Microparticulas Ards

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Am J Physiol Lung Cell Mol Physiol 303: L364–L381, 2012.

Review First published June 22, 2012; doi:10.1152/ajplung.00354.2011.

Microparticles and acute lung injury


Mark McVey,1,2 Arata Tabuchi,1 and Wolfgang M. Kuebler1,3,4,5,6
1
The Keenan Research Centre at the Li Ka Shing Knowledge Institute of St. Michael’s, University of Toronto, Toronto,
Ontario, Canada; 2Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada; 3Department of Surgery,
University of Toronto, Toronto, Ontario, Canada; 4Department of Physiology, University of Toronto, Toronto, Ontario,
Canada; 5Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany; and 6German Heart Institute, Berlin,
Germany
Submitted 4 November 2011; accepted in final form 18 June 2012

McVey M, Tabuchi A, Kuebler WM. Microparticles and acute lung injury. Am


J Physiol Lung Cell Mol Physiol 303: L364 –L381, 2012. First published June 22,
2012; doi:10.1152/ajplung.00354.2011.—The pathophysiology of acute lung injury
(ALI) and its most severe form, acute respiratory distress syndrome (ARDS), is
characterized by increased vascular and epithelial permeability, hypercoagulation
and hypofibrinolysis, inflammation, and immune modulation. These detrimental
changes are orchestrated by cross talk between a complex network of cells,
mediators, and signaling pathways. A rapidly growing number of studies have
reported the appearance of distinct populations of microparticles (MPs) in both the
vascular and alveolar compartments in animal models of ALI/ARDS or respective
patient populations, where they may serve as diagnostic and prognostic biomarkers.
MPs are small cytosolic vesicles with an intact lipid bilayer that can be released by
a variety of vascular, parenchymal, or blood cells and that contain membrane and
cytosolic proteins, organelles, lipids, and RNA supplied from and characteristic for
their respective parental cells. Owing to this endowment, MPs can effectively
interact with other cell types via fusion, receptor-mediated interaction, uptake, or
mediator release, thereby acting as intrinsic stimulators, modulators, or even
attenuators in a variety of disease processes. This review summarizes current
knowledge on the formation and potential functional role of different MPs in
inflammatory diseases with a specific focus on ALI/ARDS. ALI has been associ-
ated with the formation of MPs from such diverse cellular origins as platelets,
neutrophils, monocytes, lymphocytes, red blood cells, and endothelial and epithe-
lial cells. Because of their considerable heterogeneity in terms of origin and
functional properties, MPs may contribute via both harmful and beneficial effects
to the characteristic pathological features of ALI/ARDS. A better understanding of
the formation, function, and relevance of MPs may give rise to new promising
therapeutic strategies to modulate coagulation, inflammation, endothelial function,
and permeability either through removal or inhibition of “detrimental” MPs or
through administration or stimulation of “favorable” MPs.
acute respiratory distress syndrome; permeability; coagulation; inflammation; en-
dothelial function

