Abstract

Accelerated atherosclerotic calcification is responsible for plaque burden, especially in diabetes. The regulatory mechanism for atherosclerotic calcification in diabetes is poorly characterized. Here we show that deletion of PARP-1, a main enzyme in diverse metabolic complications, attenuates diabetic atherosclerotic calcification and decreases vessel stiffening in mice through Runx2 suppression. Specifically, PARP-1 deficiency reduces diabetic arteriosclerotic calcification by regulating Stat1-mediated synthetic phenotype switching of vascular smooth muscle cells and macrophage polarization. Meanwhile, both vascular smooth muscle cells and macrophages manifested osteogenic differentiation in osteogenic media, which was attenuated by PARP-1/Stat1 inhibition. Notably, Stat1 acts as a positive transcription factor by directly binding to the promoter of Runx2 and promoting atherosclerotic calcification in diabetes. Our results identify a new function of PARP-1, in which metabolism disturbance-related stimuli activate the Runx2 expression mediated by Stat1 transcription to facilitate diabetic arteriosclerotic calcification. PARP-1 inhibition may therefore represent a useful therapy for this challenging complication.

Highlights

  • Vascular calcification is prevalent in patients suffering from diabetes and has been strongly associated with adverse cardiovascular outcome[1,2]

  • We demonstrate for the first time that Poly [ADP-ribose] polymerase 1 (PARP-1) plays a novel role in the regulation of osteogenic differentiation in both vascular smooth muscle cells (VSMCs) and macrophages regardless of the high glucose (HG) milieu

  • Our results suggest that PARP-1 regulates hyperglycemia-accelerated arteriosclerotic calcification by targeting Stat1/Runx[2] axis

Read more

Summary

Introduction

Vascular calcification is prevalent in patients suffering from diabetes and has been strongly associated with adverse cardiovascular outcome[1,2]. Arteriosclerotic calcification, which represents a significant predictor of susceptibility to plaque rupture, is directly driven by the osteogenic differentiation of VSMCs and the. Runx[2] binds to the osteoblast-specific cis-acting element 2, which is found in the promoter regions of both osteocalcin and alkaline phosphatase[9], and regulates their expression. This may explain the requirement for Runx[2] as a molecular switch for osteogenic differentiation and the inability of any parallel pathway to overcome its absence.

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.