The amygdala is a well known area of the brain that acts like a smoke detector for threats, stimulating the brain and body into a fight, flight or freeze defensive response. The AMY plays a large role in two aspects of neuroplasticity: it controls cortisol secretion and thus affects the metabolic ways a neuron can live or die, and it regulates salience effects for threat. Hyper-sensitive, easily triggered stress response or high number of triggering events coded by the AMY could lead to profound cell damage due to being in a metabolic endangered state such that minor insults could kill them. The bed nucleus is connected to the same areas of the brain as the AMY. It mediates long-term anxiety. Thus it is highly implicated in worry or psychological stress and may be responsible for the slow attrition of neurons seen in individuals who are low in a dominance hierarchy or have poor emotional regulation skills. Upregulation of these areas could lead to increased excitoxic cell death and Under-regulation of these areas could lead to reduced salience effects and negative consequences on multiple systems (Sopolsky, 2005).
  • 27. 27 Activity Dependent - Previous Firing of Cells: Recent previous firing predisposes the area to fire again under similar conditions (Antonov, Antonova, Kandel, & Hawkins, 2003). The cells are thus primed to highlight certain relationships in external contexts by firing more easily in the internal context. In a highly upregulated context of firing there is a loss of differentiation between firing patterns. If firing becomes indiscriminant and not related to situational events it could increased LTP, the encoding of inaccurate patterns of events and subsequently a catastrophic destabilization of neural context. Under-regulation of firing rate could lead to increased LTD due to inability for one part of a pattern of firing to trigger an action potential in related systems. Switching Neurological Sets: Another key aspect of the brain physiology that could enhance the development of AD is the reduction of the ability to switch between functional systems. Recent studies have found that there are major neurological organizational systems for broad classes of types of life tasks (Taylor, Seminowicz, & Davis, 2009). Purely cognitive tasks require different neurological demands than physical tasks or emotional learning tasks. A study of individuals with early AD has found that there is a reduction of the ability to shift between these larger scale functional systems. The insula, which often plays a gating function between cognitive and emotional systems, no longer activates freely in those in early stage AD. A key impact could be that mental states and memories that require the ability to shift between functional systems could be difficult to access. This could lead a destabilization of neuronal memory constellations and learned helplessness in accessing memories. Attention and Signal Noise: Working memory has a limited amount of space for processing information. Pain, psychological stress, and internal sensations could all co-opt significant portions of working memory. This could lead to a feedback loop of increased focus
  • 28. 28 on the pain or stressor, causing more pain and stress, and so on. If the pain and stress are occupying significant amounts of working memory space, concentration goes down, the metabolic effects of stress go up, and the ability to attend to and learn from the environment goes down. This could lead to lack of neurogenesis, loss of nerve cells and reduced dendritic density through increased signal noise ratio. Metabolic Effects Apoptotic Regulatory Factors Multiple alterations in indicators of apoptosis are seen in individuals with AD. Amiloid B protein, thought to be a major factor in the formation of AD, has been shown to be neurotoxic (Cotman & Anderson, 1995). Alterations in reactive oxygen have been noted in individuals with AD. Reactive oxygen is a major chemical used by the immune system to induce apoptosis in the body. T-lymphocytes, the cells responsible for this immune function, are down-regulated in aging but upregulated in individuals with AD. This study finds “elevation of [Ca2+]i appears to be a prerequisite for apoptosis, which is suggested to be involved in the neuronal death occurring in AD. An increased [Ca2+]i in AD is consistent with processes leading to neurodegeneration in AD” (Sulgera, Dumais-Hubera, Zerfassa, Henna & Aldenhoffb, 1998). Alterations in caspase-8 have been noted, “[a] role for caspase-8 and the receptor-mediated apoptotic pathway as a mechanism leading to the activation of caspase-3 within neurons of the AD brain” (Rohna, Headb, Nessea, Cotmanb and Cribbs, 2001). Alterations in the mitochondrial-produced AIF (apoptic inducing factor) have been noted in individuals with AD. Apoptic Catastrophe There are multiple major paths that can lead to a catastrophic apoptic cascade in the subsystems of the brain. One path leads to apoptic cascade through a metabolic upregulation in
  • 29. 29 cellular energy production (Choi, 2004). The second is through destabilization of cellular connectivity through alterations in the homeostatic range of neuroplastic change. In this process there are transformations in the brain’s potential to learn. This dysregulation could lead to destabilization of a functional system or brain-wide, leading to loss of synapses, reduction of neurogenesis and cell death. The third is through dysregulation in the process of cellular reproduction, transcription, translation and the life cycle of a cell. The fourth is systemic dysregulation of major homeostatic processes. In this process the destabilization of the neurological environment is established by lack of vital chemicals, disruptions in autoregulatory process (i.e., blood sugar, sleeping, metabolism, oxygenation, excretion, illness). The fifth is dysregulation of the relationships between major psychoneurological functional areas, including dysregulation of switching between functional systems, dissociation between functional areas in a system, over-coupling between functional systems and increased irrelevant cueing for a psychobiological functional area. The path to apoptic cascade includes multiple types of causal events, including predispositions, synergistic events and non-linear dynamics. There is an interplay in the brain between genetic determinants and contextual events that sculpt its development (Thompson et. al., 2001; Caspi & Moffitt, 2006). The context of learning is created by an algorithmic pattern of associative events that allows for the mapping of the external realities, internal aptitudes and behavioral patterns in the biological relationship between cells. This process is likely in part mapped genetically and determined by a self-organizing process defined by the biological possibilities interacting with external events. Of note children who face trauma, are raised under high stress or experience disruptions in parenting and have differently organized cortical and sub-cortical areas (Schore, 2001). Life-span Dependent Psychoneurochemical Changes