ACUTE LUNG INJURY (ALI) as a result from either direct or ceeds to a fibrotic phase in which the lung is burdened with
indirect mechanical, toxic, infectious, or inflammatory chal- scarring as it remodels while attempting to heal. Despite the
lenges to the lung presents initially as dyspnea and hypoxemia, well-documented benefit of protective ventilatory strategies and
which can rapidly progress to respiratory failure. ALI and its increasing knowledge of the etiological mechanisms of ARDS,
most severe form, the acute respiratory distress syndrome mortality rates remain relatively stagnant around 40% (133), with
(ARDS), are characterized by bilateral exudative chest infiltrates incidence estimates of up to 75 per 100,000 (171).
visible by roentgenograms with no evidence of left heart failure A broad spectrum of different cell types contribute to the
and a PaO2/FIO2 ratio ⬍300 (ALI) or even ⬍200 (ARDS) (151, pathogenesis of ARDS, including alveolar epithelial and vas-
171). Within days, ALI progresses from an initial inflammatory cular endothelial cells that undergo changes in permeability
exudative phase with a leaky edematous lung to a proliferative leading to edema formation and alveolocapillary injuries, as
phase involving fibrin deposition and a concomitant decrease in well as platelets and immune cells such as polymorphonuclear
respiratory compliance that finally, if the patient survives, pro- neutrophils (PMNs), alveolar macrophages, and monocytes
that are critically involved in inflammatory responses (104,
108, 170, 171). In contrast to the characteristic anticoagulant
Address for reprint requests and other correspondence: W. M. Kuebler, The
Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s
and profibrinolytic basal state of the lung, ALI is associated
Hospital, 209 Victoria St., M5B 1W8 Toronto, Ontario, Canada (e-mail: with a prothrombotic and antifibrinolytic shift that favors
kueblerw@smh.ca). deposition of fibrin and accelerates and potentiates inflamma-
L364 1040-0605/12 Copyright © 2012 the American Physiological Society http://www.ajplung.org
Review
MICROPARTICLES AND ACUTE LUNG INJURY L365
tion (19). Considerable research has focused on the direct layer has a characteristically high level of negatively charged
interaction and communication between different cell types phospholipids, particularly phosphatidylserine (PS). MPs may
both in vivo and in vitro and has revealed important patho- contain membrane and cytosolic proteins, transcription factors,
physiological aspects of ALI. However, a rapidly growing genetic material such as ribosomal RNA (rRNA), messenger
number of studies have recently started to recognize the critical RNA (mRNA), and microRNAs (miRNA) as well as lipids or
role of microparticles (MPs) as biomarkers, communication even organelles supplied from parent cells (22, 32, 109, 116).
shuttles between these different cells and cell types, and Occasionally, membrane vesicles of less than 0.1 ␮m are
independent functional effectors in a large variety of diseases subcategorized as nanoparticles and have been proposed to
ranging from endemic pathologies such as hypertension, dia- originate from lipid rafts on parent cell membranes; accord-
betes, and atherosclerosis to frequently lethal syndromes such ingly, they may display distinct protein endowments and func-
as ALI (7, 16, 27, 28, 31, 36, 44, 60, 65, 79, 99, 118, 129, 135, tions (reviewed in Refs. 77, 134, 155).
138, 155, 172). Here we provide a brief overview on the main Microparticles subserve intercellular information exchange.
characteristics of MPs, review the current knowledge on their The distinct cellular elements retained from precursor cells
role in inflammatory disease with a particular focus on ALI, allow MPs to function as transcellular delivery systems for the
and oppose their proposed detrimental and beneficial effects in exchange of biological signals. Notably, information exchange
lung injury, thus providing a rationale for their future use as between MPs and target cells can occur in a bidirectional
both therapeutic targets and tools. manner, thus generating a continuous and dynamic exchange
of antigens, cell signaling molecules, and genetic material that
Microparticles
opens an entire new spectrum of opportunities for tightly
The first accounts of MPs date back to 1967 at which time regulated (or dysregulated) signal propagation.
they were considered to be nothing more than cellular dust In principle, MPs can transfer information from the MP-
(177). In fact, MPs are tiny cell-derived, intact vesicles that are generating donor cell to a wide range of target cells either by
formed by partitioning off from activated or apoptotic eukary- direct cell-cell contact for delivery of genetic material, pro-
otic cells (Fig. 1). MPs are defined by their size, which ranges teins, and lipids or alternatively by remote interaction through
between 50 nm and 1 ␮m, and their distinctive lipid layer secretion of soluble mediators and effectors by MPs. Currently,
composition. MPs have intact lipid bilayers, yet their outer we distinguish at least four distinct mechanisms how MPs may

Fig. 1. Release of microparticles (MPs) from


the surface of a polymorphonuclear neutro-
phil (PMN). Scanning electron micrographs
show a fixed resting PMN (scale bar: 1.5
␮m) (A) and a fixed PMN stimulated with
0.1 ␮M N-formylmethionine leucyl-phenyl-
alanine for 5 min (scale bar: 1.5 ␮m) dis-
playing large pseudopodia and, in addition,
budding of small vesicles of 70 –300 nm in
size from the PMN cell membrane (B; inset,
arrowhead; scale bar 400 nm). C: transmis-
sion electron micrographs following immu-
nogold labeling show expression of cell sur-
face antigens such as complement receptor 1
on isolated MPs (scale bar: 150 nm). D: thin
sections of MPs precipitated with Dyna-
beads prove vesicles to be bilamellar struc-
tures. Reproduced from Hess et al. (69);
Copyright 1999. The American Association
of Immunologists, Inc.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
L366 MICROPARTICLES AND ACUTE LUNG INJURY

