Matching Items (11)
Filtering by

Clear all filters

152325-Thumbnail Image.png
Description
The brain is a fundamental target of the stress response that promotes adaptation and survival but the repeated activation of the stress response has the potential alter cognition, emotion, and motivation, key functions of the limbic system. Three structures of the limbic system in particular, the hippocampus, medial prefrontal cortex

The brain is a fundamental target of the stress response that promotes adaptation and survival but the repeated activation of the stress response has the potential alter cognition, emotion, and motivation, key functions of the limbic system. Three structures of the limbic system in particular, the hippocampus, medial prefrontal cortex (mPFC), and amygdala, are of special interest due to documented structural changes and their implication in post-traumatic stress disorder (PTSD). One of many notable chronic stress-induced changes include dendritic arbor restructuring, which reflect plasticity patterns in parallel with the direction of alterations observed in functional imaging studies in PTSD patients. For instance, chronic stress produces dendritic retraction in the hippocampus and mPFC, but dendritic hypertrophy in the amygdala, consistent with functional imaging in patients with PTSD. Some have hypothesized that these limbic region's modifications contribute to one's susceptibility to develop PTSD following a traumatic event. Consequently, we used a familiar chronic stress procedure in a rat model to create a vulnerable brain that might develop traits consistent with PTSD when presented with a challenge. In adult male rats, chronic stress by wire mesh restraint (6h/d/21d) was followed by a variety of behavioral tasks including radial arm water maze (RAWM), fear conditioning and extinction, and fear memory reconsolidation to determine chronic stress effects on behaviors mediated by these limbic structures. In chapter 2, we corroborated past findings that chronic stress caused hippocampal CA3 dendritic retraction. Importantly, we present new findings that CA3 dendritic retraction corresponded with poor spatial memory in the RAWM and that these outcomes reversed after a recovery period. In chapter 3, we also showed that chronic stress impaired mPFC-mediated extinction memory, findings that others have reported. Using carefully assessed behavior, we present new findings that chronic stress impacted nonassociative fear by enhancing contextual fear during extinction that generalized to a new context. Moreover, the generalization behavior corresponded with enhanced functional activation in the hippocampus and amygdala during fear extinction memory retrieval. In chapter 5, we showed for the first time that chronic stress enhanced amygdala functional activation during fear memory retrieval, i.e., reactivation. Moreover, these enhanced fear memories were resistant to protein synthesis interference to disrupt a previously formed memory, called reconsolidation in a novel attempt to weaken chronic stress enhanced traumatic memory. Collectively, these studies demonstrated the plastic and dynamic effects of chronic stress on limbic neurocircuitry implicated in PTSD. We showed that chronic stress created a structural and functional imbalance across the hippocampus, mPFC, and amygdala, which lead to a PTSD-like phenotype with persistent and exaggerated fear following fear conditioning. These behavioral disruptions in conjunction with morphological and functional imaging data reflect a chronic stress-induced imbalance between hippocampal and mPFC regulation in favor of amygdala function overdrive, and supports a novel approach for traumatic memory processing in PTSD.
ContributorsHoffman, Ann (Author) / Conrad, Cheryl D. (Thesis advisor) / Olive, M. Foster (Committee member) / Hammer, Jr., Ronald P. (Committee member) / Sanabria, Federico (Committee member) / Arizona State University (Publisher)
Created2013
152286-Thumbnail Image.png
Description
Chronic restraint stress impairs hippocampal-mediated spatial learning and memory, which improves following a post-stress recovery period. Here, we investigated whether brain derived neurotrophic factor (BDNF), a protein important for hippocampal function, would alter the recovery from chronic stress-induced spatial memory deficits. Adult male Sprague-Dawley rats were infused into the hippocampus

