• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • No language data
  • Tagged with
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 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

Discovery of a Novel Regulatory Mechanism of TNK1 by 14-3-3 and Its Ubiquitin Association Domain Provides a Potential Therapeutic Targeting Opportunity in Cancer

Chan, Tsz Yin 03 August 2020 (has links)
While a relatively limited number of known oncogenes underlie a large percentage of cancers, a variety of new genes have emerged as low-frequency cancer drivers. Each of these new oncogenes represents a frontier for targeted therapy. However, the discovery of low-frequency targetable oncogenic drivers is challenging. This study focuses on the poorly understood Tyrosine kinase non-receptor-1 (TNK1), which has been reported to have both oncogenic and tumor suppressive functions. TNK1 has been identified to promote cancer cells survival and promote chemoresistance in multiple independent studies. On the other hand, whole-body constitutive deletion of TNK1 in mice caused an increase in spontaneous carcinomas and lymphomas. All in all, with no known regulatory mechanism and substrates of TNK1, the precise biological role of TNK1 is still unclear. To understand how TNK1 is regulated, we employed a proteomic approach to identify TNK1 interactors. We found out that TNK1 interacts with the phospho-binding protein 14-3-3 and this interaction is mediated by a cluster of MARK-mediated phosphorylations within the proline-rich domain. 14-3-3 binding retains TNK1 in the cytosol and maintains TNK1 in an inactive state. Release of TNK1 from 14-3-3 binding drives TNK1 to a heavy membrane fraction, where it becomes highly active. One unique feature of TNK1 is an ubiquitin association domain (UBA) on its C-terminus. Our data suggest that the UBA domain of TNK1 binds to poly-ubiquitin chains in nondiscriminatory manner. Remarkably, point mutations within the UBA that disrupt ubiquitin binding abolish TNK1 activation and oncogenic signaling, suggesting, to our knowledge, a unique UBA-centric mechanism of tyrosine kinase regulation. Finally, we used a structure-guided approach to identify a small molecule inhibiting TNK1 with high potency and selectivity. Such compound, TP-5801, inhibits TNK1 dependent STAT3 phosphorylation. TP-5801 also prolongs the survival of mice injected via tail vein with TNK1-driven Ba/F3 cells and reduces tumor burden in a subcutaneous xenograft model. In conclusion, our data reveal a mechanism of TNK1 regulation that controls its oncogenic tyrosine kinase activity and a potential strategy for TNK1 inhibition.
2

Examination of 14-3-3 Interactors Identifies a Novel Mechanism of Regulation for the Ubiquitin Binding Kinase TNK1 That Can Be Targeted to Block Tumor Growth

Egbert, Christina Marie 09 August 2022 (has links) (PDF)
Decades of research have begun to identify oncogenic mut-drivers that are responsible for driving a large percentage of cancers. These high frequency mut-drivers have therapeutics in the clinic for patient treatment. However, there is another group of low frequency mut-drivers that fail to rise above the noise of the high frequently drivers. These low frequency drivers represent a group of genes with untapped potential for new targeted therapies. However, identifying these drivers can be difficult. This study focuses on identifying new functional phosphorylations using the phospho-docking protein 14-3-3. The family of 14-3-3 proteins have been linked to many oncogenic pathways due to the diversity in their client protein interactions. One critical problem in studying 14-3-3 interactors is uncovering the docking site on the phospho-binding partner. Our work indicates that intrinsic disorder and unbiased mass spectrometry identification rate of a given phosphorylation are important for improving the selection of a 14-3-3 docking site. Using a machine learning model, we developed a tool that combines current available 14-3-3 prediction data and our observations about disorder and phosphorylation observation to predict 14-3-3 binding sites. Our publicly available tool "14-3-3-site-finder" produces a rank order list of potential 14-3-3 docking sites that could help overcome the time-consuming process of identifying the correct site. In our efforts of identifying functional phosphorylations with 14-3-3, we have observed that 14-3-3 interacts with a non-receptor tyrosine kinase, TNK1. TNK1 is a poorly characterized kinase that has essentially nothing known about its substrates, function or regulation. TNK1 has been implicated in both tumor suppressor and oncogenic roles. Particularly, a Hodgkin Lymphoma cell line is dependent on a truncated form of TNK1 for growth. Our work uncovers the first mechanism of regulation for this kinase. We found that MARK kinase phosphorylates TNK1 within the proline rich domain allowing 14-3-3 to dock on this phosphorylation. 14-3-3 binding restrains TNK1 in the cytosol and holds TNK1 in an inactive state. Upon the release of 14-3-3, TNK1 moves to a membrane fraction where it is active. One unique feature of TNK1 is that it has a c-terminal ubiquitin association domain (UBA). In vitro ubiquitin pulldowns indicate that the TNK1 UBA has no preference for linkage type or length of ubiquitin. Further, biolayer interferometry indicates that the UBA binds ubiquitin tightly. Mutation of residues within the ubiquitin:TNK1 interface prevent ubiquitin binding and decrease TNK1 activity, preventing downstream oncogenic signaling, suggesting a UBA-centric mechanism of regulation for TNK1. Finally, we developed a small molecule inhibitor, TP-5801, that selectively targets TNK1. TP-5801 prevents downstream TNK1 phosphorylation of STAT3. Further, TP-5801 prolonged the life of mice injected with TNK1 driven Ba/F3 cells. Taken together, our data reveal the first mechanism of kinase regulation for TNK1 involving 14-3-3 binding and ubiquitin association as well as the development of a TNK1 specific therapeutic
3

A Machine Learning Approach that Integrates Clinical Data and PTM Proteomics Identifies a Mechanism of ACK1 Activation and Stabilization in Cancer

Loku Balasooriyage, Eranga Roshan Balasooriya 08 August 2022 (has links)
Identification of novel cancer driver mutations is crucial for targeted cancer therapy, yet a difficult task especially for low frequency drivers. To identify cancer driver mutations, we developed a machine learning (ML) model to predict cancer hotspots. Here, we applied the ML program to 32 non-receptor tyrosine kinases (NRTKs) and identified 36 potential cancer driver mutations, with high probability mutations in 10 genes, including ABL1, ABL2, JAK1, JAK3, and ACK1. ACK1 is a member of the poorly understood ACK family of NRTKs that also includes TNK1. Although ACK1 is an established oncogene and high-interest therapeutic target, the exact mechanism of ACK1 regulation is largely unknown and there is still no ACK1 inhibitor in clinical use. The ACK kinase family has a unique domain arrangement with most notably, a predicted ubiquitin association (UBA) domain at its C-terminus. While the presence of a functional UBA domain on a kinase is unique to the ACK family, but the role of the UBA domain on ACK1 is unknown. Interestingly, the ML program identified the ACK1 Mig6 homology region (MHR) and UBA domains truncating mutation p633fs* as a cancer driver mutation. Our data suggest that the ACK1 UBA domain helps activate full-length ACK1 through induced proximity. It also acts as a mechanism of negative feedback by tethering ACK1 to ubiquitinated cargo that is ultimately degraded. Indeed, our preliminary data suggest that truncation of the ACK1 UBA stabilizes ACK1 protein levels, which results in spontaneous ACK1 oligomerization and activation. Furthermore, our data suggests removal of the MHR domain hyper activates ACK1. Thus, our data provide a model to explain how human mutations in ACK1 convert the kinase into an oncogenic driver. In conclusion, our data reveal a mechanism of ACK1 activation and potential strategies to target the kinase in cancer.

Page generated in 0.2726 seconds