Abstract

BackgroundHexose-6-Phosphate Dehydrogenase (H6PD) is a generator of NADPH in the Endoplasmic/Sarcoplasmic Reticulum (ER/SR). Interaction of H6PD with 11β-hydroxysteroid dehydrogenase type 1 provides NADPH to support oxo-reduction of inactive to active glucocorticoids, but the wider understanding of H6PD in ER/SR NAD(P)(H) homeostasis is incomplete. Lack of H6PD results in a deteriorating skeletal myopathy, altered glucose homeostasis, ER stress and activation of the unfolded protein response. Here we further assess muscle responses to H6PD deficiency to delineate pathways that may underpin myopathy and link SR redox status to muscle wide metabolic adaptation.MethodsWe analysed skeletal muscle from H6PD knockout (H6PDKO), H6PD and NRK2 double knockout (DKO) and wild-type (WT) mice. H6PDKO mice were supplemented with the NAD+ precursor nicotinamide riboside. Skeletal muscle samples were subjected to biochemical analysis including NAD(H) measurement, LC-MS based metabolomics, Western blotting, and high resolution mitochondrial respirometry. Genetic and supplement models were assessed for degree of myopathy compared to H6PDKO.ResultsH6PDKO skeletal muscle showed adaptations in the routes regulating nicotinamide and NAD+ biosynthesis, with significant activation of the Nicotinamide Riboside Kinase 2 (NRK2) pathway. Associated with changes in NAD+ biosynthesis, H6PDKO muscle had impaired mitochondrial respiratory capacity with altered mitochondrial acylcarnitine and acetyl-CoA metabolism. Boosting NAD+ levels through the NRK2 pathway using the precursor nicotinamide riboside elevated NAD+/NADH but had no effect to mitigate ER stress and dysfunctional mitochondrial respiratory capacity or acetyl-CoA metabolism. Similarly, H6PDKO/NRK2 double KO mice did not display an exaggerated timing or severity of myopathy or overt change in mitochondrial metabolism despite depression of NAD+ availability.ConclusionsThese findings suggest a complex metabolic response to changes in muscle SR NADP(H) redox status that result in impaired mitochondrial energy metabolism and activation of cellular NAD+ salvage pathways. It is possible that SR can sense and signal perturbation in NAD(P)(H) that cannot be rectified in the absence of H6PD. Whether NRK2 pathway activation is a direct response to changes in SR NAD(P)(H) availability or adaptation to deficits in metabolic energy availability remains to be resolved.

Highlights

  • Hexose-6-Phosphate Dehydrogenase (H6PD) is a generator of NADPH in the Endoplasmic/ Sarcoplasmic Reticulum (ER/sarcoplasmic reticulum (SR))

  • We subjected Tibialis Anterior (TA) skeletal muscle from WT and H6PD knockout (H6PDKO) mice to an unbiased metabolomic screen performed by LCMS to better understand the role H6PD plays in muscle cell metabolism and SR redox maintenance

  • Nicotinamide Riboside Kinase 2 (NRK2) is dispensable in H6PDKO myopathy We investigated the role of the NRK2 pathway in defending muscle function in the absence of H6PD using double knockout (DKO) mice

Read more

Summary

Introduction

Hexose-6-Phosphate Dehydrogenase (H6PD) is a generator of NADPH in the Endoplasmic/ Sarcoplasmic Reticulum (ER/SR). Interaction of H6PD with 11β-hydroxysteroid dehydrogenase type 1 provides NADPH to support oxo-reduction of inactive to active glucocorticoids, but the wider understanding of H6PD in ER/ SR NAD(P)(H) homeostasis is incomplete. A major enzyme that reduces NADP+ within the SR lumen is hexose-6-phosphate dehydrogenase (H6PD) which oxidises glucose-6-phosphate derived from glycolysis to generate NADPH [5, 6]. H6PD NADPH generation supports 11β-HSD1 oxoreductase activity for glucocorticoid production [3]. In ACRD, the lack of H6PD and its associated NADPH production switches 11β-HSD1 activity towards glucocorticoid oxidation (and NADP+ reduction) resulting in increased tissue glucocorticoid clearance and relative insensitivity. H6PD knockout (H6PDKO) mice reflect this biochemistry, manifesting an ACRD-like phenotype, showing tissue glucocorticoid insensitivity and HPA axis activation [6]. The liver has impaired ability to stimulate gluconeogenesis, with increased glycogen synthesis rates, while adipose tissue has impaired ability to store and mobilise lipid [11, 12]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call