• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 4
  • 2
  • 2
  • Tagged with
  • 10
  • 7
  • 6
  • 5
  • 4
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Investigating the role of autism gene Ube3a in the transcriptional regulation in neurons

Magsino, Emmanuel 17 June 2016 (has links)
UBE3A is a protein with dual functions as an E3 ubiquitin ligase and as a Steroid Hormone Receptor (SHR) transcriptional coactivator. It is expressed ubiquitously in tissues and has significant importance in neurons, where it is expressed exclusively from the maternal allele. Thus, while UBE3A has a wide variety of targets in various tissues, it is especially important in the brain as UBE3A regulates several aspects of neuronal growth, function, and maintenance. Therefore deficits of Ube3a cause Angelman Syndrome (AS) and increased dosage causes Autism Spectrum Disorder (ASD), two neurological disorders. The pathological phenotype of both diseases involves behavioral dysfunctions in learning, motor skills, and sociability. Through microarray studies, our laboratory has found that Ube3a is involved in the regulation of neuronal proteins such as CBLN1, which has been found to have significant importance in parallel fiber synapse formation onto Purkinje cells. To determine how regulation of CBLN1 occurs, mutant variants of human Ube3a isoform III were then generated. E3 ligase-dead, substrate-binding defective, nonphosphorylatable mutant, and phosphor-mimetic mutants were produced and inserted into a pLVX-IRES-mCherry vector. A quantitative transcriptional analysis demonstrated that increasing wild-type (WT) UBE3A decreased Cbln1 expression. The ligase dead mutant mimicked the WT suggesting that E3 ligase activity is not required in the regulation of Cbln1. The nonphosphorylatable mutant demonstrated an increase in Cbln1 expression, which may be due to a dominant negative effect on native UBE3A causing its degradation. The phosphor-mimetic mutant had no statistical effect. This may be due to its inability to enter the nucleus and affect transcription. The substrate-binding mutant also showed no statistical effect possibly because of its inability to bind to any substrate and that may be necessary to regulate transcription. These preliminary results demonstrate that UBE3A may be regulating CBLN1 at the transcriptional level independent of its E3 ubiquitin ligase function. Future studies will be required to more precisely determine the mechanisms involved in UBE3A’s regulation of CBLN1. / 2018-06-16T00:00:00Z
2

Efficacy of Increased Ube3a Protein Levels in the Brain in Rescuing the Phenotype of an Angelman Syndrome Mouse

Daily, Jennifer L. 01 January 2012 (has links)
Angelman syndrome (AS), a genetic disorder occurring in approximately one in every 15,000 births, is characterized by severe mental retardation, seizures, difficulty speaking and ataxia. The gene responsible for AS was discovered to be UBE3A and encodes an E6-AP ubiquitin ligase. A unique feature of this gene is that it undergoes maternal imprinting in a neuron-specific manner. In the majority of AS cases, there is a mutation or deletion in the maternally inherited UBE3A gene, although other cases are the result of uniparental disomy or mismethylation of the maternal gene. While most human disorders characterized by severe mental retardation involve abnormalities in brain structure, no gross anatomical changes are associated with AS. Although it was previously believed that UBE3A was imprinted in a brain region-specific manner, primarily in the hippocampus and cortex, recent evidence indicates that there is a widespread knockdown of Ube3a protein throughout the AS mouse brain. As a result, it became necessary to evaluate AS human brain samples to verify the relevance and accuracy of the AS mouse model. It was determined that Ube3a is deficient throughout all major brain regions in humans with AS. The remainder of this dissertation work was focused on determining if increased UBE3A expression in the AS mouse brain would be sufficient to rescue the AS phenotype. The results show that adeno-associated virus-mediated UBE3A delivery is not effective in the AS neonatal brain. In the adult AS mouse brain, however, it increased Ube3a in the hippocampus to near wild-type levels. This was sufficient to rescue the associative fear conditioning learning deficit in the AS mouse and improve learning and memory in the Morris water maze. These studies are the first to demonstrate that increased protein production in the adult AS mouse is sufficient to improve the AS phenotype, indicating that the symptoms of AS are not necessarily embryonic developmental.
3

