Return to search

A Characterization of MK-STYX, A Catalytically Inactive Phosphatase Regulating Mitochondrial Apoptosis

<p> Chemoresistance is a highly significant problem affecting a diverse array of cancers at all clinical stages. In an attempt to identify molecular mechanisms leading to chemoresistance, we performed a RNAi screen against all known and putative kinases and phosphatases in the human genome. The knockdown of one of these genes, MK-STYX, resulted in potent chemoresistance in response to a diverse array of chemotherapeutic agents. As many of these drugs function through the induction of the apoptotic program, we hypothesized that the RNAi-mediated knockdown of MK-STYX blocks the cellular response to chemotherapeutic-induced apoptosis. </p><p> To investigate this hypothesis, we determined the ability of both control and MK-STYX knockdown cells to undergo apoptosis after exposure to an array of cell death inducing agents with different mechanisms of action. The results of these experiments demonstrated that MK-STYX knockdown protects against intrinsic, but not extrinsic apoptotic stimuli. These data were recapitulated with knockdown of the pro-apoptotic genes caspase-9 and Bax/Bak, suggesting that MK-STYX may modulate the regulation of one of these key apoptotic regulatory nodes. We demonstrated that the loss of MK-STYX blocks cytochrome c release, placing the apoptotic deficiency at the level of Bax/Bak-mediated mitochondrial outer membrane permeabilization, or MOMP. MK-STYX was found to localize to the mitochondria, but is neither released from the mitochondria upon apoptotic stress nor localized proximal to the machinery currently known to control MOMP. These results are summarized in Chapter 2. </p><p> In an effort to more fully define molecular mechanism of MK-STYX, we performed an unbiased TAP-tagging experiment to identify its interaction partners. The most significant and unique protein identified was the mitochondrial phosphatase PTPMT1. Interestingly, MK-STYX is a catalytically inactive dual specificity phosphatase, and catalytically inactive phosphatases have a precedent for regulating the activity and/or localization of active phosphatases. Because of this potential phosphatase regulatory mechanism, as well as similar localization patterns of both genes, we chose to further explore the interaction between PTPMT1 and MK-STYX. </p><p> Due to the robust survival phenotype seen in MK-STYX knockdown cells when treated with chemotherapeutic, we predicted that the knockdown of PTPMT1 may have a similar phenotype. Surprisingly, we found that PTPMT1 knockdown causes a Bax/Bak dependent cell death, suggesting that MK-STYX and PTPMT1 may functionally oppose one another in the mitochondria. Experiments in which both enzymes are downregulated show that PTPMT1 is epistatic to MK-STYX, as cells are resensitized to chemotherapeutic agents and cytochrome c release under these conditions. Interestingly, PTPMT1 was recently shown to be an important enzyme in the cardiolipin biosynthetic pathway, positively regulating the synthesis of this mitochondrial lipid. The genetic interaction provided by the robust changes in viability seen when these enzymes are downregulated suggests that MK-STYX may function to dampen PTPMT1 enzymatic activity. This allows us to hypothesize that the loss of MK-STYX results in increased cardiolipin biosynthesis, leading to altered mitochondrial membrane composition and subsequently, an altered apoptotic response. These results are summarized in Chapters 3 and 4. </p><p> We further hypothesize that the upregulation of cardiolipin levels directly inhibits the ability of Bax/Bak to permeabilize the outer mitochondrial membrane, effectively blocking the induction of mitochondrial apoptosis. These data suggest a novel mechanism by which dysregulated cardiolipin can facilitate chemoresistance, and suggest that this pathway could be exploited by recurrent cancers to evade therapies.</p><p>

Identiferoai:union.ndltd.org:PROQUEST/oai:pqdtoai.proquest.com:10274627
Date21 July 2017
CreatorsNiemi, Natalie Marie
PublisherVan Andel Research Institute
Source SetsProQuest.com
LanguageEnglish
Detected LanguageEnglish
Typethesis

Page generated in 0.1728 seconds