PT - JOURNAL ARTICLE AU - Audrey Courboulin AU - Marjorie Barrier AU - Tanya Perreault AU - Pierre Bonnet AU - Veronique L. Tremblay AU - Roxane Paulin AU - Ève Tremblay AU - Caroline Lambert AU - Maria H. Jacob AU - Sandra N. Bonnet AU - Steeve Provencher AU - Sébastien Bonnet TI - Plumbagin reverses proliferation and resistance to apoptosis in experimental PAH AID - 10.1183/09031936.00084211 DP - 2012 Sep 01 TA - European Respiratory Journal PG - 618--629 VI - 40 IP - 3 4099 - http://erj.ersjournals.com/content/40/3/618.short 4100 - http://erj.ersjournals.com/content/40/3/618.full SO - Eur Respir J2012 Sep 01; 40 AB - Like cancer, pulmonary arterial hypertension (PAH) is characterised by a pro-proliferative and anti-apoptotic phenotype. In PAH, pulmonary artery smooth muscle cell (PASMC) proliferation is enhanced and apoptosis suppressed. The sustainability of this phenotype requires the activation of pro-survival transcription factors, such as signal transducer and activator of transcription (STAT)3 and nuclear factor of activated T-cells (NFAT). There are no drugs currently available that are able to efficiently and safely inhibit this axis. We hypothesised that plumbagin (PLB), a natural organic compound known to block STAT3 in cancer cells, would reverse experimental pulmonary hypertension. Using human PAH-PASMC, we demonstrated in vitro that PLB inhibits the activation of the STAT3/NFAT axis, increasing the voltage-gated K+ current bone morphogenetic protein receptor type II (BMPR2), and decreasing intracellular Ca2+ contentration ([Ca2+]i), rho-associated coiled-coil containing protein kinase (ROCK)1 and interleukin (IL)-6, contributing to the inhibition of PAH-PASMC proliferation and resistance to apoptosis (proliferating cell nuclear antigen (PCNA), TUNEL, Ki67 and anexine V). In vivo, PLB oral administration decreases distal pulmonary artery remodelling, mean pulmonary artery pressure and right ventricular hypertrophy without affecting systemic circulation in both monocrotaline- and sugen/chronic hypoxia-induced PAH in rats. This study demonstrates that the STAT3/NFAT axis can be therapeutically targeted by PLB in human PAH-PASMC and experimental PAH rat models. Thus, PLB could be considered a specific and attractive future therapeutic strategy for PAH.