Role of CBLN1’S RE-1 transcriptional regulatory sequences in gene repression

Cruz, Tristan 12 July 2017 (has links)
BACKGROUND: Cbln1 is a gene whose expression is negatively correlated to seizures. Krishnan et al. has recently shown that seizures synergize with transcriptional co-regulator Ube3a to repress Cbln1 expression, which ultimately manifests as ASD associated behavioral discrepancies. (Krishnan et al., 2017) Seizures increase the expression of REST and the Cbln1 gene contains an intronic RE-1 binding site previously shown to interact with REST. This could therefore be a point of convergence for the transcriptional downregulation of Cbln1. OBJECTIVE: To determine if Cbln1’s RE-1 sequences confers gene repression to minimal promoter reporter system by REST and Ube3a. METHODS: Desired RE-1 sequences from Cbln1 were subcloned into a pGL3-basic vector using specific restriction enzymes that flanked each DNA region. Specific oligonucleotide target sequences were annealed together and ligated into the plasmid vector before transfecting into live HEK293T cells. A minimal luciferase promoter with just enough sequence for the polymerase to sit was also ligated into the cassette. A luciferase assay was then conducted on the plated cells under exposure to separate testing conditions such as excess Ube3a, REST, dnREST, and various combinations of these factors to determine the effect of these TFs on gene expression controlled by Cbln1’s RE-1 site. RESULTS: REST strongly, and Ube3a weakly, repressed the minimal promoter reporter construct when Cbln1’s RE-1 sequences were added. REST occluded the repressive effects produced by Ube3a indicating that their effects are not additive or synergistic. CONCLUSION: Both REST, that is increased by seizures (Krishnan et al., 2017), and Ube3a (more weakly) can repress gene expression when Cbln1’s RE-1 binding sequences are added. These repressive effects may help explain how seizures and Ube3a can decrease Cbln1 expression that ultimately leads to reduced sociability. / 2019-10-31
4

Social Behavior and Gene Expression Disturbances in Mouse Models of Angelman Syndrome and Idic15 Autism

Stoppel, David Christopher January 2014 (has links)
Reciprocal changes in UBE3A gene dosage cause two neurodevelopmental disorders. Maternally inherited deletions of UBE3A cause Angelman syndrome, characterized by intellectual disability, motor defects, seizures, and a pathognomonic increased social motivation. Whereas maternally inherited triplications of UBE3A as in Idic15 Autism underlie decreased sociability and increased repetitive restrictive behaviors of this disorder. Identifying the cellular compartments and neuronal subtypes where excess and loss of Ube3a impair behavior is essential to understanding and potentially treating the disorders. In Chapter 2, we show that mouse models of maternal loss of UBE3A (Ube3a-mKO, Angelman syndrome) and triplication of UBE3A (Ube3a-2x, Idic15) have opposite effects on social behavioral and cortical gene expression. Social preference and social vocalizations are reduced in Ube3a-2x and increased in Ube3a-mKO mimicking the human phenotypes. Using a nuclear localizing signal tagged Ube3a transgenic mouse (Ube3a-NLS), we show that Ube3a acts in the nucleus to impair social behavior and cortical gene expression. Many of the genes reciprocally regulated by nuclear Ube3a are part of an Autism protein-interaction network. In Chapter 3, we demonstrate Ube3a-2x mice have increased aggression, an important Autism comorbidity. In contrast, maternal loss of Ube3a reduces aggression, consistent with the gregarious, amiable nature of individuals with Angelman syndrome. We then mapped the loci where increased Ube3a increases aggression. Increased aggression was observed when Ube3a was targeted to glutamatergic and vasopressinergic but not to GABAergic or oxytocinergic neurons. In Chapter 4, we show that in mice, social behavior is downregulated by social experience via Ube3a. In wild-type mice, altering their social environment strongly regulates their social behavior: individual housing causes hypersocial whereas group housing causes hyposocial behavior. In Ube3a-2x animals, group housing caused an excessive downregulation of social behavior whereas single housing fully rescued their social behavior deficits. By contrast, in Ube3a-mKO animals, the suppressive effects of group housing on social experience was largely blocked, suggesting Ube3a is required for this process. In summary, this thesis characterizes the role of UBE3A gene dosage in regulating social and aggression behaviors and identifies the subcellular compartment and neuronal subtypes where changes in Ube3a gene dosage disturb the homeostasis of these behaviors.
5