Chronic restraint stress impairs hippocampal-mediated spatial learning and memory, which improves following a post-stress recovery period. Here, we investigated whether brain derived neurotrophic factor (BDNF), a protein important for hippocampal function, would alter the recovery from chronic stress-induced spatial memory deficits. Adult male Sprague-Dawley rats were infused into the hippocampus with adeno- associated viral vectors containing the coding sequence for short interfering (si)RNA directed against BDNF or a scrambled sequence (Scr), with both containing the coding information for green fluorescent protein to aid in anatomical localization. Rats were then chronically restrained (wire mesh, 6h/d/21d) and assessed for spatial learning and memory using a radial arm water maze (RAWM) either immediately after stressor cessation (Str-Imm) or following a 21-day post-stress recovery period (Str-Rec). All groups learned the RAWM task similarly, but differed on the memory retention trial. Rats in the Str-Imm group, regardless of viral vector contents, committed more errors in the spatial reference memory domain than did non-stressed controls. Importantly, the typical improvement in spatial memory following recovery from chronic stress was blocked with the siRNA against BDNF, as Str-Rec-siRNA performed worse on the RAWM compared to the non-stressed controls or Str-Rec-Scr. These effects were specific for the reference memory domain as repeated entry errors that reflect spatial working memory were unaffected by stress condition or viral vector contents. These results demonstrate that hippocampal BDNF is necessary for the recovery from stress-induced hippocampal dependent spatial memory deficits in the reference memory domain.
ContributorsOrtiz, J. Bryce (Author) / Conrad, Cheryl D. (Thesis advisor) / Olive, M. Foster (Committee member) / Taylor, Sara (Committee member) / Bimonte-Nelson, Heather A. (Committee member) / Arizona State University (Publisher)
Created2013
135225-Thumbnail Image.png
Description
Monoamine neurotransmitters (e.g., serotonin, norepinephrine, and dopamine) are powerful modulators of mood and cognitive function in health and disease. We have been investigating the modulation of monoamine clearance in select brain regions via organic cation transporters (OCTs), a family of nonselective monoamine transporters. OCTs are thought to complement the actions

Monoamine neurotransmitters (e.g., serotonin, norepinephrine, and dopamine) are powerful modulators of mood and cognitive function in health and disease. We have been investigating the modulation of monoamine clearance in select brain regions via organic cation transporters (OCTs), a family of nonselective monoamine transporters. OCTs are thought to complement the actions of selective monoamine transporters in the brain by helping to clear monoamines from the extracellular space; thus, assisting to terminate the monoamine signal. Of particular interest, stress hormones (corticosterone; CORT) inhibit OCT3-mediated transport of monoamine, to putatively lead to prolonged monoamine signaling. It has been demonstrated that stress levels of CORT block OCT3 transport in the rat hypothalamus, an effect that likely underlies the rapid, stress-induced increase in local monoamines. We examined the effect of chronic variable stress (CVS) on the development of mood disorders and OCT3 expression in limbic and hypothalamic regions of the rat brain. Animals subjected to CVS (14-days with random stressor exposure two times/day) showed reduced body weight gain, indicating that CVS was perceived as stressful. However, behavioral tests of anxiety and depressive-like behaviors in rats showed no group differences. Although there were no behavioral effects of stress, molecular analysis revealed that there were stress-related changes in OCT3 protein expression. In situ hybridization data confirmed that OCT3 mRNA is expressed in the hippocampus, amygdala, and hypothalamus. Analysis of Western blot data by two-way ANOVA revealed a significant treatment effect on OCT3 protein levels, with a significant decrease in OCT3 protein in the amygdala and hippocampus in CVS rats, compared to controls. These data suggest an important role for CORT sensitive OCT3 in the reduction of monoamine clearance during stress.
ContributorsBoyll, Piper Savannah (Author) / Orchinik, Miles (Thesis director) / Conrad, Cheryl (Committee member) / Talboom, Joshua (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2016-05
135343-Thumbnail Image.png
Description
The stress response facilitates our ability to deal effectively with threatening situations, but exposure to severe or chronic stressors can lead to undesirable neural, physiological, and behavioral outcomes. Chronic stress is associated with structural changes in the rat hippocampus, with corresponding deficits in learning and memory. Recent studies have uncovered

