Abstract

Coenzyme Q (CoQ) is a lipid present in all cell membranes. One of the multiple metabolic functions of CoQ is to transport electrons in the reaction catalyzed by sulfide:quinone oxidoreductase (SQOR), the first enzyme of the oxidation pathway of sulfides (hydrogen sulfide, H2S). Early evidence of a defect in the metabolism of H2S in primary CoQ deficiency came from yeast studies in Schizosaccharomyces pombe strains defective for dps1 and ppt1 (homologs of PDSS1 and COQ2, respectively), which have H2S accumulation. Our recent studies in human skin fibroblasts and in murine models of primary CoQ deficiency show that, also in mammals, decreased CoQ levels cause impairment of H2S oxidation. Patient fibroblasts carrying different mutations in genes encoding proteins involved in CoQ biosynthesis show reduced SQOR activity and protein levels proportional to the levels of CoQ. In Pdss2kd/kd mice, kidney, the only organ clinically affected, shows reduced SQOR levels and downstream enzymes, accumulation of H2S, and glutathione depletion. Pdss2kd/kd mice have also low levels of thiosulfate in plasma and urine, and increased C4–C6 acylcarnitines in blood, due to inhibition of short-chain acyl-CoA dehydrogenase. Also in Coq9R239X mice, the symptomatic organ, cerebrum, shows accumulation of H2S, reduced SQOR, increase in thiosulfate sulfurtransferase and sulfite oxidase, and reduction in the levels of glutathione and glutathione enzymes, leading to alteration of the biosynthetic pathways of glutamate, serotonin, and catecholamines. Coq9R239X mice have also reduced blood pressure, possible consequence of H2S-induced vasorelaxation. Since liver is not clinically affected in Pdss2 and Coq9 mutant mice, the effects of the impairment of H2S oxidation in this organ were not investigated, despite its critical role in metabolism. In conclusion, in vitro and in vivo studies of CoQ deficient models provide evidence of tissue-specific H2S oxidation impairment, an additional pathomechanism that should be considered in the understanding and treatment of primary CoQ deficiency.

Highlights

  • SULFIDE METABOLISM AND MITOCHONDRIASulfide metabolism in mammalian cells includes the transsulfuration and the hydrogen sulfide (H2S) oxidation pathways

  • The enzymes involved in the mitochondrial H2S oxidation pathway are sulfide:quinone oxidoreductase (SQOR), sulfur dioxygenase (SDO; known as ETHE1 or persulfide dioxygenase), sulfite oxidase (SO), thiosulfate sulfurtransferase or rhodanese (TST), and thiosulfate reductase (TR) (Tables A1, A2)

  • We investigated the tissue-specific effects of Coenzyme Q (CoQ) deficiency on H2S oxidation in three mouse models with different phenotype associated to CoQ deficiency: Pdss2kd/kd mice, which carry a spontaneous mutation in Pdss2, which encodes the subunit 2 of polyprenyl-diphosphate synthase, the first enzyme of CoQ biosynthesis (Peng et al, 2004; Saiki et al, 2005), and two knock-in mice harboring mutation in Coq9 (Garcia-Corzo et al, 2013; Luna-Sanchez et al, 2015), which encodes COQ9, a protein that interact with COQ7, the enzyme responsible for the hydroxylation of demethoxyubiquinone to 5-hydroxyquinone (Figure 2; Garcia-Corzo et al, 2013)

Read more

Summary

Introduction

SULFIDE METABOLISM AND MITOCHONDRIASulfide metabolism in mammalian cells includes the transsulfuration (biosynthetic) and the hydrogen sulfide (H2S) oxidation (catabolic) pathways. In Pdss2kd/kd mice, kidney, the only organ clinically affected, shows reduced SQOR levels and downstream enzymes, accumulation of H2S, and glutathione depletion.

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