Disrupted Synaptic Transmission and Abnormal Short-term Synaptic Plasticity in an Angelman Syndrome Mouse Model

January 2017 (has links)
abstract: Angelman syndrome (AS) is a neurodevelopmental disorder characterized by developmental delays, intellectual disabilities, impaired language and speech, and movement defects. Most AS cases are caused by dysfunction of a maternally-expressed E3 ubiquitin ligase (UBE3A, also known as E6 associated protein, E6-AP) in neurons. Currently, the mechanism on how loss-of-function of the enzyme influences the nervous system development remains unknown. We hypothesize that impaired metabolism of proteins, most likely those related to E6-AP substrates, may alter the developmental trajectory of neuronal structures including dendrites, spines and synaptic proteins, which leads to disrupted activity/experience-dependent synaptic plasticity and maturation. To test this hypothesis, we conducted a detailed investigation on neuronal morphology and electrophysiological properties in the prefrontal cortex (PFC) layer 5 (L5) corticostriatal pyramidal neurons (target neurons). We found smaller soma size in the maternal Ube3a deficient mice (m-/p+; 'AS' mice) at postnatal 17-19 (P17-19), P28-35 and older than 70 days (>P70), and decreased basal dendritic processes at P28-35. Surprisingly, both excitatory and inhibitory miniature postsynaptic currents (mEPSCs and mIPSCs) decreased on these neurons. These neurons also exhibited abnormalities in the local neural circuits, short-term synaptic plasticity and AMPA/NMDA ratio: the excitatory inputs from L2/3 and L5A, and inhibitory inputs from L5 significantly reduced in AS mice from P17-19; Both the release probability (Pr) and readily-releasable vesicle (RRV) pool replenishment of presynaptic neurons of the target neurons were disrupted at P17-19 and P28-35, and the change of RRV pool replenishment maintained through adulthood (>P70). The AMPA/NMDA ratio showed abnormality in the L5 corticostriatal neurons of PFC in AS mice older than P28-35, during which it decreased significantly compared to that of age-matched WT littermates. Western Blot analysis revealed that the expression level of a key regulator of the cytoskeleton system, Rho family small GTPase cell division control protein 42 homolog (cdc42), reduced significantly in the PFC of AS mice at P28-35.These impairments of synaptic transmission and short-term synaptic plasticity may account for the impaired neuronal morphology and synaptic deficits observed in the PFC target neurons, and contribute to the phenotypes in AS model mice. The present work reveals for the first time that the E6-AP deficiency influences brain function in both brain region-specific and age-dependent ways, demonstrates the functional impairment at the neural circuit level, and reveals that the presynaptic mechanisms are disrupted in AS model. These novel findings shed light on our understanding of the AS pathogenesis and inform potential novel therapeutic explorations. / Dissertation/Thesis / Doctoral Dissertation Neuroscience 2017
6

Astrocytic roles in regulating dendritic spine maturation in UBe3A-dependent autism spectrum disorder

Gardner, Zachary V. 17 June 2023 (has links)
Autism spectrum disorders (ASDs) are a diverse class of neurodevelopmental disorders with various aberrant cellular phenotypes such as dysfunctional neurotransmission and irregular neuronal morphology. ASDs have a broad underlying genetic background with many genes linked to their etiology. UBE3A has been identified as a top gene candidate associated with ASD, and overexpression of UBE3A via copy-number variation confers hallmark ASD behaviors in humans and transgenic mice. Our previous work revealed that synapse formation was negatively affected in the Ube3A-ASD mouse model (Ube3A 2X Tg, or simply “2X Tg”). However, the cellular and molecular mechanisms underlying the synaptic dysregulation remain unknown. We sought to identify a cell-type specific mechanism by which these morphological changes were conferred. We found that selective overexpression of Ube3A in neurons failed to induce changes in dendritic spine maturation. In contrast, overexpression of Ube3A in astrocytes resulted in alterations in spines and synapses. Further, we identified thrombospondin-2 (TSP2), a secreted astrocytic glycoprotein promoting synaptogenesis and spinogenesis, is involved in the defective spine maturation. Ube3A overexpression confers a loss of transcriptional down-regulation of TSP2 in astrocytes, and the medium of astrocyte cultures with Ube3A overexpression was sufficient to trigger spine changes similar to that observed in mixed cultures that globally overexpress Ube3A. Importantly, depletion of TSP2 promoted similar loss of dendritic spine maturation, whereas supplement of TSP2 to 2X Tg astrocyte media was able to rescue the spine defects. Furthermore, overexpression of Ube3A in an astrocyte-specific manner recapitulated aberrant dendritic spine maturation as well as autism-like behaviors displayed in 2X Tg mice. Collectively, these findings reveal an astrocytic dominance in initiating ASD pathobiology at the neuronal and behavior levels.
7