The stress response facilitates our ability to deal effectively with threatening situations, but exposure to severe or chronic stressors can lead to undesirable neural, physiological, and behavioral outcomes. Chronic stress is associated with structural changes in the rat hippocampus, with corresponding deficits in learning and memory. Recent studies have uncovered an inherent neuroplasticity that allows the hippocampus to recover from these stress-induced neural changes. Underlying mechanisms likely involve several different cellular and molecular pathways. In order to gain a more comprehensive understanding of these pathways, we investigated differences in protein expression throughout the timeline of chronic stress and recovery. Male Sprague-Dawley rats were randomly assigned to chronic restraint stress for 6hr/d/10d or 6hr/d/21d, stress for 6hr/d/21d followed by a recovery period of no stress for 10 or 21 days, or a control group. The proteome from the hippocampus of these rats was sequenced using liquid chromatography tandem mass spectrometry (LC-MS/MS) and analyzed. We hypothesized that chronic stress alters interneuronal signaling in the hippocampus by enhancing or attenuating the expression of proteins responsible for synaptic plasticity (functional) and neuronal structure (morphology). So far we have found that structural proteins, such as alpha-internexin, homer protein homolog 3, neurofilament light, and vimentin were significantly altered by chronic stress and recovery. In contrast, proteins necessary for or associated with myelination such as 2',3'-cyclic-nucleotide 3'-phosphodiesterase, myelin-associated glycoprotein, myelin basic protein S, and myelin proteolipid protein were significantly downregulated by chronic stress. Collectively, these results will provide a resource for further investigations into the mechanisms of the brain's recovery from chronic stress.
ContributorsKachemov, Marketta Marilyn (Author) / Orchinik, Miles (Thesis director) / Pirrotte, Patrick (Committee member) / Conrad, Cheryl (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2016-05
134581-Thumbnail Image.png
Description
Chronic stress often leads to cognitive deficits, especially within the spatial memory domain mediated by the hippocampus. When chronic stress ends and a no-stress period ensues (i.e., washout, WO), spatial ability improves, which can be better than non-stressed controls (CON). The WO period is often the same duration as the

Chronic stress often leads to cognitive deficits, especially within the spatial memory domain mediated by the hippocampus. When chronic stress ends and a no-stress period ensues (i.e., washout, WO), spatial ability improves, which can be better than non-stressed controls (CON). The WO period is often the same duration as the chronic stress paradigm. Given the potential benefit of a post-stress WO period on cognition, it is important to investigate whether this potential benefit of a post-stress WO period has long-lasting effects. In this project, chronic restraint (6hr/d/21d) in Sprague-Dawley rats was used, as it is the minimum duration necessary to observe spatial memory deficits. Two durations of post-stress WO were used following the end of chronic restraint, 3 weeks (STR-WO3) and 6 weeks (STR-WO6). Immediately after chronic stress (STR-IMM) or the WO periods, rats were tested on various cognitive tests. We corroborated past studies that chronic stress impaired spatial memory (STR-IMM vs CON). Interestingly, STR-WO3 and STR-WO6 failed to demonstrate improved spatial memory on a radial arm water maze task, performing similarly as STR-IMM. Performance outcomes were unlikely from differences in anxiety or motivation because rats from all conditions performed similarly on an open field task and on a simple object recognition paradigm, respectively. However, performance on object placement was unusual in that very few rats explored, suggesting some degree of anxiety or fear in all groups. One possible interpretation of the unusual results of the 3 week washout group may be attributed to the different spatial memory tasks used across studies or external factors from the study. Further exploration of these other factors led to the conclusion that they did not play a role and the STR-WO3 RAWM data were anomalous to other studies. This suggests that a washout period following chronic stress may not be fully understood.
ContributorsFlegenheimer, Aaron Embden (Author) / Conrad, Cheryl (Thesis director) / Bimonte-Nelson, Heather (Committee member) / Ortiz, J. Bryce (Committee member) / School of Life Sciences (Contributor) / School of Human Evolution and Social Change (Contributor) / Barrett, The Honors College (Contributor)
Created2017-05
134278-Thumbnail Image.png
Description
The RAS/MAPK (RAS/Mitogen Activated Protein Kinase) pathway is a highly conserved, canonical signaling cascade that is highly involved in cellular growth and proliferation as well as cell migration. As such, it plays an important role in development, specifically in development of the nervous system. Activation of ERK is indispensable for

