Epigenetic regulation of vascular smooth muscle cell proliferation and neointima formation by histone deacetylase inhibition

HM Findeisen, F Gizard, Y Zhao, H Qing… - … , and vascular biology, 2011 - Am Heart Assoc
HM Findeisen, F Gizard, Y Zhao, H Qing, EB Heywood, KL Jones, D Cohn, D Bruemmer
Arteriosclerosis, thrombosis, and vascular biology, 2011Am Heart Assoc
Objective—Proliferation of smooth muscle cells (SMC) in response to vascular injury is
central to neointimal vascular remodeling. There is accumulating evidence that histone
acetylation constitutes a major epigenetic modification for the transcriptional control of
proliferative gene expression; however, the physiological role of histone acetylation for
proliferative vascular disease remains elusive. Methods and Results—In the present study,
we investigated the role of histone deacetylase (HDAC) inhibition in SMC proliferation and …
Objective
Proliferation of smooth muscle cells (SMC) in response to vascular injury is central to neointimal vascular remodeling. There is accumulating evidence that histone acetylation constitutes a major epigenetic modification for the transcriptional control of proliferative gene expression; however, the physiological role of histone acetylation for proliferative vascular disease remains elusive.
Methods and Results
In the present study, we investigated the role of histone deacetylase (HDAC) inhibition in SMC proliferation and neointimal remodeling. We demonstrate that mitogens induce transcription of HDAC 1, 2, and 3 in SMC. Short interfering RNA–mediated knockdown of either HDAC 1, 2, or 3 and pharmacological inhibition of HDAC prevented mitogen-induced SMC proliferation. The mechanisms underlying this reduction of SMC proliferation by HDAC inhibition involve a growth arrest in the G1 phase of the cell cycle that is due to an inhibition of retinoblastoma protein phosphorylation. HDAC inhibition resulted in a transcriptional and posttranscriptional regulation of the cyclin-dependent kinase inhibitors p21Cip1 and p27Kip. Furthermore, HDAC inhibition repressed mitogen-induced cyclin D1 mRNA expression and cyclin D1 promoter activity. As a result of this differential cell cycle-regulatory gene expression by HDAC inhibition, the retinoblastoma protein retains a transcriptional repression of its downstream target genes required for S phase entry. Finally, we provide evidence that these observations are applicable in vivo by demonstrating that HDAC inhibition decreased neointima formation and expression of cyclin D1 in a murine model of vascular injury.
Conclusion
These findings identify HDAC as a critical component of a transcriptional cascade regulating SMC proliferation and suggest that HDAC might play a pivotal role in the development of proliferative vascular diseases, including atherosclerosis and in-stent restenosis.
Am Heart Assoc