Estudo de expressão do gene UBE3A em neurônios derivados de células-tronco da polpa dentária de pacientes com a síndrome de Angelman / Study of UBE3A expression in dental pulp stem cells - derived neurons from patients with Angelman syndrome

Cruvinel, Estela Mitie 22 June 2011 (has links)
Síndrome de Angelman (AS - MIM 105830) é causada pela ausência de função do gene UBE3A que codifica uma proteína ubiquitina - ligase (E6-AP). Esse gene é expresso bialelicamente em vários tecidos exceto no cérebro, onde a expressão é preferencialmente materna. O RNA anti-senso de UBE3A é considerado o regulador dessa expressão diferencial entre os alelos, e faz parte de um transcrito grande que só o alelo paterno é expresso devido ao imprinting genômico; no cérebro, esse transcrito se entende até a região anti-senso de UBE3A, mas nos demais tecidos o transcrito é menor e não engloba a região anti-senso. Este trabalho visa obter um modelo para estudo da AS. Células-tronco da polpa do dente (SHEDs) de pacientes com deleção do segmento 15q11-q13 ou mutação no gene UBE3A foram caracterizadas e submetidas à diferenciação neuronal. A diferenciação foi analisada através do estudo de RNA e proteínas para marcadores neuronais e, também, por testes funcionais. As SHEDs são células-tronco mesenquimais e constituem uma população heterogênea. Essas células ou algumas dessas células já expressam algumas proteínas neuronais ou de células excitáveis como nestina, β-tubulina III, MAP2 e proteína de canais dependentes de voltagem de sódio e potássio. Um ponto interessante é que as SHEDs apresentam baixa expressão do UBE3A anti-senso e a expressão do UBE3A nas células de pacientes é menor que 50% da expressão encontrada nas células de controles, que pode indicar a ocorrência de expressão preferencial materna desse gene em outros tipos celulares além de neurônios maduros. Quando induzidas à diferenciação neurogênica, a maioria das linhagens controles apresentou aumento da expressão de MAP2 e, principalmente, β-tubulina III; e a maioria das linhagens de pacientes com AS não apresentou aumento notável na expressão dessas proteínas, exceto uma linhagem de paciente que aumentou a expressão de β-tubulina III. As células induzidas à diferenciação apresentaram aumento estatisticamente significativo da condutância de sódio através de canais de sódio dependentes de voltagem. Com a análise de expressão de UBE3A e do UBE3A anti-senso é possível afirmar que a expressão deles não alterou com a diferenciação neuronal. Assim, é possível concluir que as células-tronco da polpa do dente, com o protocolo de diferenciação neurogênica, progrediram na via de diferenciação, mas a maioria das células não atingiu o estágio de maturação necessário para que ocorresse o imprinting do UBE3A ou a via de diferenciação não ia em direção a neurônios que apresentam imprinting do UBE3A. / Angelman syndrome (AS - MIN 105830) is caused by the loss of function of the maternal UBE3A gene, which encodes an ubiquitin protein ligase (E6-AP). UBE3A displays biallelic expression in most of tissues, but maternal predominant expression is observed in the brain. A RNA antisense that is paternally expressed in some regions in the brain is considered to be responsible for this tissue-specific imprinting; UBE3A antisense is part of a large transcript that starts at SNURF-SNRPN gene and is paternally expressed, and in the brain this transcript includes UBE3A antisense region however in other tissues this region is not included. The aim of the present study is to develop a new model for studying AS. Dental pulp stem cells (SHEDs) were characterized and differentiated by an already described protocol. SHEDs intrinsically express some neuronal proteins as nestin, β-tubulin III, MAP2 and voltage-gated sodium channels and potassium channels. Interestingly, SHEDs also present a low expression of UBE3A antisense, and UBE3A expression in cells from patients with AS is lower than 50% of the cells from normal control, so it is possible that preferential maternal expression of this gene might occur in some cells beyond mature neurons. After the neuronal differentiation, most control lineages and one lineage of AS patients had an increase of MAP2 and β-tubulin III expression. Two control lineages and most lineages from AS patients did not have a notable increase of expression of these proteins. Neuronal differentiated cells displayed an increase in conductance through voltage-gated sodium channels. Analysis of UBE3A and UBE3A antisense expression in SHEDs and cells induced to differentiate into neurons indicated no changes in their expression. Thus, after neuronal differentiation induction, dental pulp stem cells progressed through neuronal differentiation pathway. However, most cells did not reach the stage which UBE3A imprinting occurs or the neuronal differentiation is resulting in a cell that do not present UBE3A imprinting.
8

