• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 43
  • 6
  • 6
  • 5
  • 1
  • Tagged with
  • 71
  • 71
  • 71
  • 21
  • 15
  • 12
  • 11
  • 11
  • 9
  • 9
  • 8
  • 7
  • 7
  • 6
  • 6
  • 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.
31

The Ron Receptor Tyrosine Kinase in Tissue Morphogenesis

Meyer, Sara January 2009 (has links)
No description available.
32

The Ron Receptor Tyrosine Kinase as a Mediator of Inflammation and Tumorigenesis

Paluch, Andrew M. 28 June 2016 (has links)
No description available.
33

Role of the SCF/KIT signalling pathway in embryonic stem cells

Fraser, Lindsay January 2011 (has links)
Murine embryonic stem (ES) cells are derived from the inner cell mass of the developing embryonic blastocyst. These cells can self renew which allows them to be propagated indefinitely in the laboratory and they can differentiate into cell types derived from all three germ layers. Manipulation of the mouse genome using gene targeting techniques in conjunction with ES cell technology has provided valuable insights into embryonic development and cell lineage specification. KIT is a trans-membrane receptor tyrosine kinase (RTK) that dimerises upon binding to its ligand, stem cell factor (SCF) resulting in the auto-phosphorylation of intracellular kinase domains. This activity is crucial for the transmission of signals from the cell surface to the nucleus. KIT is expressed on stem and progenitor cells of many lineages and defects in the SCF/KIT signaling pathway causes detrimental effects at both the cellular and physiological level. This project aimed to investigate the role of the SCF/KIT signalling pathway during murine ES cell differentiation and survival. To assess the role of SCF/KIT signalling in ES cell proliferation and survival, we knocked out the c-kit gene in mouse ES cells to produce heterozygous (KitW-lacZ/+) and KIT Null (KitW-lacZ/W-lacZ) cell lines. The self renewal and differentiation profile of these cell lines revealed an auxiliary role for SCF/KIT during ES cell self renewal and an absolute role in survival upon in vitro differentiation. This phenotype of apoptosis upon differentiation was recapitulated in wild type E14 ES cells treated with a KIT neutralising antibody (ACK2). Wild type cells that were treated with the JNK inhibitor, SP600125 had a comparable phenotype to KIT null cells indicating that this could be one of the mediators of KIT signalling that has a protective role in the survival of differentiating ES cells. We hypothesised that blocking classical apoptotic pathways might prevent the death on differentiation observed in KIT null cells. However, neither blocking the pro-apoptotic P38 pathway with the chemical inhibitor PD169316 nor over-expressing the pro-survival protein BCL2 in KIT Null cells could prevent their apoptosis upon differentiation phenotype. This strongly suggests that these pathways are not involved in KIT mediated survival of differentiating ES cells. Although compensatory mechanisms are thought to exist for defective KIT signaling in vivo, an absolute role is assigned to KIT during ES cell differentiation. Further analysis of micro array data comparing gene expression from wild type E14 and KIT Null cell lines may reveal the specific mechanisms of KIT mediated survival during differentiation onset.
34

Régulation de la signalisation du récepteur MET par la protéine SOCS1 dans le carcinome hépatocellulaire / Regulation of MET signaling by SOCS1 in hepatocellular carcinoma