  • 30. 30 In older adults there is often a confluence of difficulties that could increase the likelihood of apoptic cascade due to either metabolic factors or learning-based factors. As adults age there is an change in the production of GABA (one of the brain’s main inhibitory neurotransmitters) and a decrease of the regulatory inhibition of gabanergic neurons by adenosine triphosphate (ATP) (Marczynski, 1998). The increase of inhibitory neurotransmitters could increase the likelihood of raising the resting state of the brain to a point well below the threshold for action potential creating significant difficulty for the encoding of a new patterns. Reduction in sleep and sleep disturbances are common among elders (Huanga, Liua, Wangb, van Somerenc, Xub, & Zhou, 2000). Lack of sleep in younger adults has been associated with precipitous drop-offs in neurogenesis and thus a slower ability to replace cells when they die (Mueller, Pollock, Lieblich, Epp, Galae, & Mistlberger, 2008). REM sleep is the time of the most major daily increase in acytocholine, a chemical involved in the regulation of attention, encoding memory and the rate of plasticity. REM sleep is significantly reduced as modern Westerners age (Ehlersa & Kupfe, 1989). Another key time period of susceptibility for women is when entering menopause (Solertea, Fioravantia, Racchib, Trabucchic, Zanettib, & Govonid, 1999). Reduction in estrogen has been associated with the increased chance of development of neurodegenerative disorders and low levels of estrogen replacement were found to be a mild protective factor against AD in initial research (Vegetoa, Benedusia, & Maggi, 2008; Solertea et. al., 1999). Another key time of vulnerability is in infancy. During this time even seemingly minor increases in stress can lead to marked changes patterns of stress reactivity and regulation apoptic factors in the hippocampus throughout a lifetime (Liu et. al., 1997; Anand & Scalzo, 2000; Weaver, Grant, & Meaney, 2002). Other likely times of disruptions are in times of rapid
  • 31. 31 neurological development, such as myolenation of the frontal lobe in late teens or major life transitions. Often older adults live in an impoverished learning context due to several reasons: a. significant over-learning about relevant environmental events and thus a lack of novelty, b. lack of novel environmental cues and reduction of mobility, c. diminished perceptual strength due to changes in sensory input, d. poor exercise, and e. poor social support due to ageism in Western societies. An impoverished learning context has been associated with loss of synaptic density and cell loss in neural structures (Sandeman & Sandeman, 2000; Turnera & Greenough, 1985; van Praag, Shubert, Zhao, & Gage, 2005). Assessing the individual for disruptions that happen at key points of developmental transition could lead to effective interventions. Systems Based Approach: Implications for the modeling of brain aging and treatment of AD As we have seen before there are multiple ways that any system can become unstable. Alzheimer’s disorder has many different antecedent events (Attix & Welsh, Bohmer; Chen, Kagan, Hirakura, & Xie, 2000; Sorg et. al., 2007; Peskind, Wilkinson, Petrie, Schellenberg, & Raskind, 2001). As one prominent researcher notes pithily, when you have seen one case of Alzheimer’s, you have seen one case of Alzheimer’s (Whitehouse & George, 2008). In the systems approach it is assumed that there are many possible ways that the brain and body could produce what looks like Alzheimer’s disorder. Each of these ways of creating symptoms called Alzheimer’s Dementia will have a specific conglomeration of factors. The role of this section will be to outline how to use this approach in identifying key dysfunctions in the brain system and to provide means to reregulate these processes. The first step in this process would be to identify on a case-by-case basis which major systems are dysregulated. This would require identifying possible disruptions that could lead to
  • 32. 32 the symptoms at hand. Some key areas of systemic disruptions we have identified are dysregulation of CORT, disruptions to oscillation between parasympathetic and sympathetic nervous system functioning, disruptions in plasticity, dysregulation of cellular senescence, and dysregulation of dopamine or acytocholine systems. These factors could be exacerbated by natural aging effects on the neural systemic contexts. Events such as changes in GABA production and the phase transition from pre-menopausal to post-menopausal neurochemistry could create synergistic effects with the above systems. These disruptions could be identified by multiple types of tests such as diurnal cortisol tests, measuring for overall telomerase production, assessing glutamate production, and assessing if GABA is presented in age-appropriate levels. These and other findings could be assigned a statistical range of likely contribution to the disease process (identified through studies and the existing literature), the additive effects, synergistic effects and catalyzation points for phase change in the brain could be also be quantified. A good multivariate analysis could assess the impacts, interactions and synergies and find threshold levels for phase changes in the brain. The second step would be to assess if there are global or local effects of the dysregulation. This could be vital information for how to assess which systems are more likely to be vulnerable and which systems are main drivers for the dysregulation. The third step is to identify typically occurring protective defensive responses in these systems and the steps and stages of return to systemic coherence after an insult. Of particular interest are areas vulnerable to feedback loops. Often when a given system is taxed or is injured it will enter an allostatic response as it attempts to mitigate the damage as it occurs and repair damage that is existing. The ability of the tissue to instigate this pattern and then return to baseline once the threat is gone are both vital to the health of the organism. As Dr. Kurr noted in
  • 33. 33 his work with the neural adapted sinibus virus, sometimes these defensive responses can themselves trigger other protective responses and enter into a deadly feedback loop. The fourth step is to identify what resources the system needs to meet the demands of completing the protective response. Sapolsky, in a 2001 talk on Alzheimer’s disorder and over- activation of cortisol, describes that cells, when stimulated by cortisol, can enter into an energy crisis. The demands for energy in the hippocampal tissue exceed the tissue’s ability to meet those demands. The dendritic connections are sloughed off and many protective measures are taken to avoid the metabolic crisis that occurs if the cell is over-stimulated. Sapolsky (2001) goes onto to state that if the cells are given an easy to digest form of glucose, thus providing enough energy to complete the defensive measures and meet the demands of the cell, the crisis is averted and the cells will not die. The fifth step is to identify the secondary, tertiary, etc., systemic effects and note if any of these effects will not re-regulate once the regulatory boundary is re-established for the system of primary dysregulation (e.g., hippocampal tissue loss will be stopped and reversed through reregulation of CORT; on the other hand, in another system the reregulation of telomerase will not lead to reduction of stress-induced premature cellular senescence). The effects of a primary dysregulation on other systems could be that situational stress leading to increased CORT production is leading to: a. increasing the rate of cellular senescence; b. reduced cognitive functioning that synergistically compounds these effects through enhancing poor eating habits, c. increased anhadonia through disruption in the dopamine systems, leading to lack of motivation and impoverished social environments; and d. increased aggression disrupting the ability the primary social support system to provide regulation for stress reactivity.
