Files

Abstract

Mitochondrial diseases are rare and severe conditions with debilitating symptoms. Biochemical defects in mitochondria however are common. The difference between these two frequencies is suspected to lie in the capability of the cells to adapt to the homeostatic disbalance. Cells activate retrograde signalling pathways in response to mitochondrial dysfunction, activating cellular transcriptional programs to correct for the disbalance. The full extent of the retrograde signals that are induced by mitochondrial dysfunctions still remains unknown. Studying these retrograde signalling pathways can lead to insights how to make the difference between restoring the homeostasis and a detrimental disease. Investigating the transcriptional responses to artificially induced mitochondrial dysfunction has proven to be lucrative in unveiling novel responses to mitochondrial defects. In this work we studied the transcriptional responses to three chemical models of mitochondrial respiratory chain dysfunction. In line with recently published work, we found a commonly upregulated stress response in all models. Interestingly, shared between all the models was a downregulation of genes enriched in cholesterol biosynthesis pathways. Upon closer examination this encompassed, almost without exception, all transcripts of the mevalonate pathway. This coordinated downregulation of transcripts had functional consequences on the output of the pathway, lowering the abundance of cholesterol precursor metabolites. Other metabolites in more upstream regions of the pathway were changed differently depending on the complex affected, suggesting other regulatory influences involved. Upon closer mechanistic investigation to the cause of the transcriptional suppression we found the main transcription factor responsible for mevalonate pathway gene transcription, the Sterol Response Element Binding Protein 2 (SREBP2), to be inhibited during complex I impairment. This was most evident in sterol depleted conditions where SREBP2 did not activate to normal levels. A decreased activation of SREBP2 is signature for an elevated concentration of ER-cholesterol. Indeed, other ER-bound cholesterol sensitive proteins confirmed this, showing a similar decreased abundance following complex I impairment in both normal and low lipid conditions. To investigate the source of this cholesterol we measured various cholesterol levels. No changes in the levels of total cholesterol (free cholesterol plus esterified cholesterol) were detected. Also, the fraction of free cholesterol was not significantly affected by complex I inhibition. Using the fluorescent cholesterol stain filipin, we were able to measure a modest but reproducible increase in intracellular free cholesterol. This provided novel mechanistic insight into the inhibition of ER sterol sensors and mevalonate pathway activity by mitochondrial dysfunction. Several well-known retrograde signalling pathways AMPK, ISR and mTOR did not explain the changes observed. The exact mechanism through which mitochondria incite an increase in intracellular free cholesterol remains unknown. Taken together this work can provide a mechanistic link between mitochondrial function and intracellular cholesterol homeostasis and could possibly link mitochondrial function to circulating lipoprotein homeostasis. Further work should focus on elucidating the possible mechanism, where differences in complex functions could provide a hint in the right direction.

Details

PDF