Gui, Yirui January 2014 (has links)
Résumé : La répression fréquente du gène encodant pour le « suppressor of cytokine signaling 1 » (SOCS1) dans le carcinome hépatocellulaire (CHC) et la forte susceptibilité des souris déficientes pour SOCS1 à développer des tumeurs hépatiques expérimentales suggèrent que SOCS joue un rôle de suppresseur de tumeur. Cette notion est supportée par les études impliquant la répression de l’expression de SOCS1 via des évènements épigénétiques ou par les microARN dans plusieurs autres types de cancers. Les mécanismes moléculaires sous-jacents au rôle potentiel de suppresseur de tumeur de SOCS1 dans le foie demeurent à ce jour inconnus. Bien que les récepteurs à activité tyrosine kinase (RTK) sont reconnus pour induire l’expression de l’ARNm de SOCS1, le rôle et les mécanismes par lesquels SOCS1 peut réguler la signalisation des RTK sont incertains. Le RTK MET, qui a pour ligand le facteur de croissance des hépatocytes (HGF), régule plusieurs fonctions cellulaires normales. La dérégulation de la signalisation du récepteur MET joue des rôles importants dans la pathogenèse du CHC. Des études ont démontré que l’activation de MET promeut la prolifération, l’invasion et la migration des cellules cancéreuses du foie ainsi que leur dissémination métastatique. La signalisation aberrante de MET est un trait commun de plusieurs autres cancers et serait à l’origine de l’émergence de la résistance à la chimiothérapie. Dans ce projet, j’ai investigué les mécanismes moléculaires par lesquels SOCS1 régule l’activité du récepteur MET. Mes résultats indiquent que le foie des souris Socs1[indice supérieur -/-]Ifng[indice supérieur -/-] se régénère plus rapidement que celui des souris contrôles. Suivant une stimulation au HGF, les hépatocytes issus des souris Socs1[indice supérieur -/-] Ifng[indice supérieur -/-] présentent une augmentation de la signalisation de MET, de la migration et de la prolifération cellulaires. L’expression exogène de SOCS1 dans différentes lignées cellulaires d’hépatocarcinomes humains et murins inhibe la signalisation induite par HGF. De plus, SOCS1 diminue la prolifération, la croissance indépendante de l’anchrage et la migration dans ces lignées de CHC in cellulo et réduit de façon significative leur croissance dans les essais de xénogreffes chez les souris immunodéficientes. Mes résultats suggèrent que l’activation de la signalisation HGF-MET induit la transcription du gène SOCS1, suivi par une interaction physique entre SOCS1 et MET. L’analyse de divers mutants de SOCS1 révèle que cette interaction implique principalement les domaines SH2 et « kinase inhibitory region » (KIR) de SOCS1. L’activité kinasique de MET est requise pour cette interaction puisque l’interaction entre SOCS1 et un mutant kinase-inactif de MET est fortement réduite. SOCS1 est aussi phosphorylé en aval de MET sur quatre résidus tyrosine (Tyr). Quoique ces résidus Tyr représentent théoriquement des sites d’interaction pour des protéines adaptatrices possédant des domaines de liaison aux phospho-Tyr, elles ne semblent pas impliquées dans l’interaction de SOCS1 avec MET. Je démontre également que SOCS1 induit l’ubiquitination de MET via l’élongation de chaînes de polyubiquitine de type K48, conduisant à sa dégradation par le protéasome. Cette modulation négative de MET par SOCS1 dans les cellules CHC survient indépendamment de la voie de dégradation lysosomale de Cbl qui est partagée par plusieurs autres RTK. // Abstract : Frequent repression of the gene coding for the suppressor of cytokine signaling 1 (SOCS1) in hepatocellular carcinoma (HCC) and increased susceptibility of SOCS1 deficient mice to experimental hepatocarcinogenesis suggest a tumor suppressor role for SOCS1. This notion is supported by epigenetic and micro-RNA-mediated blockade of SOCS1 expression in several other cancers. Molecular mechanisms underlying the putative tumor suppressor function of SOCS1 in the liver have not been elucidated yet. Although receptor tyrosine kinases (RTK) can induce SOCS1 mRNA expression, the role and mechanisms of SOCS1 in regulating RTK signaling are not yet clear. c-Met is the RTK for hepatocyte growth factor (HGF) and mediates several normal cellular functions. HGF signaling and MET activation also play important roles in the pathogenesis of HCC. Experimental studies have shown that the activated MET promotes proliferation, invasion and migration of liver cancer cells and enhances metastasis. Aberrant MET signaling is a hallmark of many other cancers and underlies the emergence of chemoresistant clones. In this project, I investigated the molecular mechanisms by which SOCS1 regulates MET RTK activity. My results illustrate that the Socs1[superscript -/-]Ifng[superscript -/-] liver regenerates at a faster rate than the control one. Following HGF stimulation, hepatocytes from Socs1[superscrip -/-]Ifng[superscript -/-] mice display increased MET signaling, cell migration and proliferation. Forced expression of SOCS1 inhibits HGF-induced signaling pathways in different human or murine hepatoma cell lines. Furthermore, SOCS1 also decreases cell proliferation, anchorage-independent growth, and migration of HCC cell lines in cellulo, and results in significant inhibition of their growth as xenografts in immunodeficient mice. My findings show that activation of HGF-MET signaling results in transcriptional activation of SOCS1 gene, followed a physical interaction between SOCS1 and MET. Analysis of various SOCS1 mutants reveals that this interaction is mediated primarily via the SH2 and the kinase inhibitory region (KIR) domain of SOCS1. MET kinase activity is required for this interaction since SOCS1 binding to a kinase-dead MET mutant is dramatically reduced. MET promotes phosphorylation of SOCS1 on four tyrosine (Tyr) residues. Although these Tyr might represent potential binding sites for adaptors containing phospho-Tyr-binding domains, they do not appear to be involved in the interaction of SOCS1 with MET. I also show that SOCS1 induces polyubiquitination of MET via K48-ubiquitin chain elongation leading to its degradation by proteasomes. The SOCS1-mediated downmodulation of MET expression in HCC cells occurs independently of the Cbl-mediated lysosomal degradation pathway shared by many other RTKs. Taken together, my findings show that SOCS1 attenuates HGF-induced cellular functions by targeting the activated MET receptor for proteasomal degradation.
35

