• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 2
  • 1
  • Tagged with
  • 4
  • 4
  • 4
  • 3
  • 3
  • 3
  • 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

Application of Proximity Ligation Assay for Multidirectional Studies on Transforming Growth Factor-β Pathway

Zieba, Agata January 2012 (has links)
A comprehensive understanding of how the body and all its components function is essential when this knowledge is exploited for medical purposes. The achievements in biological and medical research during last decades has provided us with the complete human genome and identified signaling pathways that governs the cellular processes that facilitates the development and maintenance of higher order organisms. This has brought about the realization that diseases such as cancer is a consequence of genomic aberrations that effects these signaling pathways, endowing cancer cells with the capacity to circumvent homeostasis by acquiring features like self-sustained proliferation and insensitivity to apoptosis. The increased understanding of biology and medicine has been made possible by the development of advanced methods to carry out biological and clinical analyses. The demands of a method often differ regarding in what context it will be applied. It may be acceptable for method to be laborious and time consuming if it is used in basic research, but for medical purposes molecular methods need to be fast and straightforward to perform. Innovative technologies should preferentially address the demands of both researchers and clinicians and provide data not possible to obtain by other methods. An example of such a method is the in situ proximity ligation assay (in situ PLA). In this thesis I have used this method to determine the activity status, at the single-cell level, of the transforming growth factor-β (TGF-β) signaling pathway and activating protein-1 (AP-1) family of transcription factors.  Both of these pathways are frequently involved in cancer development and progression. In addition to this research I herein also present further modifications of in situ PLA, and analyses thereof, to increase the utility and resolution of this assay.
2

Regulation of cell polarity and invasion by TGF-β and BMP signaling

Shahidi Dadras, Mahsa January 2017 (has links)
Transforming growth factor β (TGF-β) and bone morphogenetic protein (BMP) signaling pathways are involved in many physiological processes during embryonic and adult life. TGF-β promotes epithelial to mesenchymal transition (EMT). We identified a gene target of TGF-β signaling, encoding the salt-inducible kinase 1 (SIK1). A potential substrate of this kinase, the polarity protein Par3, is an established regulator of tight junction assembly. SIK1 associates with Par3, can potentially phosphorylate Par3 and leads to its degradation, contributing to tight junction disassembly. Glioblastoma multiforme (GBM) is a common malignancy in the central nervous system, characterized by high heterogeneity, invasiveness, and resistance to therapy. One of the causes of heterogeneity and therapy-resistance is the existence of glioblastoma stem cells (GSCs). TGF-β signaling promotes self-renewal while BMP signaling induces differentiation of GSCs. Snail is a potent inducer of the EMT in carcinomas. However, in the context of GBM, Snail induces BMP signaling and represses TGF-β signaling through interaction with SMADs, the signaling mediators of TGF-β and BMP. In conclusion, Snail differentially regulates the activity of the opposing BMP and TGF-β pathways, thus promoting an astrocytic fate switch and repressing stemness in GSCs. Although profound changes in cell polarity is a hallmark of invasive malignancies, little is known about the role of the polarity machinery in tumor suppression. Patient transcriptomic data suggested low Par3 expression, correlating with poor survival of the GBM patients. Par3 silencing decreased the GSC self-renewal capacity and enhanced their invasiveness. Transcriptomic analysis indicates that loss of Par3 leads to downregulation of genes encoding mitochondrial enzymes that generate ATP. These results support a novel role of Par3 in GBM, beyond its contribution to junctional contacts between cells. Another regulator of TGF-β and BMP signaling is the liver kinase B1 (LKB1). According to GBM patient mRNA analysis, high levels of LKB1 correlate with poor prognosis. Silencing of LKB1 in GSCs impairs invasion and self-renewal capacity due to downregulation of genes involved in these processes. Moreover, loss of LKB1 induces mitochondrial dysfunction, leading to decreased ATP levels. Collectively, this thesis has delivered a group of novel regulatory pathways that control critical aspects of cancer cell polarity, invasion and stemness.
3

Étude des conséquences fonctionnelles de la mutation SGO1 K23E sur la voie de signalisation TGF-β

Gosset, Natacha 06 1900 (has links)
No description available.
4

Caractérisation moléculaire du syndrome CAID : mise en évidence des rôles non canoniques de SGO1 dans la régulation de la signalisation TGF-β et de l'épigénomique.

