Abstract

This study addressed the hypothesis that soluble epoxide hydrolase (sEH), which metabolizes endothelium-derived epoxyeicosatrienoic acids, plays a role in vascular calcification. The sEH inhibitor trans-4-(4-(3-adamantan-1-yl-ureido)-cyclohexyloxy)-benzoic acid (t-AUCB) potentiated the increase in calcium deposition of rat aortic rings cultured in high-phosphate conditions. This was associated with increased tissue-nonspecific alkaline phosphatase activity and mRNA expression level of the osteochondrogenic marker Runx2. The procalcifying effect of t-AUCB was prevented by mechanical aortic deendothelialization or inhibition of the production and action of epoxyeicosatrienoic acids using the cytochrome P450 inhibitor fluconazole and the antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE), respectively. Similarly, exogenous epoxyeicosatrienoic acids potentiated the calcification of rat aortic rings through a protein kinase A (PKA)-dependent mechanism and of human aortic vascular smooth muscle cells when sEH was inhibited by t-AUCB. Finally, a global gene expression profiling analysis revealed that the mRNA expression level of sEH was decreased in human carotid calcified plaques compared to adjacent lesion-free sites and was inversely correlated with Runx2 expression. These results show that sEH hydrolase plays a protective role against vascular calcification by reducing the bioavailability of epoxyeicosatrienoic acids.

Highlights

  • Vascular calcification is a highly regulated process observed during aging, hypertension, diabetes, and chronic kidney disease (CKD)

  • There was an increase in tissue-nonspecific alkaline phosphatase (TNAP) activity (Figure 1F) and a decrease in pyrophosphate (PPi) level (Figure 1G), an endogenous inhibitor of ectopic calcification which is hydrolyzed by TNAP [16,17,18], in the culture supernatants of high-phosphate-cultured aortic rings

  • T3h.8emdMecrPeai saenidnaSMcuMltuHrCe dmuRraNtiAonexopf r7esdsaioyns ilsevneeleadnedd tthoeinindcurceeasseiginniRficuannxt2a, oMrtsixc2caalncdifiScoatxi9onle[v1e6l]s. cTohnefidrmecerdeatsheeintrSaMnsMitiHonC omf RvNasAcuelxapr rceeslslsiotnowlevaredl aanndotshteeoincchroenadseroigneRnuicnxp2h,eMnostxy2paen. dInSoaxd9dlietvioenls, tchoenrfeirwmaesd athneitnrcarnesaitsieodn TofNvAasPcualcatrivcietlylsatnodwadredcraenaosesteinocthhoenldervoeglsenoifc pthheeninohtyibpiet.oIrnoafdcdailtcioifinc, athtieorne PwPais, aasn pinrecrveiaosuesdlyTsNhAowPnacitnivaitoyrtaansdisdoelactreedasferoinmthuerelmevicelsraotsf t[h1e4].inhTihbiistopraotfhwcaalcyifiiscaitniovnolvPePdi, ians mineralization deposition facilitating the formation of the final product of calcium phosphate salt reaction hydroxyapatite, but it may directly promote the transition of vascular smooth muscle cells (VSMCs) to a osteochondrogenic phenotype [31,32]

Read more

Summary

Introduction

Vascular calcification is a highly regulated process observed during aging, hypertension, diabetes, and chronic kidney disease (CKD). It contributes to elevated risk of cardiovascular events and mortality [1,2,3,4]. Increasing experimental evidence shows that the endothelium is a source of osteoprogenitor cells and regulates vascular calcification through the release of signaling molecules such as nitric oxide (NO), which prevents VSMC osteochondrogenic differentiation [5,6,7]. Based on the measurements of plasma DHET levels, a recent clinical study suggested that EETs may be protective against the development of vascular calcification [13]

Objectives
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