interact with their target cells that are illustrated schematically such as for example angiogenesis in endothelial cells (64).
in Fig. 2. First, MPs are able to stimulate outside-in signaling Because miRNAs have recently been recognized to contribute
in target cells via presentation of membrane-associated ligands to injurious mechanisms in ALI (183), transfer of miRNA from
that directly bind to their respective surface receptors, or via MPs to inflammatory or lung parenchymal cells may present an
release of secreted factors such as for example cytokines that exciting novel pathomechanism in this disease that clearly
act on the target cell via receptor-mediated or receptor-inde- deserves further exploration.
pendent mechanisms (102). Second, MPs can be internalized Origin of microparticles. Parent cells of MPs include but are
into recipient cells, a process that has also been proposed to not limited to platelets, megakaryocytes, monocytes, epithelial
underlie the rapid clearance of circulating MPs from the blood cells, lymphocytes, PMNs, endothelial cells, red blood cells,
(41). For example, fluorescently tagged platelet MPs (PMPs) reticulocytes, mast cells, stem cells, astrocytes, glial cells,
can be endocytosed by brain microvascular endothelial cells smooth muscle cells, and cancer cells (16, 20, 23, 139). Similar
and are subsequently detectable in endosomes and lysosomes to the wide range of parent cells, MP formation can be
(50). Third, MPs may fully or partially fuse with the target cell, triggered by diverse physiological, pathophysiological, or ex-
allowing for complete or selective transfer of contents includ- perimental stimuli as discussed in greater detail below.
ing membrane and cytosolic proteins, bioactive lipids (109, Unstimulated, resting cells actively maintain their mem-
139), or even whole cell organelles, as recently demonstrated brane asymmetry via phospholipid transporter enzymes, which
for the microvesicular delivery of mitochondria by mesenchy- localize negatively charged phospholipids on the inner leaflet,
mal stem cells (73). Last, MPs may deliver genetic material in facing into the cell. When cellular homeostasis is disturbed or
form of mRNA and miRNA. The encoding mRNAs can be lost such as in apoptosis, the activities of these phospholipid
directly translated to alter the target cell proteome (6, 45), flippases, floppases, and scramblases are dysregulated, leading
whereas miRNAs are small noncoding RNAs that can modu- to hastened changes in membrane composition of the inner and
late gene expression in target cells at the posttranscriptional outer membrane leaflets. This process is a characteristic feature
level, in that they bind to complementary sequences on target of MP formation, since it allows for the relocation of the
mRNAs, commonly resulting in translational repression and PS-rich layer of the inner leaflet to the exterior leaflet (116,
gene silencing. The export of miRNA from donor cells is 155). The resulting changes in membrane composition and
largely regulated by ceramide (166), a notion that gains rele- regular asymmetry in conjunction with processes such as
vance in light of the parallel implication of ceramide in MP intracellular calcium accumulation and calpain activation re-
generation (155), which may thus facilitate miRNA egress with viewed in detail elsewhere (116, 119) cause membrane bleb-
MPs in a coordinated fashion. At the level of the target cell, bing and ultimately the formation of MPs (Fig. 3).
subsequent miRNA entry is facilitated either by endocytosis of Externalization of PS is a precursor step in MP formation,
MPs or by fusion of their lipid bilayer with the plasma allowing for a common shared surface marker for identification
membrane (166). Lately, miRNAs have been detected in a of many MPs that can be assayed by annexin V binding.
wide variety of different MPs as generated from peripheral Interestingly, however, it appears that there are MPs that do not
blood cells in humans (70) or mesenchymal (39) or embryonic stain appreciably for annexin V, which adds controversy to the
stem cells (180), respectively, and miRNA transfer by MPs has identification and strict definition of what constitutes an MP
been demonstrated to critically modulate target cell responses (116, 153). Furthermore, it seems that some MPs can also be

Fig. 2. Mechanisms of interactions between


MPs and target cells. Interaction between MPs
and their target cells can occur via essentially
4 different mechanisms: outside-in signaling
via ligand-receptor interaction or secreted fac-
tors (top right); internalization and lysosomal
processing of MPs facilitates for example sub-
sequent presentation of MP antigens on the
surface of target cells (bottom right); fusion-
mediated transfer of surface receptors, pro-
teins, and lipids (bottom left); and delivery of
genetic material such as mRNA or miRNA
(top left). MPs may fuse either completely or
temporarily with target cells resulting in either
complete or selective transfer of MP contents.
Redrawn and modified from a figure origi-
nally published in Beyer et al. (22).