  • 34. 34 The sixth step would be to identify means to work with the system to re-establish a functional regulatory range. A key assessment in this is to identify when to use “global” or top down interventions vs. when to use focused bottom up interventions. Bottom up interventions address the regulation of the whole system by changing a single system, function, protein or chemical. Top down interventions change the regulation in subsystems by re-regulating the entire context of the body (e.g. increasing sleep and re-establishing sleep architecture can change metabolism, neurogenesis, BDNF levels and work functioning etc.). In the case in which dysregulation of CORT is the major driver for dysregulation building in the ability for the nervous system to smoothly down-regulate through establishment of a parasympathetic braking of the sympathetic system could be vital. This could be accomplished through multiple means. Increased sleep and re-establishing healthy sleep architecture could lead to down-regulation of the stress system. Increase in number and duration of pleasurable events could increase the parasympathetic tone. Social engagement also increases parasympathetic tone. Many stress reduction techniques found in CBT, MBSR and other protocols could reduce the stress response (Koszyckia, Bengera, Shlika, & Bradwejna, 2007; Praissman, 2008). This could then leave the system more able to form new memories and regrow the hippocampal tissue. The seventh step is to assess the functioning of affected systems post re-regulation of the primary dysregulated system. Any systems that do not re-establish their functional range may need to have a process that addresses their functional dysregulation. Assessing in this case for dysregulation and factors reducing the system’s ability to return to functional range will likely allow the clinician to help this system return to functional capacity. It may in fact turn out that the stress reactivity set in motion alterations that have made a stable phase shift. Finding means to hold the regulatory boundaries for that system so that it can shift to the previous functional
  • 35. 35 relationship could then reduce the dysregulation in that system. As an example, if the CORT secretion has reduced telomerase production to a level where oxidative stress can lead classes of cells to premature senescence, reduction of CORT with likely not re-regulate this area (Serrano & Blasco, 2002). Increases in factors leading neurogenesis, re-regulation of telomerase and increased antioxidants could begin the process of interrupting the cascade of cell death that was established by the dysregulation of cellular senescence. Increased REM sleep could help to increase neurogenesis and re-regulate any disruptions to the acytocholine system established by the elevations of CORT (Mirescu & Gould, 2006; Cameron & McKay, 1999; Mueller, Pollock, Lieblich, Epp, Galae, & Mistlberger, 2008; Mohapela, Giampiero, Kokaiaa, & Lindvalla, 2005; Vazquez & Baghdoyan, 2001). It is important to note that there are many ways to affect any given system and the many effects that any given system can have. This implies that the above formulation is only a guide for possible interventions and modeling. If the main dysregulation was in neuroplasticity and not CORT there would be multiple other types of interventions and the systems that are subsequently affected would differ. Taking a systems approach to treatment and working with the health in the system, it may be possible to reverse some of the loss of functioning and extend the health and mental life for individuals with and without cognitive impairment till much closer to the end of their life. This could enhance the quality of life across the lifespan. This description of Alzheimer’s disorder is meant as an example of a broader class of disorders of regulation and ways to map cases of AD that have other causal events leading to the main dysregulation. Disorders of regulation are marked by changes in auto-regulation that can lead to disruptions in the functioning of multiple systems in the body, brain and mind. Alzheimer’s disorder, like other disorders of regulation, can have multiple events that lead to its
  • 36. 36 onset. These events lead to disruption in functional dynamics and destabilization of the system. A complex functioning system like the human brain and body also has many processes that help establish and maintain healthy systemic functioning. Just as this class of disorders can be created by the body’s own mechanisms working out of synchronization, they likely can be healed through re-establishment of the functional regulators of the biological system.
  • 37. 37 References Adamafio, N. A. (2009). The hormone-emotion-behavioral gene-neuromessenger labyrinth: Pertinent questions. African Journal of Biochemistry Research, 3(10), 326-331. Adamec, R., & Young, B. (2000). Neuroplasticity in specific limbic system circuits may mediate specific kindling induced changes in animal affect—implications for understanding anxiety associated with epilepsy. Neuroscience Biobehaviral Review. 24(7), 705-23. doi:10.1016/S0149-7634(00)00032-4. Artola, A. (2008). Diabetes, stress, and ageing-related changes in synaptic plasticity in hippocampus and neocortex - The same metaplastic process? European Journal of Pharmacology. 585(1),153-62. doi:10.1016/j.ejphar.2007.11.084. Attix, D., & Welsh-Bohmer, K. (2006) Geriatric Neuropsychology. New York, NY: Guilford Press. Azmitia1, E. C. (2001). Modern views on an ancient chemical: serotonin effects on cell proliferation, maturation, and apoptosis. Brain Research Bulletin, 56(5), 413-424. doi:10.1016/S0361-9230(01)00614-1 Anand, K. J. S., & Scalzo, F. M. (2000). Can Adverse Neonatal Experiences Alter Brain Development and Subsequent Behavior? Biological Neonatology Fetal and Neonatal Research, 77, 69-82. DOI: 10.1159/000014197 Bear, M. (2003). The changing brain, MIT WORLD http://mitworld.mit.edu/video/152 Beaumont, D., Lehmann A. B., & James, O. F. W. (1989). Protein Turnover in Malnourished Elderly Subjects: The Effects of Refeeding. Oxford Journal of Medicine, 18(4), 235-240. Becker, K. (2003). The common variants/multiple disease hypothesis of common complex genetic disorders. Medical Hypothesis, 62(2), 309-317. ISSN: 0306-9877.