The RAS/MAPK (RAS/Mitogen Activated Protein Kinase) pathway is a highly conserved, canonical signaling cascade that is highly involved in cellular growth and proliferation as well as cell migration. As such, it plays an important role in development, specifically in development of the nervous system. Activation of ERK is indispensable for the differentiation of Embryonic Stem Cells (ESC) into neuronal precursors (Li z et al, 2006). ERK signaling has also shown to mediate Schwann cell myelination of the peripheral nervous system (PNS) as well as oligodendrocyte proliferation (Newbern et al, 2011). The class of developmental disorders that result in the dysregulation of RAS signaling are known as RASopathies. The molecular and cell-specific consequences of these various pathway mutations remain to be elucidated. While there is evidence for altered DNA transcription in RASopathies, there is little work examining the effects of the RASopathy-linked mutations on protein translation and post-translational modifications in vivo. RASopathies have phenotypic and molecular similarities to other disorders such as Fragile X Syndrome (FXS) and Tuberous Sclerosis (TSC) that show evidence of aberrant protein synthesis and affect related pathways. There are also well-defined downstream RAS pathway elements involved in translation. Additionally, aberrant corticospinal axon outgrowth has been observed in disease models of RASopathies (Xing et al, 2016). For these reasons, this present study examines a subset of proteins involved in translation and translational regulation in the context of RASopathy disease states. Results indicate that in both of the tested RASopathy model systems, there is altered mTOR expression. Additionally the loss of function model showed a decrease in rps6 activation. This data supports a role for the selective dysregulation of translational control elements in RASopathy models. This data also indicates that the primary candidate mechanism for control of altered translation in these modes is through the altered expression of mTOR.
ContributorsHilbert, Alexander Robert (Author) / Newbern, Jason (Thesis director) / Olive, M. Foster (Committee member) / Bjorklund, Reed (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2017-05
134855-Thumbnail Image.png
Description
Chronic stress impairs spatial working memory, attention set-shifting, and response inhibition. The relationship between these functions and the potential underlying neurocircuitry, such as the medial prefrontal cortex (mPFC), needs further research to understand how chronic stress impacts these functions. This study focused on the infralimbic (IL) and prelimbic (PRL) regions

Chronic stress impairs spatial working memory, attention set-shifting, and response inhibition. The relationship between these functions and the potential underlying neurocircuitry, such as the medial prefrontal cortex (mPFC), needs further research to understand how chronic stress impacts these functions. This study focused on the infralimbic (IL) and prelimbic (PRL) regions of the mPFC, to examine its involvement in two behavioral tasks, fixed minimum interval (FMI) and radial arm water maze (RAWM), following chronic stress, and the relationship between the two paradigms. A previous study failed to find a significant correlation between spatial working memory and response, both functions mediated by the PFC, even though chronic stress disrupted both outcomes. Thus, the purpose of this study was to investigate the functional activation of the mPFC, following chronic stress in these two paradigms, in order to gain an understanding of the neurocircuitry involved within this region. The behavioral outcomes were performed prior to my involvement in the project, and the results corroborate previous findings that chronic stress impairs response inhibition on FMI and spatial working memory on RAWM. My honors thesis involved quantifying the immunohistochemistry-stained tissue to assess the functional activation of the mPFC. Over the course of six months, my work involved identifying the border between IL and PRL regions by overlaying captured images of tissues, starting at a lower magnification of 40x. Afterwards, images were recaptured at higher magnifications (100x) to quantify Fos-like counts of functional activation. No overt changes were found following chronic stress in Fos-like counts after performance on FMI or RAWM. However, response inhibition on the FMI task showed a relationship with the IL function; non-stressed rats displayed a positive correlation between response inhibition and Fos-like profiles. In contrast, chronically stressed rats revealed a negative correlation between response inhibition and Fos-like profiles. The IL cortex is revealed to facilitate extinction of a learned behavior. Thus, these results present a possible interpretation that there is an association, non-stressed rats suppressing a previously learned response, being formed.
ContributorsLe, Brittany Quynh (Author) / Conrad, Cheryl (Thesis director) / Sanabria, Federico (Committee member) / Judd, Jessica (Committee member) / School of Molecular Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2017-05
134795-Thumbnail Image.png
Description
Chronic restraint stress leads to apical dendritic retraction in CA3 pyramidal neurons and often no quantifiable changes in CA1 dendritic complexity. When chronic stress ends, a post-stress recovery period results in an enhancement in CA3 dendritic complexity. We investigated the relationship between CA3 and CA1 pyramidal neurons to determine whether

