Abstract

The high-energy sulfate donor 3′-phosphoadenosine-5′-phosphosulfate (PAPS), generated by human PAPS synthase isoforms PAPSS1 and PAPSS2, is required for all human sulfation pathways. Sulfotransferase SULT2A1 uses PAPS for sulfation of the androgen precursor dehydroepiandrosterone (DHEA), thereby reducing downstream activation of DHEA to active androgens. Human PAPSS2 mutations manifest with undetectable DHEA sulfate, androgen excess, and metabolic disease, suggesting that ubiquitous PAPSS1 cannot compensate for deficient PAPSS2 in supporting DHEA sulfation. In knockdown studies in human adrenocortical NCI-H295R1 cells, we found that PAPSS2, but not PAPSS1, is required for efficient DHEA sulfation. Specific APS kinase activity, the rate-limiting step in PAPS biosynthesis, did not differ between PAPSS1 and PAPSS2. Co-expression of cytoplasmic SULT2A1 with a cytoplasmic PAPSS2 variant supported DHEA sulfation more efficiently than co-expression with nuclear PAPSS2 or nuclear/cytosolic PAPSS1. Proximity ligation assays revealed protein–protein interactions between SULT2A1 and PAPSS2 and, to a lesser extent, PAPSS1. Molecular docking studies showed a putative binding site for SULT2A1 within the PAPSS2 APS kinase domain. Energy-dependent scoring of docking solutions identified the interaction as specific for the PAPSS2 and SULT2A1 isoforms. These findings elucidate the mechanistic basis for the selective requirement for PAPSS2 in human DHEA sulfation.

Highlights

  • The high-energy sulfate donor 3؅-phosphoadenosine-5؅phosphosulfate (PAPS), generated by human PAPS synthase isoforms PAPSS1 and PAPSS2, is required for all human sulfation pathways

  • In knockdown studies in human adrenocortical NCI-H295R1 cells, we found that PAPSS2, but not PAPSS1, is required for efficient DHEA sulfation

  • We used human adrenocortical NCI-H295R1 cells, which we found to express high levels of SULT2A1 mRNA (⌬CT value 13.4 Ϯ 1.3 relative to 18S rRNA, Ϯ S.D., see Table S1) and almost identical mRNA levels of PAPSS1 and PAPSS2 (⌬CT values 16.1 Ϯ 0.8 and 15.8 Ϯ 0.9, respectively)

Read more

Summary

Introduction

The high-energy sulfate donor 3؅-phosphoadenosine-5؅phosphosulfate (PAPS), generated by human PAPS synthase isoforms PAPSS1 and PAPSS2, is required for all human sulfation pathways. Energy-dependent scoring of docking solutions identified the interaction as specific for the PAPSS2 and SULT2A1 isoforms These findings elucidate the mechanistic basis for the selective requirement for PAPSS2 in human DHEA sulfation. 3Ј-phosphoadenosine-5Ј-phosphosulfate (PAPS) synthases, the enzymes responsible for sulfate activation, are represented by only two genes in humans, PAPSS1 and PAPSS2 [4]. This gene pair is evolutionary conserved in all vertebrate genomes investigated so far; RNA splice forms and additional teleost-specific gene duplications of PAPSS2 are the only exceptions [4]. The clinical phenotype of human loss-offunction mutations in the gene encoding PAPSS2 have suggested differential roles for PAPSS1 and PAPSS2 in human sul-

Methods
Results
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