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
MICROPARTICLES AND ACUTE LUNG INJURY L367
in their production. Different mechanisms of fusion between
cells and MPs that could lead to phenotypic modulation of MPs
and/or other cells have been recently reviewed in detail by
Quesenberry and colleagues (139). A particularly striking ex-
ample in this respect is the provocative hypothesis that endo-
thelial progenitor cells may origin from the fusion of PMPs
with mononuclear cells as suggested by proteomic analyses
(137). Repetitive fusion events with tissue factor (TF)-express-
ing cells may also account for the expression of TF on various
types of cells and MPs that would not otherwise express this
marker. For example, platelets can become TF positive after
contact with TF-expressing MPs shed from PMNs and mono-
cytes (145). Following stimulation with endotoxin [lipopoly-
saccharide (LPS)] or protein kinase C activation by phorbol
Fig. 3. Pathways of MP release from parent cells. Cell activation and/or
stimulation of proapoptotic signaling cascades cause a rapid loss in lipid
myristate acetate (PMA), platelets, and subsequent PMPs may
membrane asymmetry due to dysregulation of flippases, floppases, and scram- obtain their TF antigens from fusion with either monocytes or
blases via sudden increases in intracellular calcium, resulting in externalization monocyte microparticles (MMPs) (28). Beyond mixing of
of phosphatidylserine (PS). Concurrent cytoskeletal reorganization and con- distinct lineage markers between cells and MPs there are also
traction cause blebbing and subsequent shedding of MPs into the extracellular challenging distinctions between MPs of similar parent cell
space. MP formation from membrane lipid rafts may lead to release of MPs
that maintain part of the unique signaling characteristics of these platforms. origins. Subtle variations in parent cells can be difficult to
Redrawn and modified from a figure originally published in Chironi et al. (36). detect because of phenotypic similarities of their respective
MPs, as in the case of platelet vs. megakaryocyte-derived MPs
(54, 143). Hence, a series of elaborate experimental techniques
formed before regular membrane asymmetry is disrupted, such have been developed to allow for the exact identification of the
as in cultured endothelial cells (155). Specifically, Davizon and origin of MPs and the functional significance of these cell-
colleagues (42) have linked MP formation to lipid rafts in cell derived vesicles.
membranes, raising the possibility that lipid rafts may be Characterization of microparticles. In the stepwise process
actively involved in MP formation under some conditions, and of experimentally isolating MPs there are considerable techni-
that MPs may maintain part of the unique functional and cal variables that can have impact on the recovery and pheno-
signaling properties of membrane lipid rafts (Fig. 3). Currently, type of MPs obtained. Some of these procedural differences
the cellular machinery implicated in MP formation constitutes include the actual phlebotomy, the use of anticoagulants, stor-
an active area of research (reviewed in Ref. 134) that is age conditions, length and number of freeze-thaw cycles,
expected to yield important insights not only into the mecha- centrifugation, washing, and pelleting or filtration of MPs; and
nisms of MP formation but likewise into its regulation and specific recommendations (Table 1) have been developed to
function. optimize each of these steps (186). A series of review articles
In healthy humans, circulating MPs are largely derived from have scrutinized the impact of these variables that underline the
platelets with some contributions from leukocytes and mini- sensitive and precarious nature of MP isolation and stress how
mally from endothelial cells (34, 36). Size and composition of caution is required to avoid undesired differences in MP
this population of basal MPs depend on physiological variables isolates obtained from slight variations in handling and pro-
including exercise, menstrual cycle, age, or exposure to ex- cessing procedures (91, 134, 153, 155). The rapidly growing
treme environments such as SCUBA diving, in addition to number of publications pertaining to MPs showcases a diverse
countless pathological conditions, some of which are discussed array of techniques for preparation and isolation of MPs that is
in later sections of this review (34, 55, 163). bound to generate considerable variability in terms of the final
PMPs have a short circulating half-life of less than 10 min in products assayed (78, 134). In recent years, this dilemma has
rabbits (141) but were shown to circulate for ⬃30 min in mice stimulated workshops and meetings specifically dedicated to
(53). MP clearance from the circulation occurs predominantly the attempt to limit variability and standardize the study of
by endothelial cell internalization (41) or splenic removal, and MPs; namely, the International Society on Thrombosis and
the half-life of PMPs is markedly prolonged in splenectomized Haemostasis has made strides to centralize methodological
mice compared with controls (129). Moreover, a recent study approaches for MP isolation (90).
in thrombocytopenic humans reported a markedly longer half- Detection and characterization of MPs has involved strate-
life for circulating MPs that was on the order of 5– 6 h (142, gies such as antibody-capture ELISA assays, flow cytometry,
143). The considerable variation in half-lives in these studies functional coagulation assays, electron microscopy (Fig. 1),
suggests a major influence of both host and MP variables such atomic force or confocal microscopy, HPLC, capillary electro-
as species, blood cell levels, and mode of activation, as well as phoresis, and mass spectrometry (11, 24, 37, 43, 75, 77–79, 89,
potential methodological variability with respect to isolation 91, 115, 129, 137, 153, 155, 162, 174, 181, 185). Each of these
and assay for MPs between these different studies. techniques comes with its specific advantages and limitations,
Tracking the origin of MPs is complicated not only by the as summarized in Table 2. Because of its general availability
wide variety of possible parent cell sources and triggering and versatility, flow cytometry has become the most commonly
stimuli, but in addition by the fact that these cell-derived used technique for characterization of MPs and discerning their
vesicles can be and presumably are constantly phenotypically parent cell of origin depending on the expression of character-
modified by contact with different cell types at different steps istic surface markers (Fig. 4). Most strategies with flow cy-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
L368 MICROPARTICLES AND ACUTE LUNG INJURY