Estudo de expressão do gene UBE3A em neurônios derivados de células-tronco da polpa dentária de pacientes com a síndrome de Angelman / Study of UBE3A expression in dental pulp stem cells - derived neurons from patients with Angelman syndrome

Estela Mitie Cruvinel 22 June 2011 (has links)
Síndrome de Angelman (AS - MIM 105830) é causada pela ausência de função do gene UBE3A que codifica uma proteína ubiquitina - ligase (E6-AP). Esse gene é expresso bialelicamente em vários tecidos exceto no cérebro, onde a expressão é preferencialmente materna. O RNA anti-senso de UBE3A é considerado o regulador dessa expressão diferencial entre os alelos, e faz parte de um transcrito grande que só o alelo paterno é expresso devido ao imprinting genômico; no cérebro, esse transcrito se entende até a região anti-senso de UBE3A, mas nos demais tecidos o transcrito é menor e não engloba a região anti-senso. Este trabalho visa obter um modelo para estudo da AS. Células-tronco da polpa do dente (SHEDs) de pacientes com deleção do segmento 15q11-q13 ou mutação no gene UBE3A foram caracterizadas e submetidas à diferenciação neuronal. A diferenciação foi analisada através do estudo de RNA e proteínas para marcadores neuronais e, também, por testes funcionais. As SHEDs são células-tronco mesenquimais e constituem uma população heterogênea. Essas células ou algumas dessas células já expressam algumas proteínas neuronais ou de células excitáveis como nestina, β-tubulina III, MAP2 e proteína de canais dependentes de voltagem de sódio e potássio. Um ponto interessante é que as SHEDs apresentam baixa expressão do UBE3A anti-senso e a expressão do UBE3A nas células de pacientes é menor que 50% da expressão encontrada nas células de controles, que pode indicar a ocorrência de expressão preferencial materna desse gene em outros tipos celulares além de neurônios maduros. Quando induzidas à diferenciação neurogênica, a maioria das linhagens controles apresentou aumento da expressão de MAP2 e, principalmente, β-tubulina III; e a maioria das linhagens de pacientes com AS não apresentou aumento notável na expressão dessas proteínas, exceto uma linhagem de paciente que aumentou a expressão de β-tubulina III. As células induzidas à diferenciação apresentaram aumento estatisticamente significativo da condutância de sódio através de canais de sódio dependentes de voltagem. Com a análise de expressão de UBE3A e do UBE3A anti-senso é possível afirmar que a expressão deles não alterou com a diferenciação neuronal. Assim, é possível concluir que as células-tronco da polpa do dente, com o protocolo de diferenciação neurogênica, progrediram na via de diferenciação, mas a maioria das células não atingiu o estágio de maturação necessário para que ocorresse o imprinting do UBE3A ou a via de diferenciação não ia em direção a neurônios que apresentam imprinting do UBE3A. / Angelman syndrome (AS - MIN 105830) is caused by the loss of function of the maternal UBE3A gene, which encodes an ubiquitin protein ligase (E6-AP). UBE3A displays biallelic expression in most of tissues, but maternal predominant expression is observed in the brain. A RNA antisense that is paternally expressed in some regions in the brain is considered to be responsible for this tissue-specific imprinting; UBE3A antisense is part of a large transcript that starts at SNURF-SNRPN gene and is paternally expressed, and in the brain this transcript includes UBE3A antisense region however in other tissues this region is not included. The aim of the present study is to develop a new model for studying AS. Dental pulp stem cells (SHEDs) were characterized and differentiated by an already described protocol. SHEDs intrinsically express some neuronal proteins as nestin, β-tubulin III, MAP2 and voltage-gated sodium channels and potassium channels. Interestingly, SHEDs also present a low expression of UBE3A antisense, and UBE3A expression in cells from patients with AS is lower than 50% of the cells from normal control, so it is possible that preferential maternal expression of this gene might occur in some cells beyond mature neurons. After the neuronal differentiation, most control lineages and one lineage of AS patients had an increase of MAP2 and β-tubulin III expression. Two control lineages and most lineages from AS patients did not have a notable increase of expression of these proteins. Neuronal differentiated cells displayed an increase in conductance through voltage-gated sodium channels. Analysis of UBE3A and UBE3A antisense expression in SHEDs and cells induced to differentiate into neurons indicated no changes in their expression. Thus, after neuronal differentiation induction, dental pulp stem cells progressed through neuronal differentiation pathway. However, most cells did not reach the stage which UBE3A imprinting occurs or the neuronal differentiation is resulting in a cell that do not present UBE3A imprinting.
9