Pim1 kinase regulates c-Kit gene translation

An, Ningfei, Cen, Bo, Cai, Houjian, Song, Jin H., Kraft, Andrew, Kang, Yubin 30 December 2016 (has links)
Background: Receptor tyrosine kinase, c-Kit (CD117) plays a pivotal role in the maintenance and expansion of hematopoietic stem/progenitor cells (HSPCs). Additionally, over-expression and/or mutational activation of c-Kit have been implicated in numerous malignant diseases including acute myeloid leukemia. However, the translational regulation of c-Kit expression remains largely unknown. Methods and results: We demonstrated that loss of Pim1 led to specific down-regulation of c-Kit expression in HSPCs of Pim1(-/-)mice and Pim1(-/-)2(-/-)3(-/-) triple knockout (TKO) mice, and resulted in attenuated ERK and STAT3 signaling in response to stimulation with stem cell factor. Transduction of c-Kit restored the defects in colony forming capacity seen in HSPCs from Pim1 (-/-) and TKO mice. Pharmacologic inhibition and genetic modification studies using human megakaryoblastic leukemia cells confirmed the regulation of c-Kit expression by Pim1 kinase: i.e., Pim1-specific shRNA knockdown down-regulated the expression of c-Kit whereas overexpression of Pim1 up-regulated the expression of c-Kit. Mechanistically, inhibition or knockout of Pim1 kinase did not affect the transcription of c-Kit gene. Pim1 kinase enhanced c-Kit S-35 methionine labeling and increased the incorporation of c-Kit mRNAs into the polysomes and monosomes, demonstrating that Pim1 kinase regulates c-Kit expression at the translational level. Conclusions: Our study provides the first evidence that Pim1 regulates c-Kit gene translation and has important implications in hematopoietic stem cell transplantation and cancer treatment.
36

