Abstract

Mast cells are the major effector cells in immunoglobulin E (IgE)-mediated allergy. The high affinity IgE receptor FcεRI, as well as G protein-coupled receptors (GPCRs) on the mast cell surface signals to phosphoinositide 3-kinase γ (PI3Kγ) to initiate degranulation, cytokine release, and chemotaxis. PI3Kγ is therefore considered as a target for treatment of allergic disorders. However, leukocyte PI3Kγ is key to many functions in innate and adaptive immunity, and attenuation of host defense mechanisms is an expected adverse effect that complicates treatment of chronic illnesses. PI3Kγ operates as a p110γ/p84 or p110γ/p101 complex, where p110γ/p84 requires Ras activation. Here we investigated if modulation of Ras-isoprenylation could target PI3Kγ activity to attenuate PI3Kγ-dependent mast cell responses without impairment of macrophage functions. In murine bone marrow-derived mast cells, GPCR stimulation triggers activation of N-Ras and H-Ras isoforms, which is followed by the phosphorylation of protein kinase B (PKB/Akt) relayed through PI3Kγ. Although K-Ras is normally not activated in Ras wild-type cells, it is able to compensate for genetically deleted N- and H-Ras isoforms. Inhibition of Ras isoprenylation with farnesyltransferase inhibitor FTI-277 leads to a significant reduction of mast cell degranulation, cytokine production, and migration. Complementation experiments expressing PI3Kγ adaptor proteins p84 or p101 demonstrated a differential sensitivity towards Ras-inhibition depending on PI3Kγ complex composition. Mast cell responses are exclusively p84-dependent and were effectively controlled by FTI-277. Similar results were obtained when GTP-Ras was inactivated by overexpression of the GAP-domain of Neurofibromin-1 (NF-1). Unlike mast cells, macrophages express p84 and p101 but are p101-dominated and thus remain functional under treatment with FTI-277. Our work demonstrates that p101 and p84 have distinct physiological roles, and that Ras dependence of PI3Kγ signaling differs between cell types. FTI-277 reduces GPCR-activated PI3Kγ responses in p84-expressing but not p101-containing bone marrow derived cells. However, prenylation inhibitors have pleiotropic effects beyond Ras and non-tolerable side-effects that disfavor further clinical validation. Statins are, however, clinically well-established drugs that have previously been proposed to block mast cell degranulation by interference with protein prenylation. We show here that Simvastatin inhibits mast cell degranulation, but that this does not occur via Ras-PI3Kγ pathway alterations.

Highlights

  • Mast cells are key effector cells in the pathology of allergic disease and chronic inflammation and are target cells of novel therapeutic strategies [1]

  • We investigated which Ras isoforms would qualify for phosphoinositide 3-kinase g (PI3Kg) signaling in mast cells

  • The specific action of GGTI-298 on geranylgeranylation was confirmed by the accumulation of ungeranyl-geranylated Rap1A, while prelamin A did not accumulate in its non-farnesylated form in mast cells and macrophages (Figure S1). These results suggest that GGTI-298 does not affect PI3Kg signaling in mast cells by interference with Ras activation, but is likely to intercept upstream activation by geranyl-geranylated Gg subunit subtypes, which seems to be set that Simvastatin blocks degranulation by pleiotropic action and up differently in macrophages

Read more

Summary

Introduction

Mast cells are key effector cells in the pathology of allergic disease and chronic inflammation and are target cells of novel therapeutic strategies [1]. Multivalent antigen/IgE complexes cross-link FceRI, and trigger a protein tyrosine kinase cascade involving Src family kinase activation and Syk (spleen tyrosine kinase) translocation. We have demonstrated earlier that G-protein coupled receptor (GPCR) ligands synergize with IgE/antigen to stimulate mast cell degranulation [4,5,6]. IgE/antigen stimulated mast cells release adenosine, which yields an autocrine augmentation of mast cell activation via the Gai-coupled A3 adenosine receptor [7], triggering activation of phosphoinositide 3-kinase g [PI3Kg [4,5,6]]. An alternative activation mechanism downstream of FceRI involves the Ca2+-mediated activation of protein kinase C b (PKCb), which phosphorylates and activates PI3Kg [8]

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