  • 38. 38 Bermudez, Y., Erasso, D., Johnson, N., Alfonso, M., Lowell, M., and Kruk, P. (2006). Telomerase confers resistance to caspase-mediated apoptosis. Clinical Intervention Aging, 1(2), 155–167. DOI 10.2147/CIA.S. Berkeley, California, LINK to PDF: http://docs.google.com/viewer?a=v&q=cache:Xccxp4gQcEQJ:www.er.doe.gov/bes/repor ts/files/CS_rpt.pdf+Coherence+in+complex+systems&hl=en&gl=us&pid=bl&srcid=AD GEESiDxzbmZac3kdV_hc4UmFDZXuZAK5kbR8Fl- Wr5q89rZtuO4SAjxZvf7d4XlONL05ZpQsHE1uCo__2r_3fGBxBu2aRXTtedkFPCTv6q YdAip-xjHZATR8qaiFPwHncKGYJYf9oM&sig=AHIEtbT-PLhHUBxrmx3- pyZM2GOE5xsZFw Blasco, M. A. (2005). Telomeres and human disease: aging, cancer, and beyond. Nature Review Genetics, 6(8), 611-622. PMID 16136653. Brett, M., Johnsrude, I. S., & Owen, A. M. (2002). Opinion: The problem of functional localization in the human brain. Nature Reviews Neuroscience 3, 243-249. doi:10.1038/nrn756. Bremner, D., Narayan, M., Anderson, E., Staib, L., Miller, H., and Charney D., (2000). Hippocampal Volume Reduction in Major Depression. American Journal of Psychiatry, 157, 115-118. PMID: 10618023. Bruzz, P., Green, S., Byar. D., Brinton, L., & Schairer, C. (1985). Estimating the population attributable risk factors using case-control data. American Journal of Epidemiology, 122(5), 904-914. PMID: 4050778. Bourdeau, I., Bard, C., Noël, B., Leclerc, I., Cordeau, M., Bélair, M., Lesage, J., Lafontaine, L., and Lacroix, A. (2002). Loss of Brain Volume in Endogenous Cushing’s Syndrome and
  • 39. 39 Its Reversibility after Correction of Hypercortisolism. The Journal of Clinical Endocrinology & Metabolism, 87(5), 1949-1954. PMID: 11994323. Bullido, M. J., Artiga, M. J., María Recuero, M., Migue, I., García, A., Aldudo, J., Lendon, C., Han, S., Morris, M., Frank, A., Vázquez, J., Goate, A. & Valdivieso, F. (1998). A polymorphism in the regulatory region of APOE associated with risk for Alzheimer's dementia. Nature Genetics, 18, 69–71. doi:10.1038/ng0198-69. Calabresi, P., Gubellini, P., Centonze, D., Picconi, B., Bernardi, G., Chergui, K., Svenningsson, P., Fienberg, A., & Greengard, P. (2000). Dopamine and cAMP-Regulated Phosphoprotein 32 kDa Controls Both Striatal Long-Term Depression and Long-Term Potentiation, Opposing Forms of Synaptic Plasticity. The Journal of Neuroscience, 20(22), 8443-8451. PMID: 11069952. Cameron, H. A., McKay, R. D. (1999). Restoring production of hippocampal neurons in old age. Nature Neuroscience 2, 894-897. doi:10.1038/13197. Casparya, D. M., Milbrandta, J. C. & Helfert, R. H. (1995). Central auditory aging: GABA changes in the inferior colliculus. Experimental Gerontology, 30(3-4), 349-360. doi:10.1016/0531-5565(94)00052-5. Chang, S. (2004). Modeling aging and cancer in the telomerase knockout mouse. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis. 576(1-2), 39-53. Chen, QS, Kagan, BL, Hirakura, Y, Xie, C. (2000). Impairment of hippocampal long-term potentiation by Alzheimer amyloid beta-peptides. Journal of Neuroscience Research, 60(1), 65-72. PMID: 10723069.
  • 40. 40 Choia, J., Faucea S., & Effros, R. (2007). Reduced telomerase activity in human T lymphocytes exposed to cortisol. Brain, Behavior, and Immunity, 22(4), 600–605. doi: 10.1016/j.bbi.2007.12.004. Choi, D. (2004). Excitotoxic cell death. Journal of Neurobiology, 23(9), 1261–1276. PMID: 1361523. Cohen, S., Graham, M., Lovrecz, G., Bache, N., Robinson, P., & Reddel, R. (2007). Protein composition of catalytically active human telomerase from immortal cells. Science 315(5820), 1850–3. doi:10.1126/science.1138596. Cohen, H., Kotlera, M., Matara, M., Kaplana, Z., Loewenthala, U., Miodownika, H., & Cassuto, Y. (1998). Analysis of heart rate variability in posttraumatic stress disorder patients in response to a trauma-related reminder. Biological Psychiatry, 44(10), 1054-1059. PMID: 9821570. Cotman, C., and Anderson, A. (1995). A potential role for apoptosis in neurodegeneration and Alzheimer's disease. Molecular Neurobiology, 10, 19-45. doi:10.1007/BF02740836. Coyle, J. T., & Puttfarcken, P. (1993). Oxidative stress, glutamate, and neurodegenerative disorders. Science, 262(5134), 689-695. doi:10.1126/science.7901908. Davies, M. J., & Pomerance, A. (1972). Pathology of atrial fibrillation in man. British Heart Journal, 34, 520-525. doi:10.1136/hrt.34.5.520. de Diego-Otero, Y., Romero-Zerbo, Y., el Bekay, R., Decara, J., Sanchez, L., Rodriguez-de Fonseca, F., & del Arco-Herrera, I. (2009). Alpha-tocopherol protects against oxidative stress in the fragile X knockout mouse: an experimental therapeutic approach for the Fmr1 deficiency. Neuropsychopharmacology, 34(4), 1011–26. doi:10.1038/npp.2008.152.