Table 1. Technical variables and recommendations for MP isolation


Variables Current Recommendation

Venipuncture needle & tourniquet ⬎21-gauge needle, light or no tourniquet to avoid platelet activation
2
Blood collection anticoagulant citrate, CTAD (as EDTA can activate platelets)
2 transportation & time to centrifuge avoid agitation, ⬍2 h to centrifuge (as PMPs increase over time)
PFP preparation speed & time of centrifuge wide variety of speeds and times
platelets may persist in PFP after a single centrifugation step
2
(Storage) number of freeze & thaw cycles ⫺80°C, single freeze & thaw cycle (multiple cycles reduce number of MPs)
2
(Further purification of MPs) speed & time of centrifuge wide variety of speeds and times
2 further purification can reduce background signal from plasma at the risk of losing MPs
Analysis
Flow chart of variables and current recommendations for the isolation, storage, and purification of microparticles (MPs) as proposed for platelet microparticles
(PMPs) by Zwicker and colleagues (186). CTAD, citrate theophylline adenosine and diphyridamole; PFP, platelet-free plasma.

tometry involve identification of MPs based on forward and captured the full extent of variations possible since the char-
side scatter characteristics as well as annexin V staining for PS acterization of MPs under different conditions is incomplete
in conjunction with analysis of markers characteristic for and we currently lack the tools to accurately assay antigens
specific parent cells to test for their contribution to the indi- present at very low abundances on the surface of MPs. MPs
vidual MPs being assayed. The use of flow cytometry to this come in a variety of sizes ranging from smaller nanoparticles
end bears a series of strengths, but also some limitations (77, such as those often produced by red blood cells up to MPs large
78, 89, 147, 153) that have been discussed in detail in recent enough to rival platelets (134). Similar parent cells are able to
literature. Proteomics presents an important alternative tool for produce a spectrum of different-sized MPs depending upon
particularly small particles not amenable to flow cytometry whether MP formation occurs spontaneously at baseline or
(117) since it can yield comprehensive information on charac- following stimulation (25, 74). Even cell lines such as Jurkat T
teristic protein expression patterns that may be utilized to cells yield a spectrum of different-sized MPs (164). Notably,
identify and/or characterize MPs. This approach has been used this heterogeneity in size can directly impact MP surface
in a series of investigations to assay different aspects of the marker expression and function as shown for platelet MPs that
human MP proteome (75, 96). contain different antigens such as growth factors, chemokines,
Heterogeneity of microparticles. Our current understanding and receptors depending on their size (74).
