The search result changed since you submitted your search request. Documents might be displayed in a different sort order.
  • search hit 6 of 16
Back to Result List

The E3 ligase TRIM25 negatively interferes with the translation of caspase-2–therapeutic implications for sensitizing colon carcinoma cells to intrinsic apoptosis

  • An overexpression of the E3 ubiquitin ligase TRIM25 is implicated in several human cancers and frequently correlates with a poor prognosis and occurrence of therapy resistance in patients. Previous studies of our group have identified the mRNA encoding the pro-apoptotic caspase-2 as a direct target of the ubiquitous RNA binding protein human antigen R (HuR). The constitutive HuR binding observed in colon carcinoma cells negatively interferes with the translation of caspase-2 mainly through binding to the 5' untranslated region (UTR) of caspase-2 and thereby confers an increased survival of tumor cells. The main objective of this thesis was to unravel novel regulatory proteins critically involved in the control of caspase-2 translation and their impact on therapeutic drug resistance of human colon carcinoma cells. By employing RNA affinity chromatography in combination with mass-spectrometry, among several putative caspase-2 mRNA binding proteins, we have identified the tripartite motif-containing protein 25 (TRIM25) as novel caspase-2 translation regulatory protein in colon carcinoma cells. The constitutive TRIM25 binding to caspase-2 mRNA in two different human colorectal carcinoma cell lines was validated by ribonucleoprotein (RNP)-immunoprecipitation (RIP)-RT-PCR assay and by means of biotin-labeled RNA-pull-down assay. Since caspase-2 is a caspase which is particularly involved in the DNA-damage-induced apoptosis, I tested the functional relevance of negative caspase-2 regulation by TRIM25 for chemotherapeutic drug-induced cell death of different adenocarcinoma cells by RNA interference (RNAi)- mediated loss-of-function and gain-of-function approaches. In the first part of the thesis, I could demonstrate that transient silencing of TRIM25 caused a significant increase in caspase-2 protein levels without affecting the amount of corresponding mRNAs. Mechanistically, the TRIM25 silencing-triggered increase in caspase-2 was totally impaired by cycloheximide, indicating that the stimulatory effects on caspase-2 levels depend on protein synthesis. This finding was corroborated by RNP/polysomal fractionation, which revealed that the transient knockdown of TRIM25 caused a significant redistribution of caspase-2 transcripts from the fraction of RNP particles to that from translationally active polyribosomes. The second part of my thesis aimed at the elucidation of the functional consequences of the negative caspase-2 regulation by TRIM25 for enhanced tumor cell survival. Thereby, I found that the siRNA-mediated knockdown of TRIM25 caused a significant increase in the chemotherapeutic drug-induced cleavage of caspase-3 and to elevations in cytoplasmic cytochrome c levels implicating that TRIM25 depletion did mainly affect the intrinsic apoptotic pathway. Concordantly, the ectopic expression of TRIM25 caused a reduction in caspase-2 protein levels, concomitant with an attenuated sensitivity of tumor cells to doxorubicin. To test the functional impact of caspase-2 in the TRIM25 depletion-dependent sensitization to drug-induced apoptosis, I employed a siRNA-mediated knockdown of caspase-2. Interestingly, the strong induction of caspase-3 and -7 cleavage after doxorubicin treatment was fully impaired after the additional knockdown of caspase-2, indicating the sensitizing effects by TRIM25 knockdown depend on caspase-2. Data from this thesis identified the TRIM25 as a novel RNA-binding protein of caspase-2 mRNA, which negatively interferes with the translation of caspase-2 and which functionally contributes to chemotherapeutic drug resistance of colon carcinoma cells. Interfering with the negative TRIM25-caspase-2 axis may represent a promising therapeutic avenue for sensitizing colorectal cancers to conventional anti-tumor therapies.

Download full text files

Export metadata

Metadaten
Author:Usman NasrullahORCiDGND
URN:urn:nbn:de:hebis:30:3-571181
Place of publication:Frankfurt am Main
Referee:Achim SchmidtkoORCiDGND, Wolfgang EberhardtGND, Jochen KleinORCiDGND, Eugen ProschakORCiDGND
Document Type:Doctoral Thesis
Language:English
Date of Publication (online):2020/11/26
Year of first Publication:2020
Publishing Institution:Universitätsbibliothek Johann Christian Senckenberg
Granting Institution:Johann Wolfgang Goethe-Universität
Date of final exam:2020/10/29
Release Date:2020/12/16
Page Number:139
HeBIS-PPN:473924358
Institutes:Biochemie, Chemie und Pharmazie
Dewey Decimal Classification:5 Naturwissenschaften und Mathematik / 57 Biowissenschaften; Biologie / 570 Biowissenschaften; Biologie
6 Technik, Medizin, angewandte Wissenschaften / 61 Medizin und Gesundheit / 610 Medizin und Gesundheit
Sammlungen:Universitätspublikationen
Licence (German):License LogoDeutsches Urheberrecht