Chronic restraint stress leads to apical dendritic retraction in CA3 pyramidal neurons and often no quantifiable changes in CA1 dendritic complexity. When chronic stress ends, a post-stress recovery period results in an enhancement in CA3 dendritic complexity. We investigated the relationship between CA3 and CA1 pyramidal neurons to determine whether dendritic restructuring in CA3 neurons leads to region-specific changes in the dendritic complexity of CA1 neurons. Adult male Sprague-Dawley rats were restrained (wire mesh, 6h/d/21d) and brains were removed soon after restraint ended (Str-Imm) or after a 21d post-stress recovery period (Str-Rec). In addition, BDNF downregulation targeting the CA3 region prevents enhancement in dendritic complexity following recovery in chronically stressed rats, providing robust conditions to investigate the CA3-CA1 relationship. Consequently, rats were infused into the CA3 area with either an AAV vector with a coding sequence against BDNF (shRNA) or a sequence with no known mRNA complements (Scr). Apical and basal dendritic complexity of CA3 and CA1 was quantified by counting total dendritic bifurcations and dendritic intersections using the Sholl analysis (20 µm distances from soma). Please note that the quantification of the CA3 dendritic arbors was not part of this thesis project. The outcome of that investigation revealed that apical CA3 dendritic retraction was found in Str-Imm-Scr and Str-Rec-shRNA. For the CA1 apical area, gross dendritic bifurcation differences were not detected, but the Sholl quantification revealed regionally-enhanced dendritic complexity that varied by distance from the soma at the distal apical dendrites (Str-Imm-Scr) and proximal basal dendrites (Str-Rec-shRNA). For the latter, significant increases in basal branch points were detected with total branch point quantification method. Moreover, a correlation using all groups revealed a significant inverse relationship between CA3 apical dendritic complexity and CA1 basal dendritic complexity. The results demonstrate that chronic stress-induced CA3 apical dendritic retraction may relate to region-specific changes in CA1 dendritic complexity. The inability of past studies to detect changes in CA1 dendritic complexity may be due to the shortcoming of gross dendritic arbor measures in accounting for subtle region-specific alterations. To address this, the current study included a cohort with BDNF downregulated in the CA3 region. Overall, this suggests that decreased levels of BDNF in the hippocampus provide robust conditions in which changes to CA1 dendritic complexity can be detected.
ContributorsDaas, Eshaan Jatin (Author) / Conrad, Cheryl (Thesis director) / Orchinik, Miles (Committee member) / Ortiz, J. Bryce (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2016-12
153854-Thumbnail Image.png
Description
Evidence from the 20th century demonstrated that early life stress (ELS) produces long lasting neuroendocrine and behavioral effects related to an increased vulnerability towards psychiatric illnesses such as major depressive disorder, post-traumatic stress disorder, schizophrenia, and substance use disorder. Substance use disorders (SUDs) are complex neurological and behavioral psychiatric illnesses.