of the conditions under which MPs are formed, and their Similar to their heterogeneity in terms of origin, size, and
individual actions, is still incomplete. Little is known about endowment with antigens, MPs also differ widely with regard
exact parameters that dictate when, how, from where, why, to their functional effects. MPs can shuttle information from
which, and how many MPs are formed. What is becoming remote cells to similar or phenotypically different cells that
evident is the vast variation in terms of genetic material and might never be in direct proximity. Different MPs can affect
antigens MPs can express from their parent cells of origin or certain cells in different ways, as for example diabetics’ T
other cells or MPs they contact during formation or during cell-derived MPs can decrease endothelial nitric oxide (NO)
circulation, which can be incorporated or eventually redistrib- synthase (eNOS) and increase caveolin-1 expression causing
uted to other MPs or cells. The literature has likely not endothelial dysfunction as opposed to MPs formed by in vitro

Table 2. Advantages and limitations of techniques commonly applied in MP analysis


Enumeration Sizing Origin Throughput Cost Availability

FCM (79,89,155,185) ⫹⫹⫹ ⫹ ⫹⫹⫹ ⫹⫹ ⫺ ⫹⫹ limited sensitivity for small sized particles
(⬍500 nm)
new technologies (e.g., impedance FCM)
are being proposed to address this
limitation
ELISA (78,153,155) ⫺ ⫺ ⫹ ⫹⫹⫹ ⫹ ⫹⫹⫹ high sensitivity
Functional assays (78,129,153,155) ⫺ ⫺ ⫹ ⫹⫹⫹ ⫹ ⫹⫹⫹ no direct enumeration but able to assay
functional parameters (e.g., coagulation)
EM (11,37) ⫹ ⫹⫹ ⫹⫹ ⫺ ⫺ ⫺ can visualize organelles
CLM (162) ⫹⫹ ⫹ ⫹⫹ ⫺ ⫺ ⫹⫹ can investigate cell-MP interactions (e.g.,
internalization)
AFM (181) ⫹⫹ ⫹⫹⫹ ⫹ ⫺ ⫺ ⫺ high sensitivity
HPLC (24,43,174) ⫺ ⫹* ⫹ ⫺ ⫺ ⫹ can analyze phospholipid composition
MS (43,75,115,137) ⫺ ⫺ ⫹⫹ ⫺ ⫺ ⫺ can perform proteome and lipidome
analyses
Commonly applied techniques for the analysis of MPs and their relative strengths and weaknesses in terms of MP enumeration, MP sizing, determining the
parent cell of origin, throughput, cost, and availability. AFM, atomic force microscopy; CLM, confocal laser microscopy; ELISA, enzyme-linked immune sorbent
assay; EM, electron microscopy; FCM, flow cytometry; HPLC, high-performance liquid chromatography; MS, mass spectrometry. *Size fractionation.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
MICROPARTICLES AND ACUTE LUNG INJURY L369

Fig. 4. Characteristic surface markers indic-


ative of MP origin. Characteristic surface
markers used to detect MP populations
(right) that originate from specific parent
cells (left). CD, cluster of differentiation;
EMP, endothelial microparticle; IgG, immu-
noglobin; LMP, lymphocyte microparticle;
MegMP, megakaryocyte microparticle; MP,
microparticle; MMP, monocyte micropar-
ticle; NMP, neutrophil microparticle; PMP,
platelet microparticle; RMP, red blood cell
microparticle; vWF, von Willebrand factor.

