Inactivation of the tumour suppressor, PTEN, in smooth muscle promotes a pro-inflammatory phenotype and enhances neointima formation

SB Furgeson, PA Simpson, I Park… - Cardiovascular …, 2010 - academic.oup.com
SB Furgeson, PA Simpson, I Park, V VanPutten, H Horita, CD Kontos, RA Nemenoff…
Cardiovascular research, 2010academic.oup.com
Aims Phosphatase and tensin homolog (PTEN) is implicated as a negative regulator of
vascular smooth muscle cell (SMC) proliferation and injury-induced vascular remodelling.
We tested if selective depletion of PTEN only in SMC is sufficient to promote SMC
phenotypic modulation, cytokine production, and enhanced neointima formation. Methods
and results Smooth muscle marker expression and induction of pro-inflammatory cytokines
were compared in cultured SMC expressing control or PTEN-specific shRNA. Compared …
Aims
Phosphatase and tensin homolog (PTEN) is implicated as a negative regulator of vascular smooth muscle cell (SMC) proliferation and injury-induced vascular remodelling. We tested if selective depletion of PTEN only in SMC is sufficient to promote SMC phenotypic modulation, cytokine production, and enhanced neointima formation.
Methods and results
Smooth muscle marker expression and induction of pro-inflammatory cytokines were compared in cultured SMC expressing control or PTEN-specific shRNA. Compared with controls, PTEN-deficient SMC exhibited increased phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signalling and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activity, reduced expression of SM markers (SM-α-actin and calponin), and increased production of stromal cell-derived factor-1α (SDF-1α), monocyte chemotactic protein-1 (MCP-1), interleukin-6 (IL-6), and chemokine (C-X-C motif) ligand 1 (KC/CXCL1) under basal conditions. PI3K/Akt or mTOR inhibition reversed repression of SM marker expression, whereas PI3K/Akt or NF-κB inhibition blocked cytokine induction mediated by PTEN depletion. Carotid ligation in mice with genetic reduction of PTEN specifically in SMC (SMC-specific PTEN heterozygotes) resulted in enhanced neointima formation, increased SMC hyperplasia, reduced SM-α-actin and calponin expression, and increased NF-κB and cytokine expression compared with wild-types. Lesion formation in SMC-specific heterozygotes was similar to lesion formation in global PTEN heterozygotes, indicating that inactivation of PTEN exclusively in SMC is sufficient to induce considerable increases in neointima formation.
Conclusion
PTEN activation specifically in SMC is a common upstream regulator of multiple downstream events involved in pathological vascular remodelling, including proliferation, de-differentiation, and production of multiple cytokines.
Oxford University Press