[HTML][HTML] Phospholipase D1 mediates TNFα-induced inflammation in a murine model of TNFα-induced peritonitis

S Sethu, PN Pushparaj, AJ Melendez - PloS one, 2010 - journals.plos.org
S Sethu, PN Pushparaj, AJ Melendez
PloS one, 2010journals.plos.org
Background Tumor Necrosis Factor alpha (TNFα) is a pleiotropic cytokine extensively
studied for its role in the pathogenesis of a variety of disease conditions, including in
inflammatory diseases. We have recently shown that, in vitro, that TNFα utilizes PLD1 to
mediate the activation of NFκB and ERK1/2 in human monocytes. The aim of this study was
to investigate the role (s) played by phospholipase D1 (PLD1) in TNFα-mediated
inflammatory responses in vivo. Methodology/Findings Studies were performed in vivo using …
Background
Tumor Necrosis Factor alpha (TNFα) is a pleiotropic cytokine extensively studied for its role in the pathogenesis of a variety of disease conditions, including in inflammatory diseases. We have recently shown that, in vitro, that TNFα utilizes PLD1 to mediate the activation of NFκB and ERK1/2 in human monocytes. The aim of this study was to investigate the role(s) played by phospholipase D1 (PLD1) in TNFα-mediated inflammatory responses in vivo.
Methodology/Findings
Studies were performed in vivo using a mouse model of TNFα-induced peritonitis. The role of PLD1 was investigated by functional genomics, utilizing a specific siRNA to silence the expression of PLD1. Administration of the siRNA against PLD1 significantly reduced PLD1 levels in vivo. TNFα triggers a rapid pyrogenic response, but the in vivo silencing of PLD1 protects mice from the TNFα-induced rise in temperature. Similarly TNFα caused an increase in the serum levels of IL-6, MIP-1α and MIP-1β: this increase in cytokine/chemokine levels was inhibited in mice where PLD1 had been silenced. We then induced acute peritonitis with TNFα. Intraperitoneal injection of TNFα triggered a rapid increase in vascular permeability, and the influx of neutrophils and monocytes into the peritoneal cavity. By contrast, in mice where PLD1 had been silenced, the TNFα-triggered increase in vascular permeability and phagocyte influx was substantially reduced. Furthermore, we also show that the TNFα-mediated upregulation of the cell adhesion molecules VCAM and ICAM1, in the vascular endothelium, were dependent on PLD1.
Conclusions
These novel data demonstrate a critical role for PLD1 in TNFα-induced inflammation in vivo and warrant further investigation. Indeed, our results suggest PLD1 as a novel target for treating inflammatory diseases, where TNFα play key roles: these include diseases ranging from sepsis to respiratory and autoimmune diseases; all diseases with considerable unmet medical need.
PLOS