Searchable abstracts of presentations at key conferences in endocrinology
Endocrine Abstracts (2017) 50 OC1.3 | DOI: 10.1530/endoabs.50.OC1.3

1Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; 2Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK; 3Centre for Endocrinology, Queen Mary University of London, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, UK; 4School of Life Sciences, University of Warwick, Warwick, UK; 5School of Biosciences and Phenome Centre Birmingham, Birmingham, UK; 6Department of Endocrinology, University Hospital Wuerzburg, Wuerzburg, Germany.


Adrenocortical Carcinoma (ACC) is an aggressive malignancy with poor response to chemotherapy. Here we evaluated a potential new treatment target for ACC, focusing on the mitochondrial NADPH generator Nicotinamide Nucleotide Transhydrogenase (NNT). NNT has a central role within the mitochondrial antioxidant pathways, protecting cells from oxidative stress. Inactivating NNT mutations lead to isolated primary adrenal insufficiency, suggesting a selective vulnerability of adrenocortical cells to NNT loss. A TCGA database search confirmed increased NNT expression in ACC. Therefore, we hypothesised NNT silencing in ACC cells will induce toxic levels of oxidative stress. To explore this, we transiently knocked down NNT in NCI-H295R ACC cells by siRNA transfection. NNT silencing increased intracellular levels of oxidative stress; this resulted in a dramatic suppression of cell proliferation and higher apoptotic rates, as well as sensitising cells to chemically-induced oxidative stress. Steroidogenesis was paradoxically stimulated by NNT loss, as demonstrated by comprehensive steroid profiling. Next, we generated a stable NNT knockdown model in the same cell line (lentiviral shRNA transfection), to understand the chronic effects of NNT silencing. After culture for 1–3 months, cells adapted metabolically to stable NNT knockdown, restoring their redox balance and resilience to oxidative stress, although their proliferation remained suppressed. This was associated with higher rates of oxygen consumption. The molecular pathways underpinning the cellular response to transient and chronic NNT loss were explored in detail by RNA sequencing and whole-metabolome analysis. Transient NNT knockdown led to changes in core pathways controlling cellular proliferation and viability. Stable (chronic) knockdown was characterised by changes consistent with accelerated protein turnover and up-regulation of antioxidant polyamines, which can facilitate partial adaptation to oxidative stress. Our study provides the first pre-clinical evidence of the therapeutic merit of antioxidant targeting in ACC, as well as delineation of the long-term adaptive response of cells to oxidative stress.

Volume 50

Society for Endocrinology BES 2017

Harrogate, UK
06 Nov 2017 - 08 Nov 2017

Society for Endocrinology 

Browse other volumes

Article tools

My recent searches

No recent searches.