  • 41. 41 Dorn, L., Burgess, E., Friedman, T., Dubbert, B., Gold, P., and Chrousos, G. (1997). The Longitudinal Course of Psychopathology in Cushing’s Syndrome after Correction of Hypercortisolism. Journal of Clinical Endocrinology, 82, 912-919. PMID: 9062506. Epel, E., Blackburn, E., Lin, J., Dhabhar, F., Adler, N., Morrow, J., and Cawthon, R. (2004). Accelerated telomere shortening in response to life stress. PNAS, 101(50). 17323-17324. doi:10.1073/pnas.0408041101. Felitti, V.J., Anda, R.F., Nordenberg, D., Williamson, D.F., Spitz, A.M., & Edwards, V. (1998). The relationship of adult health status to childhood abuse and household dysfunction. American Journal of Preventive Medicine, 14, 245-258. PMID: 9635069. Falduto, M., Young, A., and Hickson, C. (1992). Exercise inhibits glucocorticoid-induced glutamine synthetase expression in red skeletal muscles. AJP-Cell Physiology, 262(1), C214-C220. PMID: 1346351. Feig, C., Peter, M. (2007). How apoptosis got the immune system in shape. European Journal of Immunology, 37(S1), S61 - S70. PMID: 17972347. Geracioti, T., Baker, D., Ekhator, N., West, S., Hill, K., Bruce, A., Schmidt, D., Rounds-Kugler, B., Yehuda, R., Keck, P., and Kasckow, J. (2001). CSF Norepinephrine Concentrations in Posttraumatic Stress Disorder. American Journal of Psychiatry 158, 1227-1230. Gerlach. J., Lublin, H., and Peacock, L. (1996). Extrapyramidal Symptoms during Long-Term Treatment with Antipsychotics: Special Focus on Clozapine and D1 and D2 Dopamine Antagonists. Neuropsychopharmacology, (14), 35S-39S. doi:10.1016/0893- 133X(95)00203-P. Gorbunova, V., Seluanov, A., & Pereira-Smith, O. (2002). Expression of Human Telomerase (hTERT) Does Not Prevent Stress-induced Senescence in Normal Human Fibroblasts but
  • 42. 42 Protects the Cells from Stress-induced Apoptosis and Necrosis. Journal of Biological Chemestry. 277(41), 38540-9. PMID: 12140282. Grenier, J., Tomkiewicz, C., Troussona, A., Rajkowskia, K., Schumachera, M., and Massaad, C. (2004). Identification by microarray analysis of aspartate aminotransferase and glutamine synthetase as glucocorticoid target genes in a mouse Schwann cell line. The Journal of steroid biochemistry and molecular biology, 97(4), 342-52, PMID: 16182522. Guzman-Marin, R., Suntsova, N., Methippara, M., Greiffenstein R., Szymusiak R., and McGinty D. (2005). Sleep deprivation suppresses neurogenesis in the adult hippocampus of rats. European Journal of Neuroscience, 22(8), 2111-2116. doi:10.1111/j.1460- 9568.2005.04376.x. Harley, C., and Villeponteau, B., (1995). Telomeres and telomerase in aging and cancer: Geron Corporation, 200 Constitution Drive. Current Opinion in Genetics & Development, 5(2), 249-255. doi:10.1016/0959-437X(95)80016-6. Harley, C. B. (1996). Telomerase and cancer, Nature Reviews Cancer, 8(3), 167-179. Hurley, M., Masha, D., & Jenner, P. (2001). Dopamine D1 receptor expression in human basal ganglia and changes in Parkinson’s disease. Molecular Brain Research, 87(2), 271-279. Hurley, M., Stubbs, C., Jenner, P. & Marsden, D. (1996). D3 receptor expression within the basal ganglia is not affected by Parkinson's disease. Neuroscience Letters, 214(2-3), 23.5-78. PMID: 8878087. Idoyaga-Vargas, V., Abulafia, D., & Calandria, J. (2001). Correlation between cortisol level and serotonin uptake in patients with chronic stress and depression. Cognitive, Affective, & Behavioral Neuroscience, 1, 388-393. doi: 10.3758/CABN.1.4.388.
  • 43. 43 Janicki, S., and Monteiro, M. (1997). Increased Apoptosis Arising from Increased Expression of the Alzheimer's Disease–associated Presenilin-2 Mutation (N141I). The Journal of Cell Biology, 139(2), 485-495. PMID: 9334350. Kannana, K., & Jain, S. (2000). Oxidative stress and apoptosis. Pathophysiology. 7(3), 153-163. PMID: 10996508. Kiel, D., Elliott. E. (1996). Chaos theory in the social sciences: foundations and applications. University of Michegan Press. Kirkwood, A., Rozas, C., Kirkwood, J., Perez, F., & Bear, M. (1999). Modulation of Long-Term Synaptic Depression in Visual Cortex by Acetylcholine and Norepinephrine. The Journal of Neuroscience, 19(5), 1599-1609. PMID: 10024347. Kirschbaum, C., Wust, S., Faig, H. G., & Hellhammer, D. H. (1992). Heritability of cortisol responses to human corticotropin-releasing hormone, ergometry, and psychological stress in humans. Journal of Clinical Endocrinology & Metabolism, 75, 1526-1530. ISSN 0021-972X. Koszyckia, D., Bengera, M., Shlika, J., & Bradwejna, J. (2007). Randomized trial of a meditation-based stress reduction program and cognitive behavior therapy in generalized social anxiety disorder. Behavior Research and Therapy, 45(10), 2518-2526. doi:10.1016/j.brat.2007.04.011. Whitehouse, P. J., & George, D., (2008). The Myth of Alzheimer's: What You Aren't Being Told About Today's Most Dreaded Diagnosis. St. Martin's Press. Lawrence, M., Redmond, D. E. Jr., Elsworth, J. D., Taylor, J. R., & Roth, R. H. (1991). The D1 receptor antagonist, SCH 23390, induces signs of parkinsonism in African green monkeys. Life Sciences, 49(25), PL229-PL234. doi:10.1016/0024-3205(91)90299-Q.
