Abstract

Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a glycan-binding receptor bearing immunoreceptor tyrosine-based inhibitory and switch motifs (ITIM and ITSM, respectively) that is selectively expressed on eosinophils, mast cells, and, to a lesser extent, basophils. Previous work has shown that engagement of Siglec-8 on IL-5–primed eosinophils causes cell death via CD11b/CD18 integrin–mediated adhesion and NADPH oxidase activity and identified signaling molecules linking adhesion, reactive oxygen species (ROS) production, and cell death. However, the proximal signaling cascade activated directly by Siglec-8 engagement has remained elusive. Most members of the Siglec family possess similar cytoplasmic signaling motifs and recruit the protein tyrosine phosphatases SHP-1/2, consistent with ITIM-mediated signaling, to dampen cellular activation. However, the dependence of Siglec-8 function in eosinophils on these phosphatases has not been studied. Using Siglec-8 antibody engagement and pharmacological inhibition in conjunction with assays to measure cell-surface upregulation and conformational activation of CD11b integrin, ROS production, and cell death, we sought to identify molecules involved in Siglec-8 signaling and determine the stage of the process in which each molecule plays a role. We demonstrate here that the enzymatic activities of Src family kinases (SFKs), Syk, SHIP1, PAK1, MEK1, ERK1/2, PLC, PKC, acid sphingomyelinase/ceramidase, and Btk are all necessary for Siglec-8–induced eosinophil cell death, with no apparent role for SHP-1/2, SHIP2, or c-Raf. While most of these signaling molecules are necessary for Siglec-8–induced upregulation of CD11b integrin at the eosinophil cell surface, Btk is phosphorylated and activated later in the signaling cascade and is instead necessary for CD11b activation. In contrast, SFKs and ERK1/2 are phosphorylated far earlier in the process, consistent with their role in augmenting cell-surface levels of CD11b. In addition, pretreatment of eosinophils with latrunculin B or jasplakinolide revealed that actin filament disassembly is necessary and sufficient for surface CD11b integrin upregulation and that actin polymerization is necessary for downstream ROS production. These results show that Siglec-8 signals through an unanticipated set of signaling molecules in IL-5–primed eosinophils to induce cell death and challenges the expectation that ITIM-bearing Siglecs signal through inhibitory pathways involving protein tyrosine phosphatases to achieve their downstream functions.

Highlights

  • Sialic acid-binding immunoglobulin-like lectin (Siglec) are cell surface proteins found predominantly, but not exclusively, on the surface of immune cells and are characterized by their propensity to bind sialic acid-containing structures

  • We demonstrate that Siglec-8 induces a signaling cascade that is uncharacteristic for an immunoreceptor tyrosine-based inhibitory motif (ITIM)-bearing receptor, involving the activities of Src family kinases (SFKs), Syk, PI3K, SHIP1, Rac1, phospholipase C (PLC), PKC, acid sphingomyelinase/ceramidase, p21 (Rac1)–activated kinase 1 (PAK1), MEK1, ERK1/2, and Bruton’s tyrosine kinase (Btk) as well as actin rearrangement

  • There was an acceleration of tyrosine phosphorylation events due to Siglec-8 engagement in IL-5–primed eosinophils and a band below 100 kDa in molecular weight that becomes prominent at 120 min in these cells

Read more

Summary

Introduction

Siglecs (sialic acid-binding immunoglobulin-like lectins) are cell surface proteins found predominantly, but not exclusively, on the surface of immune cells and are characterized by their propensity to bind sialic acid-containing structures. Siglec-8 is selectively expressed on the surface of human eosinophils and contains two cytoplasmic signaling motifs—an intracellular immunoreceptor tyrosine-based inhibitory motif (ITIM) and an immunoreceptor tyrosine-based switch motif (ITSM)— thought to be responsible for inhibitory signal transduction [2,3,4]. A rapidly mobilizable pool of intracellular CD11b/CD18, like the one trafficked to the cell membrane in response to Siglec-8 ligation [8], is known to exist in secretory vesicles in eosinophils but may be associated with granule membranes as well [9]. The source of the intracellular pool of CD11b/CD18 mobilized by Siglec-8 signaling has not been determined

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call