The Role of KRAS in Mechanosensing in Non-Small Cell Lung Cancer

Powell, Krista M 01 January 2019 (has links)
Lung cancer is the number one cause of cancer related death worldwide, with more than 1.6 million fatalities each year. Non-small cell lung cancer (NSCLC) accounts for 80-85% of all lung cancers, with KRAS being one of the most prevalent oncogenic driver mutations. Therapeutic approaches for KRAS-mutated NSCLC have been extensively explored due to the US National Cancer Institute RAS Initiative, but methods of directly targeting KRAS or downstream effectors, such as MEK, still have poor results. Previous reports have shown that KRAS-mutated NSCLC activate distinct receptor tyrosine kinases (RTKs) depending on the epithelial or mesenchymal state. Epithelial-to-mesenchymal transition (EMT) is known to play a role in the metastasis and poor prognosis of cancer, and is induced by extracellular matrix (ECM) stiffness. Hallmarks of EMT include loss of E-Cadherin and increase in Vimentin. This research investigates the role of KRAS in EMT transition due to increased ECM stiffness in KRAS mutant NSCLC, and how this affects the efficacy of KRAS and MEK inhibition. To understand how KRAS mutations in NSCLC play a role in this stiffness induced EMT, experiments were performed to detect the gene and protein expression of EMT markers, as well as possible sources of mechanosensing, including primary cilia and receptor tyrosine kinases. We hypothesized that KRAS plays a role in activation of mechanosensors and directly correlates to EMT induced by increased mechanical forces. Results show when KRAS was inhibited and there was increased mechanical forces, either from stretch or substrate stiffness, there was a decreased activation of mechanosensors. KRAS inhibition also prevented the cells from undergoing stiffness-induced EMT. This supports our hypothesis that KRAS plays a key role in ECM stiffness induced EMT. Future studies include examining the mechanism behind this phenomenon and in vivo studies.
37

Ubiquitination and Receptor Endocytosis

Haglund, Kaisa January 2004 (has links)
<p>Protein ubiquitination is an evolutionary conserved mechanism that controls a wide variety of cellular functions. Polyubiquitinated proteins are generally degraded in the proteasome, whereas monoubiquitination controls various other cellular processes, including endocytosis and endosomal sorting.</p><p>Termination of signaling by activated receptor tyrosine kinases (RTKs) largely occurs via their endocytosis and subsequent lysosomal degradation, processes accompanied by receptor ubiquitination. Cbl family proteins are major ubiquitin ligases that promote RTK ubiquitination and downregulation. We showed that epidermal growth factor (EGF) and platelet derived growth factor (PDGF) receptors are monoubiquitinated at multiple sites following their ligand-induced activation and that a single ubiquitin is sufficient for both receptor internalization and degradation. Cbl also controls EGF receptor (EGFR) downregulation by binding to CIN85, which recruits endophilins to EGFR/Cbl complexes. In the complex with activated EGFRs, Cbl directs monoubiquitination of CIN85, and the entire complex is targeted for degradation in the lysosome. We propose that multiple monoubiquitination of activated receptors and associated protein complexes ensures proper receptor sorting towards the lysosome. Importantly, the functions of Cbl are also negatively controlled in order to maintain cellular homestasis. Sprouty2 blocks EGFR downregulation by sequestering Cbl from activated EGFRs. We showed that Sprouty2 also associates with CIN85 and that this binding is required for efficient inhibition of EGFR ubiquitination and endocytosis. </p><p>Cbl is also implicated in other aspects of RTK signaling, including organization of the actin cytoskeleton. We found that growth factor receptor signals promote lamellipodia formation in neuronal cells via a complex containing Cbl, the adaptor protein ArgBP2 and Pyk2. The lamellipodia formation required intact lipid rafts and the recruitment of Crk and PI(3)K to tyrosine phosphorylated Cbl.</p><p>In conclusion, our findings contribute to a better understanding of monoubiquitin signals in downregulation of RTKs and point at a role of Cbl in the regulation of cytoskeleton dynamics.</p>
38

