Sepsis-induced potentiation of peritoneal macrophage migration is mitigated by programmed cell death receptor-1 gene deficiency

A Ayala, GF Elphick, YS Kim, X Huang… - Journal of innate …, 2014 - karger.com
A Ayala, GF Elphick, YS Kim, X Huang, A Carreira-Rosario, SC Santos, NJ Shubin, Y Chen…
Journal of innate immunity, 2014karger.com
The effect of programmed cell death receptor-1 (PD-1) on phagocyte function has not been
extensively described. Here we report that experimental mouse sepsis, cecal ligation and
puncture (CLP), induced a marked increase in peritoneal macrophage random migration,
motility and cell spread, but these changes were lost in the absence of PD-1. Alternatively,
phagocytic activity was inversely affected. In vitro cell culture imaging studies, with the
macrophage cell line J774, documented that blocking PD-1 with antibody led to aggregation …
Abstract
The effect of programmed cell death receptor-1 (PD-1) on phagocyte function has not been extensively described. Here we report that experimental mouse sepsis, cecal ligation and puncture (CLP), induced a marked increase in peritoneal macrophage random migration, motility and cell spread, but these changes were lost in the absence of PD-1. Alternatively, phagocytic activity was inversely affected. In vitro cell culture imaging studies, with the macrophage cell line J774, documented that blocking PD-1 with antibody led to aggregation of the cytoskeletal proteins α-actinin and F-actin. Further experiments looking at ex vivo peritoneal macrophages from mice illustrated that a similar pattern of α-actinin and F-actin was evident on cells from wild-type CLP mice but not PD-1-/-CLP mouse cells. We also observed that fMLP-induced migration by J774 cells was markedly attenuated using PD-1 blocking antibodies, a nonselective phosphatase inhibitor and a selective Ras-related protein 1 inhibitor. Finally, peritoneal macrophages derived from CLP as opposed to Sham mice demonstrated aspects of both cell surface co-localization with CD11b and internalization of PD-1 within vacuoles independent of CD11b staining. Together, we believe the data support a role for PD-1 in mediating aspects of innate macrophage immune dysfunction during sepsis, heretofore unappreciated.
Karger