Piché, Jessica 07 1900 (has links)
Les contractions rythmiques résultent de l’activité stimulatrice du nœud sinusal dans le cœur et des cellules interstitielles de Cajal (CICs) dans les intestins. Nous avons découvert un nouveau syndrome résultant d’une combinaison de la maladie du nœud sinusal (MNS) et de la pseudo-obstruction intestinale chronique (POIC). Ce syndrome, que nous avons nommé Chronic Atrial and Intestinal Dysrhythmia (CAID), résulte d’une mutation récessive du gène SGO1 (K23E). Cependant, les rôles connus de SGO1 n'expliquent pas l'apparition postnatale du syndrome ni la pathologie spécifique, suggérant que des rôles non canoniques de SGO1 conduisent aux manifestations cliniques observées. Cette hypothèse est supportée par la comparaison de CAID avec les autres cohésinopathies qui présentent principalement des phénotypes développementaux sans ou avec des défauts légers du cycle cellulaire. Ce projet visait à une découverte non biaisée des mécanismes non canoniques expliquant le syndrome CAID en utilisant le dogme de la biologie moléculaire (ADN→ARNm→protéine) comme ligne directrice. Pour ce faire, nous avons effectué des criblages multi-omiques sur des fibroblastes de peau de patients CAID et de contrôles sains. Les résultats des criblages ont été validés par électrophysiologie, étude des voies de signalisation pertinentes, immunohistochimie, pyroséquençage des rétrotransposons LINE-1 et quantification des marques d’histones. Nos études multi-omiques ont confirmé des changements dans la régulation du cycle cellulaire, mais aussi dans la conduction cardiaque et la fonction des muscles lisses. Plus spécifiquement, plusieurs canaux potassiques étaient sous-régulés. L’électrophysiologie a confirmé une diminution du courant potassique rectifiant entrant (IK1). L'immunohistochimie des coupes intestinales de patients CAID a confirmé l’augmentation de l’expression de SGO1 et BUB1, un régulateur de la voie de signalisation TGF-β. De plus, la voie canonique de TGF-β est augmentée et est découplée de la voie non canonique. Au niveau épigénétique, une signature unique d’hyperméthylation et de fermeture de la chromatine a été observée. Ce qui est soutenu par l’augmentation de la méthylation de H3K9me3 et de H3K27me3. En conclusion, le syndrome CAID est associé à plusieurs changements ayant possiblement un effet cumulatif plutôt que d’une seule voie de signalisation dérégulée. Nos résultats désignent la perturbation du courant IK1, la dérégulation de la signalisation TGF-β, l’hyperméthylation de l’ADN et la compaction de la chromatine comme éléments conducteurs potentiels des manifestations cliniques observées. La voie TGF-β et les changements épigénétiques peuvent être ciblées par des médicaments existants, constituant ainsi des cibles thérapeutiques prometteuses pour le traitement du syndrome CAID. / Rhythmic contractions are driven by the pacemaker activity of the cardiac sinus node and the intestinal interstitial cells of Cajal (ICC). We have discovered a new syndrome resulting from a combination of sick sinus syndrome (SSS) and chronic intestinal pseudo-obstruction (CIPO). This syndrome, which we have named Chronic Atrial and Intestinal Dysrhythmia (CAID), results from a recessive mutation in the SGO1 gene (K23E). However, the known roles of SGO1 do not explain the postnatal onset of the syndrome nor the specific pathology, suggesting that non-canonical roles of SGO1 lead to the clinical manifestations observed. This hypothesis is supported by the comparison of CAID with other cohesinopathies which mainly exhibit developmental phenotypes without or with mild cell cycle defects. This project aimed towards an unbiased discovery of noncanonical mechanisms explaining CAID using the molecular biology dogma (DNA→mRNA→protein) as a guideline. We performed multi-omic screens on skin fibroblasts from CAID patients and healthy controls. Screening results were validated by electrophysiology, study of relevant signaling pathways, immunohistochemistry, LINE-1 retrotransposon pyrosequencing, and histone marks quantification. Our multiomics analyses confirmed changes in cell cycle regulation, but also in cardiac conduction and smooth muscle function. More specifically, several potassium channels were downregulated. Electrophysiology studies confirmed a decrease in the inward rectifier potassium current (IK1). Immunohistochemistry in CAID patient’s intestinal sections confirmed overexpression of SGO1 and BUB1, a regulator of TGF-β signaling pathway. Additionally, the canonical TGF-β signaling was increased and decoupled from noncanonical signaling. At the epigenetic level, CAID patient fibroblasts have a unique signature of hypermethylation and chromatin closure. This is supported by the increased methylation of H3K9me3 and H3K27me3. In conclusion, CAID syndrome is associated with several changes that, may have a cumulative effect rather than a single deregulated signaling pathway. Our results reveal the disturbance of the IK1 current, the deregulation of TGF-β signaling, DNA hypermethylation and chromatin accessibility changes as potential conductors of intestinal and cardiac manifestations of CAID syndrome. In particular, the TGF-β pathway and epigenetic changes, may be targeted by existing drugs, thus constituting promising therapeutic targets for the treatment of CAID syndrome.

Page generated in 0.0809 seconds