Ubiquitination and Receptor Endocytosis

Haglund, Kaisa January 2004 (has links)
Protein ubiquitination is an evolutionary conserved mechanism that controls a wide variety of cellular functions. Polyubiquitinated proteins are generally degraded in the proteasome, whereas monoubiquitination controls various other cellular processes, including endocytosis and endosomal sorting. Termination of signaling by activated receptor tyrosine kinases (RTKs) largely occurs via their endocytosis and subsequent lysosomal degradation, processes accompanied by receptor ubiquitination. Cbl family proteins are major ubiquitin ligases that promote RTK ubiquitination and downregulation. We showed that epidermal growth factor (EGF) and platelet derived growth factor (PDGF) receptors are monoubiquitinated at multiple sites following their ligand-induced activation and that a single ubiquitin is sufficient for both receptor internalization and degradation. Cbl also controls EGF receptor (EGFR) downregulation by binding to CIN85, which recruits endophilins to EGFR/Cbl complexes. In the complex with activated EGFRs, Cbl directs monoubiquitination of CIN85, and the entire complex is targeted for degradation in the lysosome. We propose that multiple monoubiquitination of activated receptors and associated protein complexes ensures proper receptor sorting towards the lysosome. Importantly, the functions of Cbl are also negatively controlled in order to maintain cellular homestasis. Sprouty2 blocks EGFR downregulation by sequestering Cbl from activated EGFRs. We showed that Sprouty2 also associates with CIN85 and that this binding is required for efficient inhibition of EGFR ubiquitination and endocytosis. Cbl is also implicated in other aspects of RTK signaling, including organization of the actin cytoskeleton. We found that growth factor receptor signals promote lamellipodia formation in neuronal cells via a complex containing Cbl, the adaptor protein ArgBP2 and Pyk2. The lamellipodia formation required intact lipid rafts and the recruitment of Crk and PI(3)K to tyrosine phosphorylated Cbl. In conclusion, our findings contribute to a better understanding of monoubiquitin signals in downregulation of RTKs and point at a role of Cbl in the regulation of cytoskeleton dynamics.
39

Exploiting Drosophila as a model system for studying anaplastic lymphoma kinase in vivo

Eriksson, Therese January 2010 (has links)
Anaplastic Lymphoma Kinase (ALK) is a Receptor Tyrosine Kinase (RTK) and an oncogene associated with several human diseases, but its normal function in humans and other vertebrates is unclear. Drosophila melanogaster has an ALK homolog, demonstrating that the RTK has been conserved throughout evolution. This makes Drosophila a suitable model organism for studying not only Drosophila ALK function, but also to study mammalian forms of ALK. In Drosophila the ligand Jeb activates ALK, initiating signaling crucial for visceral mesoderm development. The activating ligand for mammalian ALK is unclear, and for this reason Drosophila was employed in a cross-species approach to investigate whether Drosophila Jeb can activate mouse ALK. Jeb is unable to activate mouse ALK, and therefore mouse ALK is unable to substitute for and rescue the Drosophila ALK mutant phenotype. This suggests that there has been significant evolution in the ALK-ligand relationship between the mouse and Drosophila. In humans ALK has recently been shown to be involved in the development of neuroblastoma, a cancer tumor in children. I have developed a Drosophila model for examining human gain of function ALK mutants found in neuroblastoma patients. The various ALK variants have acquired point mutations in the kinase domain that have been predicted to activate the RTK in a constitutive and ligand independent manner. When expressed in the fly eye, active human ALK mutants result in a rough eye phenotype, while inactive wild type ALK does not, due to the lack of an activating ligand in the fly. In this way  several of the ALK mutations identified in neuroblastoma patients could be confirmed to be activated in a ligand independent manner. Moreover, a novel ALK mutant; ALKF1174S, was discovered in a neuroblastoma patient and was in the Drosophila model shown to be a gain of function mutation, and a previously predicted gain of function mutation; ALKI1250T, was shown to be a kinase dead mutation. This fly model can also be used for testing ALK selective inhibitors, for identifying activating ligands for human ALK and for identifying conserved components of the ALK signaling pathway. Gut musculature development in Drosophila is dependent on ALK signaling, while somatic muscle development is not. Proteins of the Wasp-Scar signaling network regulate Arp2/3-complex mediated actin polymerization, and I have investigated their function in visceral and somatic muscle fusion. I found that Verprolin and other members of this protein family are essential for somatic but not visceral muscle development. Despite fusion defects in both tissues in Verprolin and other examined mutants, gut development proceeds, suggesting that fusion is not crucial for visceral mesoderm development. Hence the actin polymerization machinery functions in both somatic and visceral muscle fusion, but this process only appears to be essential in somatic muscle development. / Exploiting Drosophila as a model system for studying Anaplastic Lymphoma Kinase in vivo
40