  • 44. 44 Le Ray, D., De Sevilla, D. F., Porto, A. B., Fuenzalida, M., & Bun˜o, W. (2004). Heterosynaptic Metaplastic Regulation of Synaptic Efffcacy in CA1 Pyramidal Neurons of Rat Hippocampus. Published online in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/hipo.20021. Lidow, M. S., & Goldman-Rakic, P. S. (1994). A common action of clozapine, haloperidol, and remoxipride on D1- and D2-dopaminergic receptors in the primate cerebral cortex. PNAS, 91(10), 4353-4356. PMID: 8183912. Linga, L. L., Hughes, L. F., & Caspary, D. M. (2005). Age-related loss of the GABA synthetic enzyme glutamic acid decarboxylase in rat primary auditory cortex. Neuroscience, 132(4), 1103-1113. doi:10.1016/j.neuroscience.2004.12.043 Lisman, J., (2001). Three Ca2+ levels affect plasticity differently: the LTP zone, the LTD zone and no man's land. Journal of Phisology, 532(2), 285. Liu, D., Diorio, J., Tannenbaum, B., Caldji, C., Francis, D., Freedman, A., Sharma, S., Pearson, D., Plotsky, P. M., Meaney, M. J. (1997). Maternal Care, Hippocampal Glucocorticoid Receptors, and Hypothalamic-Pituitary-Adrenal Responses to Stress. Science, 277(5332), 1659–1662. doi:10.1126/science.277.5332.1659. Lowe, S., and Lin, A. (2000). Apoptosis in cancer. Carcinogenesis, 21(3), 485-495. PMID: 10688869. Lua, C., Fua, W., and Mattson, M. (2001). Telomerase protects developing neurons against DNA damage-induced cell death. Developmental Brain Research, 131(1-2), 167-171. doi:10.1016/S0165-3806(01)00237-1.
  • 45. 45 Lennon, S. V., Martin, S. J., Cotter, T. G. (1991). Dose-dependent induction of apoptosis in human tumour cell lines by widely diverging stimuli. Cell Proliferation. 24(2), 203–14. PMID: 2009322 . Mattson, M. (2000). Apoptosis in neurodegenerative disorders. Nature reviews. Molecular cell biology, 120(1), 120-129. PMID: 11253364. Meshorer, E., and Soreq, H. (2008). mRNA Modulations in Stress and Aging: Handbook of Neurochemistry and Molecular Neurobiology. New York, NY: Springer Science DOI10.1007/978-0-387-32671-9. Mirescu, C., & Gould, E., (2006) Stress and adult neurogenesis. Hippocampus 16, 233-238. PMID: 16411244. Mohapela, P., Giampiero, L., Kokaiaa, M., Lindvalla. O. (2005). Forebrain acetylcholine regulates adult hippocampal neurogenesis and learning. Neurobiology of Aging, 26(6), 939-946. doi:10.1016/j.neurobiolaging.2004.07.015. Mueller, A. D., Pollock, M. S., Lieblich, S. E., R. Epp, J. R., Galea, L. A. M., & Mistlberger, R. E., (2007). Sleep deprivation can inhibit adult hippocampal neurogenesis independent of adrenal stress hormones. American Journal of Physiology, Regulatory Integrative and Comparative Physiology. 294(5), R1693-703. Muir, J. (1911). My First Summer in the Sierra (Boston: Houghton Mifflin, 1911), on page 110 of the Sierra Club Books 1988 edition. Nattel, S. (2002). Review article: new ideas about atrial fibrillation 50 years on. Nature 415, 219- 226. doi:10.1038/415219a
  • 46. 46 Naka, K., Akira, T., Ikeda, I., Motoyama, I. (2003). Stress-induced Premature Senescence in hTERT-expressing Ataxia Telangiectasia Fibroblasts. The Journal of Biological Chemistry, 279, 2030-2037. doi: 10.1074/jbc.M309457200. Nichol, G., Hallstrom, A. P., Kerber, R., Moss, A. J., Ornato, J. P., Palmer, D., Riegel, B., Smith, S., Weisfeldt, M. L. (1998). American Heart Association Report on the Second Public Access Defibrillation Conference, April 17–19, 1997. Special Reports. American Heart Association, Inc. 97:1309-1314. Nilsson, J., von Euler, A., & Dalsgaard, C., (1985). Stimulation of connective tissue cell growth by substance P and substance K, Nature 315, 61 – 63. doi:10.1038/315061a0. Norman, J., (2010). The signs and symptoms which suggest the presence of high blood sugar and diabetes. Pulled from the web on 3.21.10 from: http://www.endocrineweb.com/diabetes/hyperglycemia.html Okouchi, M., Ekshyyan, O., Maracine, M., & Yee, T. (2007). Apoptosis in Neurodegeneration. Antioxidants & Redox Signaling. 9(8), 1059-1096. doi:10.1089/ars.2007.1511. Ornish, D., Lin, J., Daubenmier, J., Weidner, Y., Epel, E., Kemp, C., Jesus, M., Magbanua, M., Marlin, R., Yglecias, L., Carroll, P., & Blackburn, E. (2008). Increased telomerase activity and comprehensive lifestyle changes: a pilot study. The Lancet Oncology, 9(11). 1048-1057. PMID: 18799354. Oswald, L., Wong, D., McCaul, M., Zhou, Y., Kuwabara, H., Choi, L., Brasic, J., & Wand, G. (2005). Relationships Among Ventral Striatal Dopamine Release, Cortisol Secretion, and Subjective Responses to Amphetamine. Neuropsychopharmacology 30, 821–832. doi:10.1038/sj.npp.1300667.