Ube3a Role in Synaptic Plasticity and Neurodevelopmental Disorders.The Lessons from Angelman Syndrome.

Filonova, Irina 13 February 2014 (has links)
Angelman Syndrome (AS) is a severe neurodevelopmental disorder that affects 1:12000 newborns. It is characterized by mental retardation, delayed major motor and cognitive milestones, seizures, absence of speech and excessive laughter. The majority of AS cases arise from deletions or mutations of UBE3A gene located on the chromosome 15q11-13. UBE3A codes for E3-ubiquitin ligase that target specific proteins for degradation. To date, a wide variety of Ube3a substrates has been identified. The accumulation of Ube3a-dependent proteins and their effect on the multitude of signal transduction pathways are` considered the main cause of the AS pathology. While the majority of research has been directed towards target identifications, the overall role of Ube3a in activity-dependent synaptic plasticity has been greatly overlooked. The present work is designed to fill some of these knowledge gaps. Chapter 2 is focused on the activity-dependent aspect of Ube3a expression following neuronal stimulation in vivo and in vitro. We examined total Ube3a expression followed by KCl depolarization in neuronal primary culture. By utilizing a subcellular fractionation technique, we were able to determine which cellular pools are responsive to the depolarization. Next, a fear conditioning paradigm (FC) was used to activate neurons in the paternal Ube3a-YFP reporter mouse brain. This mouse model allowed us to resolve spatial and temporal alterations of the maternal and the paternal Ube3a in hippocampus and cortex followed by FC. In accordance to KCl depolarization results, we observed alterations in Ube3a protein but at later time points. Furthermore, we investigated if the absence of activity-dependent Ube3a changes has any effect on learning and memory kinase activation. We utilized KCl and FC to determine synaptic activity-induced ERK 1/2 phosphorylation in acute hippocampal slices and in CA1 area of hippocampus of wild type (Ube3a m+/p+) and Ube3a deficient mice (Ube3a m-/p+). We demonstrated that Ube3a loss leads to impaired activity-dependent ERK 1/2 phosphorylation. It has been established that Ube3a m-/p+ mice have a profound deficit in LTP, implying the importance of this ligase in excitatory synaptic transmission. The abnormal LTP could be partially explained by an aberrant CaMKII function, decreased activity-dependent ERK 1/2 phosphorylation and reduced phosphatase activity. These proteins have also been implicated in another form of synaptic plasticity such as long-term depression (LTD). Chapter 3, we investigated the contribution of Ube3a to NMDAR - dependent and - independent LTD. Our data showed that Ube3a m-/p+ P21-30 animals exhibit the impairments in both forms of LTD. Next, we focused on elucidating molecular mechanism underlying the reduced mGluR1/5-LTD. We discovered that mGluR1/5 kinase activation such as ERK, mTOR and p38 is not affected by Ube3a loss. In concordance with previous work, we detected increased Arc expression together with abnormal AMPAR distribution in the Ube3a m-/p+ hippocampus. Surprisingly, the mGluR1/5 induced GluR2 trafficking was normal. Our findings infer that elevated Arc levels together with the increased internalization of AMPAR may result in compromised basal state of the synapses leading to a more depression-like state in Ube3a m-/p+ mice. Evidence points that loss of Ube3a produces alterations in a variety of activity-dependent signal transduction cascades that may ultimately result in impaired synaptic plasticity and cognition. Similar to AS, abnormal molecular and behavioral phenotypes have already been observed in other mouse models of human mental retardation such as Fragile X Mental Retardation Syndrome (FXS). Chapter 4 is set to explore if any correlation can be found in between these neurodevelopmental disorders. Analysis of crude synaptoneurosomes of adult Fmr1 KO mice revealed a significant reduction in Ube3a protein. Additionally, a blunted translation of Ube3a in response to mGluR1/5 stimulation was observed. However, we didn't find any evidence of direct interaction between Ube3a mRNA and Fragile X Mental Retardation Protein (FMRP). To examine if some of the pathology seen in Fmr1 KO mice is due to Ube3a downregulation, we performed a rescue experiment by increasing overall levels of Ube3a in hippocampus of FRMP deficient mice. An exhaustive battery of behavioral testing indicated that alterations of Ube3a expression impacted only associative fear conditioning. In summary, the present work has attempted to answer some of the fundamental questions about Ube3a and its role in synaptic plasticity. We have demonstrated that Ube3a expression is modulated by synaptic activation and its activity-dependent alterations are essential for normal brain functioning. Additionally, our data suggest that Ube3a is not only significant for the synaptic excitation but also crucial for the synaptic depression. Finally, our findings indicate that the alteration of Ube3a expression may contribute to the cognitive phenotypes in other neurodevelopmental disorders such as FXS suggesting an advantage of exploring Ube3a function outside the AS research.
10

