Microparticles released by human neutrophils adhere to erythrocytes in the presence of complement

https://doi.org/10.1016/j.yexcr.2005.03.011Get rights and content

Abstract

The release of cell surface-derived microparticles, or ectosomes, has now been described for many different cell types. In various diseases characterized by systemic inflammation, the numbers of ectosomes released from specific cell-types are found increased manifold in the circulation. Their pro-inflammatory and pro-coagulant functions make them potentially important actors in disease establishment and/or progression. Until now, ectosomes have been believed to be free in the circulation. Herein, we provide evidence for sequestration of ectosomes derived from human polymorphonuclear neutrophils to erythrocytes, similarly to immune complexes. We show that ectosomes activate and bind complement in vitro. In whole blood, opsonization of ectosomes by complement mediated their immune adherence to erythrocytes through complement receptor 1. Taken together, our data suggest an important role for complement and erythrocytes in the sequestration, and possibly clearance, of blood-borne ectosomes stemming from neutrophils. The immune adherence described here may modify the biological activity and function of ectosomes.

Introduction

When challenging polymorphonuclear leukocytes (PMN) with sublytic amounts of complement, Stein and Luzio observed the release of vesicles that pinched off directly from the cell membrane (ectocytosis) [1]. A similar phenomenon occurs in many different cell types in response to a variety of stimuli. Ectocytosis occurs immediately after stimulation and is therefore timely and mechanistically distinct from apoptosis. Beside PMN, endothelial cells, monocytes, platelets, and erythrocytes are known to release microparticles by ectocytosis in vitro and in vivo [2], [3], [4], [5], [6]. For clarity, we will refer to “ectosomes” when describing such microparticles. A number of different functions have been attributed to ectosomes. For example, they allow the rapid shedding of stored interleukin-1β, released by activated THP-1 cells [7]. Ectosomes serve as intercellular protein carriers for tissue factor and the chemokine receptor CCR5 [8], [9]. As far as platelet ectosomes are concerned, their most prominent feature is a pronounced procoagulant activity. Interestingly, cell-derived adhesion molecules exposed by ectosomes of various origins are known to mediate several functions of ectosomes, e.g., hematopoietic recovery after transplantation, thrombus formation, and platelet–vessel wall interactions [10], [11], [12], [13]. The functions of ectosomes described until now are largely favoring inflammation and coagulation. In contrast, we recently described PMN ectosomes to be so far the only ectosomes having anti-inflammatory properties. Indeed, we found that PMN ectosomes downregulate the pro-inflammatory response of macrophages to stimuli such as zymosan and LPS [14].

Microparticles of diverse cellular origin are present in blood of normal individuals [15], [16], [17]. Although most such microparticles correspond probably to ectosomes originating from different cells, it is possible that some are apoptotic bodies or exosomes [16], [17], [18]. Thus we will refer here to “microparticles” when their origin has not been defined. Quantitative and qualitative changes, i.e., increased numbers of microparticles derived from different origins, were described in many diseases including sepsis [19], severe trauma [20], paroxysmal nocturnal hemoglobinuria [21], diabetes [22], [23], acute coronary syndromes [24], and lupus [2]. However, even in sepsis the number of PMN-derived microparticles is surprisingly low, when compared to the high numbers of ectosomes released by activated PMN in vitro.

Here we present evidence that most ectosomes of PMN might not circulate freely, as assumed until now. Indeed, ectosomes released by human PMN activate the classical pathway of complement and fix C4 and C3 fragments. These opsonized ectosomes bind in turn to erythrocytes via the complement receptor 1 (CD35/CR1).

Section snippets

Antibodies and reagents

Anti-human-C3d and -C4d IgG1 antibodies (Abs) were from Quidel (Santa Clara, CA), IgG1 isotype control Abs were from Diaclone Research (Besançon, France), and human C1q was from Calbiochen (San Diego, CA). Phycoerythrin (PE)-coupled anti-glycophorin A and goat anti-mouse-Ig Abs were from Pharmingen (San Diego, CA) and Southern Biotechnology Associates Inc. (Birmingham, AL), respectively. Lepirudin (Refludan®) was from Hoechst (Zürich, Switzerland). The anti-CD66b Ab was from Immunotech

Homogeneity of ectosome preparations

The ectosomes derived from PMN used in the presented work were homogeneous [26]. The purity of our preparations was attested by electron-microscopic (EM) (Fig. 1A) as well as FACScan analysis (Fig. 1B). Ectosomes had the previously described size of 50–200 nm by EM (19), and all particles were CD66b positive, i.e., derived from PMN.

Activation and binding of complement on the surface of ectosomes

Ectosomes bind C1q, the first component of the complement system [26]. We investigated whether this binding was followed by complement activation and deposition on

Discussion

We have previously shown in vitro, that in absence of serum, ectosomes bind specifically to endothelial cells as well as to monocytic THP-1 cells, but not to erythrocytes [26]. This complement-independent binding activity is likely to be mediated by adhesion molecules like selectins and integrins present on the surface of PMN ectosomes or by membrane components of ectosomes such as phosphatidylserine (PS), whose receptor is expressed on monocytes and macrophages [26], [32]. We further showed

Acknowledgments

We would like to thank Christoph Hess for his very valuable contribution regarding this work and acknowledge the help of Dr. J. Jensenius from the University of Aarhus, Denmark, who measured the activity of MBL/MASP-2 pathway. This work was supported by the Swiss National Foundation (SNF; grant 3200-066708), a grant from ZLF Bioplasma AG (Bern, Switzerland), the Stiftung für Medizinische und Biologische Forschung, and a personal grant to O. Gasser of the Fundazione per la ricerca sulla

References (42)

  • L.B. Klickstein et al.

    Complement receptor type 1 (CR1, CD35) is a receptor for C1q

    Immunity

    (1997)
  • J.A. Schifferli et al.

    Physiological and pathological aspects of circulating immune complexes

    Kidney Int.

    (1989)
  • E. Biro et al.

    Human cell-derived microparticles promote thrombus formation in vivo in a tissue factor-dependent manner Accumulation of tissue factor into developing thrombi in vivo is dependent upon microparticle P-selectin glycoprotein ligand 1 and platelet P-selectin

    J. Thromb. Haemost.

    (2003)
  • R.F. Zwaal et al.

    Pathophysiologic implications of membrane phospholipid asymmetry in blood cells

    Blood

    (1997)
  • J.M. Freyssinet

    Cellular microparticles: what are they bad or good for?

    J. Thromb. Haemost.

    (2003)
  • J.M. Stein et al.

    Ectocytosis caused by sublytic autologous complement attack on human neutrophils. The sorting of endogenous plasma-membrane proteins and lipids into shed vesicles

    Biochem. J.

    (1991)
  • V. Combes et al.

    In vitro generation of endothelial microparticles and possible prothrombotic activity in patients with lupus anticoagulant

    J. Clin. Invest.

    (1999)
  • K. Iida et al.

    Membrane vesiculation protects erythrocytes from destruction by complement

    J. Immunol.

    (1991)
  • M. Pascual et al.

    Identification of membrane-bound CR1 (CD35) in human urine: evidence for its release by glomerular podocytes

    J. Exp. Med.

    (1994)
  • N. Satta et al.

    Monocyte vesiculation is a possible mechanism for dissemination of membrane-associated procoagulant activities and adhesion molecules after stimulation by lipopolysaccharide

    J. Immunol.

    (1994)
  • M. Mack et al.

    Transfer of the chemokine receptor CCR5 between cells by membrane-derived microparticles: a mechanism for cellular human immunodeficiency virus 1 infection

    Nat. Med.

    (2000)
  • View full text