  • 47. 47 Pacak, K., Palkovits, M., Kopin, I., & Goldstein, D. (1995) Stress-Induced Norepinephrine Release in the Hypothalamic Paraventricular Nucleus and Pituitary-Adrenocortical and Sympathoadrenal Activity: In Vivo Microdialysis Studies. Frontiers in Neuroendocrinology. 16(2), 89-150. PMID: 7621982. Pacaka, P., Tjurminab, O., Palkovitsc, M., Goldsteind, D., Kocha, C., Hofff, T., Chrousosa, G. (2002). Chronic Hypercortisolemia Inhibits Dopamine Synthesis and Turnover in the Nucleus accumbens: An in vivo Microdialysis Study Vol. 76, No. 3.Pacaka, P., Tjurminab, O., Palkovitsc, M., Goldsteind, D., Kocha, C., Hofff, T., Chrousosa, G. (2002). Chronic Hypercortisolemia Inhibits Dopamine Synthesis and Turnover in the Nucleus accumbens: An in vivo Microdialysis Study, Neuroendocrinology , 76(3), 148- 157. doi: 10.1159/000064522. Panossiana, L., Porterb, V., Valenzuelaa, H., Zhua, X., Rebackb, E., Mastermanb, D., Cummingsb, J., Effrosac, R, (2003). Telomere shortening in T cells correlates with Alzheimer’s disease status. Neurobiology of Ageing, 24(1), 77-84. PMID: 12493553. Penza, K., Heim, C., and Nemeroff, C. (2003). Neurobiological effects of childhood abuse: implications for the pathophysiology of depression and anxiety. Archives of Women's Mental Health. 6(1), 15 - 22. doi:10.1007/s00737-002-0159-x Peskind, E. R., Wilkinson, C. W., Petrie, E. C., Schellenberg, G.D., & Raskind, M. A.(2001). Increased CSF cortisol in AD is a function of APOE genotype. Neurology. 56(8), 1094-8. PMID: 11320185. Popp, J., Schaper, K., Kölsch,H., Cvetanovska, G., Rommel, F., Klingmüller, D., Dodel, R., Wüllner, U., & Jessen, F. (2009). CSF cortisol in Alzheimer's disease and mild cognitive impairment. Neurobiology of Aging, 30(3), 498-500. PMID: 17716786.
  • 48. 48 Praissman, S. (2008). Mindfulness-based stress reduction: A literature review and clinician’s guide. Journal of the American Academy of Nurse Practitioners, 20(4), 212–216. DOI: 10.1111/j.1745-7599.2008.00306.x Prigogine, I., Holte, J.,(1993). Chaos: the new science. University Press of America, Inc. Lanham: Maryland. Pryor W. A. (2000). Vitamin E and heart disease: basic science to clinical intervention trials. Free Radical Biological Medicine, 28(1), 141–64. doi:10.1016/S0891-5849(99)00224-5 Rohn, Head, Nesse, Cotman, & Cribbs, (2001). Activation of Caspase-8 in the Alzheimer's Disease Brain. Neurobiology of l Disease, 8(6), 1006-1016. PMID: 11741396. Rudolph, K., Chang, S., Han-Woong, L., Blasco, M., Gottlieb, G., Greider, C., & DePinho, R. (1999). Longevity, Stress Response, and Cancer in Aging Telomerase-Deficient Mice. Cell Press, 96, 701–712. PMID: 10089885. Schafer, F.Q., Buettner, G. R. (2001). Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radical Biological Medicine, 30(11), 1191–212. doi:10.1016/S0891-5849(01)00480-4 Schaafsma, J. D., Balash, Y., Gurevich, T., Bartels, A. L., Hausdorff, J. M., & Giladi, N. (2003). Characterization of freezing of gait subtypes and the response of each to levodopa in Parkinson's disease. European Journal of Neurology, 10(4), 391 – 398. doi:10.1046/j.1468-1331.2003.00611.x. Schore, A. (2001). The effects of early relational trauma on right brain development, affect regulation, and infant mental health. Infant Mental Health Journal, 22(1-2), 201–269. doi:10.1002/1097-0355(200101/04)22:1<201::AID.
  • 49. 49 Serrano, M., & Blasco, M. (2001). Putting the stress on senescence. Current Opinion in Cell Biology, 13(6-1), 748-753. PMID: 11698192. Seuberta, W., Henninga, H. V., Schonera, W., & L'agea, M. (1968). Effects of cortisol on the levels of metabolites and enzymes controlling glucose production from pyruvate. Advances in Enzyme Regulation, 6, 153-187. doi:10.1016/0065-2571(68)90012-5. Shank, C., Awschalom, D., Murphy, D., Bawendi, M., Stupp, S., Fréchet, J., Wolynes, P. (1999). A Workshop on Complex Systems: Lawrence Berkeley National Laboratory Shea, A., Walsh, C., MacMillan, H., and Steiner, M. (2004). Child maltreatment and HPA axis dysregulation: relationship to major depressive disorder and post traumatic stress disorder in females. Psychoneuroendocrinology, 30(2), 162-78. PMID: 15471614. Shousea, M. & Ryan, W. (1984). Thalamic kindling: Electrical stimulation of the lateral geniculate nucleus produces photosensitive grand mal seizures. Experimental Neurology, 86(1), 18-32. PMID: 6434340. Starkman, M., Giordani, B., Gebarskic, S., Berent, S., Schork, A., Schteingart, D. (1999). Decrease in cortisol reverses human hippocampal atrophy following treatment of Cushing’s disease. Brain Research Reviews, 46(12), 1595-1602. ISSN:0006-3223. Starkman, M., and Schteingart, D. (1981). Neuropsychiatric Manifestations of Patients With Cushing's Syndrome: Relationship to Cortisol and Adrenocorticotropic Hormone Levels. Archive of Internal Medicine 141, 215 - 219. PMID: 6257194. Sohal, R. S. (2002). Role of oxidative stress and protein oxidation in the aging process. Free Radical Biology Medicine, 33(1), 37–44. doi:10.1016/S0891-5849(02)00856-0.