PLASTICITY MECHANISMS IN VISUAL CORTEX: ANIMAL MODELS AND HUMAN CORTEX / MECHANISMS OF REINSTATED PLASTICITY

Beshara, Simon P January 2016 (has links)
A holy grail in neuroscience is being able to control plasticity to facilitate recovery from insult in the adult brain. Despite success in animal models, few therapies have translated from bench to bedside. This thesis is aimed at addressing 2 major stumbling blocks in translation. The first gap is in our understanding of the mechanisms of plasticity-enhancing therapies, and the second is in our understanding the relevance of those mechanisms for human development. In chapters 2 and 3, I address the first gap by asking whether fluoxetine, a selective serotonin reuptake inhibitor, which reinstates juvenile-like plasticity in adult animals, reinstates a juvenile-like synaptic environment. We found evidence to suggest that fluoxetine is neuroprotective, as it rescued all of the MD-driven changes, but surprisingly we found no evidence that fluoxetine recreated a juvenile-like synaptic environment, with the exception of Ube3A. Ube3A is necessary for critical period plasticity, indicating that Ube3A may play a crucial in enhancing plasticity in the adult cortex. In chapter 4, I address whether D-serine, an amino acid that has similar effects to fluoxetine in terms of both plasticity and anti-depression, shares a common neurobiological signature with fluoxetine. I found that D-serine’s effects were strikingly similar to fluoxetine, with respect to markers of the E/I balance, indicating that it may be an effective alternative to fluoxetine. In chapter 5, I address the second gap by studying the development of 5 glutamatergic proteins in human V1. Some changes occurred early, as would be predicted from animals studies, while other changes were protracted, lasting into the 4th decade. These results will help guide the use of treatments, like fluoxetine, which effect glutamatergic proteins. iv Together the findings in this thesis significantly advances our understanding of the mechanisms involved in restating plasticity in the adult cortex, and their relevance to humans. / Dissertation / Doctor of Philosophy (PhD) / Neurons change to rewire, adapt, and recover. This plasticity is greatest early in development, so much research has focused on bringing it back in adults. There has been amazing progress in animal models, but this has not translated to humans. Two reasons for this are that we do not fully understand the mechanisms of these treatments in animals or whether those mechanisms are relevant for humans. My thesis addresses this by studying how 2 treatments, fluoxetine and D-serine, affect proteins that are important for plasticity, and how those proteins develop in the humans. I found that these treatments are neuroprotective, but do not recreate a younger state. One interesting standout is an increase in Ube3A, which is essential for juvenile plasticity. I also found that much of human development is similar to animals, but the time course for some proteins is uniquely prolonged in humans. These findings have implications for the use of plasticity-enhancing treatments at different ages.

Page generated in 0.0171 seconds