[HTML][HTML] Screen for chemical modulators of autophagy reveals novel therapeutic inhibitors of mTORC1 signaling

AD Balgi, BD Fonseca, E Donohue, TCF Tsang… - PloS one, 2009 - journals.plos.org
AD Balgi, BD Fonseca, E Donohue, TCF Tsang, P Lajoie, CG Proud, IR Nabi, M Roberge
PloS one, 2009journals.plos.org
Background Mammalian target of rapamycin complex 1 (mTORC1) is a protein kinase that
relays nutrient availability signals to control numerous cellular functions including
autophagy, a process of cellular self-eating activated by nutrient depletion. Addressing the
therapeutic potential of modulating mTORC1 signaling and autophagy in human disease
requires active chemicals with pharmacologically desirable properties.
Methodology/Principal Findings Using an automated cell-based assay, we screened a …
Background
Mammalian target of rapamycin complex 1 (mTORC1) is a protein kinase that relays nutrient availability signals to control numerous cellular functions including autophagy, a process of cellular self-eating activated by nutrient depletion. Addressing the therapeutic potential of modulating mTORC1 signaling and autophagy in human disease requires active chemicals with pharmacologically desirable properties.
Methodology/Principal Findings
Using an automated cell-based assay, we screened a collection of >3,500 chemicals and identified three approved drugs (perhexiline, niclosamide, amiodarone) and one pharmacological reagent (rottlerin) capable of rapidly increasing autophagosome content. Biochemical assays showed that the four compounds stimulate autophagy and inhibit mTORC1 signaling in cells maintained in nutrient-rich conditions. The compounds did not inhibit mTORC2, which also contains mTOR as a catalytic subunit, suggesting that they do not inhibit mTOR catalytic activity but rather inhibit signaling to mTORC1. mTORC1 inhibition and autophagosome accumulation induced by perhexiline, niclosamide or rottlerin were rapidly reversed upon drug withdrawal whereas amiodarone inhibited mTORC1 essentially irreversibly. TSC2, a negative regulator of mTORC1, was required for inhibition of mTORC1 signaling by rottlerin but not for mTORC1 inhibition by perhexiline, niclosamide and amiodarone. Transient exposure of immortalized mouse embryo fibroblasts to these drugs was not toxic in nutrient-rich conditions but led to rapid cell death by apoptosis in starvation conditions, by a mechanism determined in large part by the tuberous sclerosis complex protein TSC2, an upstream regulator of mTORC1. By contrast, transient exposure to the mTORC1 inhibitor rapamycin caused essentially irreversible mTORC1 inhibition, sustained inhibition of cell growth and no selective cell killing in starvation.
Conclusion/Significance
The observation that drugs already approved for human use can reversibly inhibit mTORC1 and stimulate autophagy should greatly facilitate the preclinical and clinical testing of mTORC1 inhibition for indications such as tuberous sclerosis, diabetes, cardiovascular disease and cancer.
PLOS