Mechanism and Therapeutic Potential of Statin-Mediated Inhibition of Tyrosine Kinase Receptors

Zhao, Tong Tong 27 October 2011 (has links)
Receptor tyrosine kinases (RTK) are key regulators of growth, differentiation and survival of epithelial cells and play a significant role in the development and progression of cancers derived from these tissues. In malignant cells, these receptors and their downstream signalling pathways are often deregulated, leading to cell hyper-proliferation, enhanced cell survival and increased metastatic potential. Furthermore, endothelial expressed RTKs regulate tumor angiogenesis allowing for tumor growth and maintenance by promoting their vascularization. Epithelial malignancies such as squamous cell carcinomas (SCC), non-small cell lung (NSCLC) and malignant mesotheliomas have very limited treatment options when presenting as metastatic disease. RTKs, particularly the epidermal growth factor (EGFR) and the vascular endothelial growth factor (VEGFR) receptors, have been shown to play significant roles in the pathogenesis of these tumor types. Statins are potent inhibitors of HMG-CoA reductase, the rate limiting enzyme of the mevalonate pathway, that are widely used as hypercholesterolemia treatments. The mevalonate pathway produces a variety of end products that are critical for many different cellular pathways, thus, targeting this pathway can affect multiple signalling pathways. Our laboratory has previously shown that lovastatin can induce tumor specific apoptosis especially in SCC and that 23% of recurrent SCC patients treated with lovastatin as a single agent showed disease stabilization in our Phase I clinical trial. Subsequently, our lab was able to demonstrate that lovastatin in combination with gefitinib, a potent inhibitor of the EGFR showed co-operative cytotoxicity when combined (Chapter 2). Furthermore, the pro-apoptotic and cytotoxic effects of these agents were found to be synergistic and to be manifested in several types of tumor cell lines including SCC, NSCLC and glioblastoma. I was able to expand upon these important findings and demonstrated that lovastatin, through its ability to disrupt the actin cytoskeleton, inhibited EGFR dimerization and activation (Chapter 3). This novel mechanism targeting this receptor has clinical implications as lovastatin treatment combined with gefitinib showed co-operative inhibitory effects on EGFR activation and downstream signalling. The RTK family of proteins share similar features with respect to activation, internalization and downstream signalling effectors. I further demonstrated that lovastatin can inhibit the VEGFR-2 in endothelial cells and mesotheliomas, where VEGF and its receptor are co-expressed driving their proliferation, and induces synergistic cytotoxicity in mesothelioma cells in combination with VEGFR-2 tyrosine kinase inhibitors (Chapter 4). These findings suggest that statins may augment the effects of a variety of RTK inhibitors in a similar fashion representing a novel combinational therapeutic approach in a wide repertoire of human cancers. More importantly, based on this work, we initiated a Phase I/II study evaluating high dose rosuvastatin and the EGFR inhibitor tarceva in SCC and NSCLC patients at our institute. This clinical evaluation will provide invaluable data that will play a role in developing this novel therapeutic strategy. Together, the work embodied in this thesis provides a model for the regulation of EGFR/VEGFR-2 activation and signalling by targeting the rho family of proteins that demonstrates a novel mechanism that can be exploited to refine current therapeutic paradigms.

Page generated in 0.0783 seconds