Evidence from the 20th century demonstrated that early life stress (ELS) produces long lasting neuroendocrine and behavioral effects related to an increased vulnerability towards psychiatric illnesses such as major depressive disorder, post-traumatic stress disorder, schizophrenia, and substance use disorder. Substance use disorders (SUDs) are complex neurological and behavioral psychiatric illnesses. The development, maintenance, and relapse of SUDs involve multiple brain systems and are affected by many variables, including socio-economic and genetic factors. Pre-clinical studies demonstrate that ELS affects many of the same systems, such as the reward circuitry and executive function involved with addiction-like behaviors. Previous research has focused on cocaine, ethanol, opiates, and amphetamine, while few studies have investigated ELS and methamphetamine (METH) vulnerability. METH is a highly addictive psychostimulant that when abused, has deleterious effects on the user and society. However, a critical unanswered question remains; how do early life experiences modulate both neural systems and behavior in adulthood? The emerging field of neuroepigenetics provides a potential answer to this question. Methyl CpG binding protein 2 (MeCP2), an epigenetic tag, has emerged as one possible mediator between initial drug use and the transition to addiction. Additionally, there are various neural systems that undergo long lasting epigenetics changes after ELS, such as the response of the hypothalamo-pituitary-adrenal (HPA) axis to stressors. Despite this, little attention has been given to the interactions between ELS, epigenetics, and addiction vulnerability. The studies described herein investigated the effects of ELS on METH self-administration (SA) in adult male rats. Next, we investigated the effects of ELS and METH SA on MeCP2 expression in the nucleus accumbens and dorsal striatum. Additionally, we investigated the effects of virally-mediated knockdown of MeCP2 expression in the nucleus accumbens core on METH SA, motivation to obtain METH under conditions of increasing behavioral demand, and reinstatement of METH-seeking in rats with and without a history of ELS. The results of these studies provide insights into potential epigenetic mechanisms by which ELS can produce an increased vulnerability to addiction in adulthood. Moreover, these studies shed light on possible novel molecular targets for treating addiction in individuals with a history of ELS.
ContributorsLewis, Candace (Author) / Olive, M. Foster (Thesis advisor) / Hammer, Ronald (Committee member) / Neisewander, Janet (Committee member) / Sanabria, Federico (Committee member) / Arizona State University (Publisher)
Created2015
131967-Thumbnail Image.png
Description
Women are twice as likely as men to develop Major Depressive Disorder (MDD), and current MDD therapies are only effective for about a third of patients. Hormonal transitions, specifically those involving estradiol (E2), have been found to contribute to this increased vulnerability in women. This study aimed to investigate potential

Women are twice as likely as men to develop Major Depressive Disorder (MDD), and current MDD therapies are only effective for about a third of patients. Hormonal transitions, specifically those involving estradiol (E2), have been found to contribute to this increased vulnerability in women. This study aimed to investigate potential mechanisms underlying the sex differences seen in MDD vulnerability, specifically the role of E2. The brain region-specific changes induced by chronic stress differ for female rats than for male rats. Therefore, we aimed to determine the effects of sex and chronic stress on E2 expression in four brain regions: the hippocampus, medial prefrontal cortex, amygdala, and cerebellum. Sprague-Dawley rats (n = 48, 24 males, 24 females; n=12/Tx group) were subjected to daily wire mesh restraint stress (6 h/21 days), and were euthanized and dissected the day following the end of chronic restraint stress (day 22). Ultra high-pressure liquid chromatography-mass spectroscopy was used to directly measure E2 in the brain regions. Quantitative real-time PCR was used to indirectly assess E2 expression via mRNA for aromatase (ARO-L) and estrogen receptors (ERβ, ERɑ, and GPR30), as well as expression of inflammatory cytokines (IL-1β and TNF-ɑ). Our findings suggest that chronic stress may lead to changes in local estradiol expression in the brain that are both sex-dependent and brain region-specific, while the data are preliminary given the small sample size. We found that expression of ARO-L mRNA, a measure of local E2 production, tended to increase in the HIPP, but decrease in the mPFC following chronic stress, and in the mPFC this pattern was only observed in males. Local estradiol production in the brain seems to act as a potential compensatory mechanism in the hippocampus, but as a protective mechanism in the mPFC, which is highly sensitive to chronic stress.
ContributorsSmith, Elliot Ann (Author) / Conrad, Cheryl (Thesis director) / Presson, Clark (Committee member) / Department of Psychology (Contributor) / Department of Physics (Contributor) / Barrett, The Honors College (Contributor)
Created2020-05