  • 50. 50 Solertea, S., Fioravantia, M., Racchib, M., Trabucchic, M., Zanettib, O., & Govonid, S. (1999). Menopause and estrogen deficiency as a risk factor in dementing illness: hypothesis on the biological basis. Maturitas, 31(2), 95-101. ISSN:0378-5122. Sorg, C., Riedl, V., Mühlau, M., Calhoun, V., Eichele, T., Läer, L., Drzezga, L., Förstl, H., Kurtz, A., Zimmer, C., & Wohlschläger, A. (2007). Selective changes of resting-state networks in individuals at risk for Alzheimer's disease. PNAS, 104(47), 18760-18765. doi: 10.1073/pnas.0708803104. Sousaa, N., Cerqueira, J., & Osborne, A. (2008). Corticosteroid receptors and neuroplasticity. Brain Research Reviews, 57(2), 561-570. PMID: 17692926. Tang, Y., Yinghua Ma, Y., Wang, J., Fan, Y., Feng, S., Lu, Q., Yu, Q., Sui, D., Rothbart, M. K., Fan, M., & Posner. M. (2007). Short-term meditation training improves attention and self-regulation. PNAS, 104(43), 17152-17156. doi: 10.1073/pnas.0707678104 Toussaint, O., Royer, V., Salmon, M., & Remacle, J. (2002). Stress-induced premature senescence and tissue. Biochemical Pharmacology, 64(5-6), 1007-1009. doi:10.1016/S0006-2952(02)01170-X. Trump, B., Berezesky, I., Chang, S., Phelps, P. (1997). USA The Pathways of Cell Death: Oncosis, Apoptosis, and Necrosis. Toxicologic Pathology, 25(1), 82-88. doi: 10.1177/019262339702500116. Ulas, J., Weihmuller, L., Brunner, L., Joyce, J., Marshall, J., and Cotman, C. (1994). Selective increase of NMDA-sensitive glutamate binding in the striatum of Parkinson's disease, Alzheimer's disease, and mixed Parkinson's disease/Alzheimer's disease patients: an autoradiographic study. Journal of Neuroscience, 14, 6317-6324. ISSN:0270-6474
  • 51. 51 Valko, M., Morris, H., & Cronin, M. T. (May 2005). Metals, toxicity and oxidative stress. Current Medical Chemestry, 12 (10), 1161–208. doi:10.2174/0929867053764635. PMID 15892631. Van Oersa, H., Kloetb, R., & Levinea, S. (1998). Early vs. late maternal deprivation differentially alters the endocrine and hypothalamic responses to stress. Developmental Brain Research 111(2), 245-252. doi:10.1016/S0165-3806(98)00143-6. Vazquez, J. & Baghdoyan, H. A. (2001). Basal forebrain acetylcholine release during REM sleep is significantly greater than during waking. American Journal of Physiological Regulatory Integrative and Comparitive Physiology. 280, R598-R601. PMID:11208592. Vegetoa, E., Benedusia, V., & Maggi, A. (2008). Estrogen anti-inflammatory activity in brain: A therapeutic opportunity for menopause and neurodegenerative diseases. Frontiers in Neuroendocrinology, 29(4), 507-519. doi:10.1016/j.yfrne.2008.04.001. von Zglinicki, T., (2002). Oxidative stress shortens telomeres. Trends Biochememical Science, 27(7), 339-44. PMID:12114022. Wall, J. T., Xu, J. & Wang. X. (2002). Human brain plasticity: an emerging view of the multiple substrates and mechanisms that cause cortical changes and related sensory dysfunctions after injuries of sensory inputs from the body. Brain Research Reviews, 39(2-3), 181-215. doi:10.1016/S0165-0173(02)00192-3. Wang, S., Oelze, B., & Axel Schumacher, A. (2008). Age-Specific Epigenetic Drift in Late- Onset Alzheimer's Disease. PLoS ONE, 3(7). doi: 10.1371/journal.pone.0002698. Weaver, I. C. G., Grant, R. J., & Meaney, M. J. (2002). Maternal behavior regulates long-term hippocampal expression of BAX and apoptosis in the offspring. Journal of Neurochemistry, 82(4), 998–1002. doi:10.1046/j.1471-4159.2002.01054.x
  • 52. 52 Weng, N., Levine, B., June, C., and Hodes, R. (1996). Regulated expression of telomerase activity in human T lymphocyte development and activation. Journal of Experimental Medicine, 183, 2471-2479. PMCID: PMC2192611. Wüst, S., Federenkoa, I., Hellhammera, D., & Kirschbaumb, C. (2000). Genetic factors, perceived chronic stress, and the free cortisol response to awakening. Psychoneuroendocrinology, 25(7), 707-720. doi:10.1016/S0306-4530(00)00021-4. Yehuda, R., Teicher, M. H., Seckl, J., Grossman, R., Morris, A., Bierer, L. (2007). Parental Posttraumatic Stress Disorder as a Vulnerability Factor for Low Cortisol Trait in Offspring of Holocaust Survivors. Archives of General Psychiatry, 64(9), 1040-1048. PMID: 17768269. Yehuda, R., Teicherbc, M., Trestmana, R., Levengooda, R., Sievera, L., (1996). Cortisol regulation in posttraumatic stress disorder and major depression: A chronobiological analysis. Biological Psychiatry, 40(2), 79-88. PMID: 8793040. Yoshinoa, A., Hovda, D., Kawamata, T., Katayama, Y., & Beckera, D. (1991). Dynamic changes in local cerebral glucose utilization following cerebral concussion in rats: evidence of a hyper- and subsequent hypometabolic state. Brain Research, 561(1), 106-119. PMID: 1797338. Young, E., Abelsona, J., Camerona, O. (2003). Effect of comorbid anxiety disorders on the Hypothalamic-Pituitary-Adrenal axis response to a social stressor in major depression. Biological Psychiatry, 56(2), 113-20. PMID: 15231443. Zulli, R., Nicosia, F., Borroni, B., Agosti, C., Prometti, P., Donati, P., Vecchi, M., Romanelli, G., Grassi, V., Padovani, A. (2005). Dispersion and Heart Rate Variability Abnormalities
  • 53. 53 in Alzheimer's Disease and in Mild Cognitive Impairment. Journal of the American Geriatrics Society, 53(12), 2135 – 2139. doi:10.1111/j.1532-5415.2005.00508.x.