Show simple item record

dc.contributor.advisorPeter C. Dedon.en_US
dc.contributor.authorGu, Chen, Ph. D. Massachusetts Institute of Technologyen_US
dc.contributor.otherMassachusetts Institute of Technology. Department of Biological Engineering.en_US
dc.date.accessioned2017-12-05T19:16:10Z
dc.date.available2017-12-05T19:16:10Z
dc.date.copyright2017en_US
dc.date.issued2017en_US
dc.identifier.urihttp://hdl.handle.net/1721.1/112512
dc.descriptionThesis: Ph. D., Massachusetts Institute of Technology, Department of Biological Engineering, 2017.en_US
dc.descriptionCataloged from PDF version of thesis.en_US
dc.descriptionIncludes bibliographical references.en_US
dc.description.abstractThe human tRNA methyltransferase 9-like (TRM9L) gene is a homolog of yeast Trm9 and human ALKBH8 and it has an important function in suppressing tumor growth in colorectal cancer. Loss of heterozygosity events on the Chromosome 8p22 loci, where TRM9L is located, are overrepresented in a wide variety of cancers, including prostate cancer, breast carcinoma, and hepatocellular carcinoma; downregulation of TRM9L expression is also observed in many different types of cancer. These findings implicate a general role potentially played by TRM9L in tumor suppression. A mechanistic understanding of TRM9L would have a broad impact in oncology. The broad objective of my thesis research is to connect mechanistically the biochemical function of TRM9L to its tumor suppressing activity. Of my special interest is TRM9L is regulated at the protein level. In this thesis, I demonstrated that TRM9L is a phosphoprotein, with phosphorylation dynamics at serines S214, S255, S279, S291, S306, and S380 correlated with protein-protein interactions and tumorigenicity. Phosphorylation levels were found to be modulated by stressors to which cells become resistant when TRM9L is silenced. For example, oxidative stress caused by hydrogen peroxide (H202) exposure increased phosphorylation on S255, S291, and S380, but phosphorylation was unchanged in response to ionizing radiation. Using chemical genetics approaches, I showed that phosphorylation of S380 is downstream of ribosomal protein S6 kinase (RSK), which is downstream of H202-activated extracellular signalregulated kinase (ERK). TRM9L mutations S214A, S255A and S380A significantly enhanced tumor growth, while S214A and S255A mutations also abolished a direct interaction between TRM9L and certain 14-3-3 isoforms. The results revealed a novel oxidative stress phosphosignaling regulatory mechanism underlying TRM9L's tumor suppressor behavior. I also demonstrated that TRM9L altered the ability of colorectal cancer cells to respond to stresses caused by reactive oxygen and nitrogen species. Results supported the idea that TRM9L reduces the cell's capacity to detoxify harmful reactive oxygen and nitrogen species and effectively makes them more toxic. Finally, my finding supported the notion that TRM9L expression downregulates the hypoxia-induced cell migration, presumably by controlling an aspect of epithelialmesenchymal transition (EMT).en_US
dc.description.statementofresponsibilityby Chen Gu.en_US
dc.format.extent129 pagesen_US
dc.language.isoengen_US
dc.publisherMassachusetts Institute of Technologyen_US
dc.rightsMIT theses are protected by copyright. They may be viewed, downloaded, or printed from this source but further reproduction or distribution in any format is prohibited without written permission.en_US
dc.rights.urihttp://dspace.mit.edu/handle/1721.1/7582en_US
dc.subjectBiological Engineering.en_US
dc.titleThe molecular basis for tumor growth suppression by tRNA methyltransferase 9-like (TRM9L)en_US
dc.typeThesisen_US
dc.description.degreePh. D.en_US
dc.contributor.departmentMassachusetts Institute of Technology. Department of Biological Engineering
dc.identifier.oclc1011594971en_US


Files in this item

Thumbnail

This item appears in the following Collection(s)

Show simple item record