activation of human T cells that express sonic hedgehog on in response to pathophysiological stimuli adds another level of
their membranes and can stimulate NO production as evi- complexity, since different triggers will stimulate MP forma-
denced by the improved endothelial function in a cardiac tion from different parent cell types. Multiple simultaneous or
ischemia-reperfusion injury model (106, 114). Alternatively, a sequential triggers can further enhance MP diversity and there-
single MP subtype such as PMPs can interact with and recruit fore potentially expand their range of function. For example,
numerous target cells such as monocytes, NK cells, and B and platelet storage conditions including platelet-rich plasma,
T cells (114). apheresis blood bank-derived human platelets, or cold storage
MP generation is triggered by a spectrum of stimuli ranging of platelets with and without contact with propyl gallate will
from physiological to experimental stimuli (Fig. 5). MPs can produce distinct PMP variants (33, 144, 168, 178). Addition of
be formed constitutively by various parent cells under physi- platelet activation inhibitors such as prostaglandin E1 (PGE1),
ological conditions but in differing amounts, resulting in a theophylline, and aprotinin during storage reduce the number
higher abundance of circulating PMPs compared with red of large but not small PMPs formed and modify their function
blood cell- or lymphocyte-derived MPs (RMP and LMP, re- in terms of platelet factor 3 activity (25). Under experimental
spectively) in healthy patients (116). Additional physiological conditions, a diverse array of potent cell stimulants such as
triggers for MP formation involve stresses such as heavy calcium ionophores, phytohemagglutinins, LPS, N-formylme-
exercise or cyclic changes such as menstruation. MP formation thionyl-leucyl-phenylalanine, and PMA can be utilized to in-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
L370 MICROPARTICLES AND ACUTE LUNG INJURY

Fig. 5. Triggers for MP formation from differ-


ent parent cells of origin. Physiological, patho-
physiological, and experimental triggers of MP
formation (right) from platelets, megakaryo-
cytes, lymphocytes, monocytes, red blood
cells, polymorphonuclear neutrophils, and en-
dothelial cells (left). APC, activated protein C;
ATII, angiotensin II; autoAb, autoantibodies;
C5a, complement factor C5a; C5B-9, comple-
ment factor C5B-9; CD40L, CD40 ligand;
CHX, cyclohexamide; EC, endothelial cell;
fMLP, N-formylmethionyl-leucyl-phenylala-
nine; IL-1, interleukin-1; L-NAME, L-NG-ni-
troarginine methyl ester; LPS, lipopolysaccha-
ride; PAF, platelet activating factor; PHA,
phytohemagglutinin; PAI-I, plasminogen acti-
vator inhibitor 1; PMA, phorbol myristate ac-
etate; PtdIns(4,5)P2, phosphatidylinositol-4,5-
bisphosphate; ROS, reactive oxygen species;
STS, staurosporine; TNF-␣, tumor necrosis
factor-␣; TRAP, thrombin receptor agonist
peptide SFLLRN.

duce MP formation with a high efficiency and reproducibility organ dysfunction syndrome, that are again associated with
(Fig. 5). elevated levels of MPs (129, 155). MPs have also been impli-
cated mechanistically on several levels in the pathophysiology
Microparticles in Inflammatory Disease of sepsis, including proinflammatory signaling pathways such
Some of the MPs characterized to date have been identified as target cell activation through reactive oxygen species pro-
as biomarkers for a wide variety of inflammatory conditions. duction (ROS) and coagulopathy in terms of MP-mediated TF
Elevated levels of circulating PMP, LMP, MMP, and endothe- presentation (113).
lial MPs (EMPs) have been shown to accompany pathological A survey comparing 36 patients with sepsis with 18 healthier
conditions such as atherosclerosis, Type 2 diabetes, vasculitis, patients without sepsis showed elevated blood levels of total
pulmonary hypertension, coronary disease, cardiopulmonary MPs, specifically PMPs and EMPs, whereas CD45⫹ LMPs
resuscitation, lupus, Crohn’s disease, and rheumatoid arthritis were lower in patients with sepsis compared with controls
(7, 27, 28, 36, 52, 118, 129, 135, 138, 155). Likewise, systemic (122). Interestingly, MPs from septic patients enhanced aortic
inflammatory states in pregnancy such as preeclampsia are contraction when transfused intravenously to mice. This effect
associated with elevated plasma levels of MPs (146). Infec- was independent of NO modulation or cyclooxygenase enzyme
tious diseases are either directly associated with increased expression but blocked by the specific thromboxane A2
levels of MPs as seen in malaria (61) or can give rise to (TXA2) antagonist SQ-29548, indicating that circulating MPs
disproportionate inflammatory responses such as the systemic may prevent vascular hyporeactivity accounting for systemic
inflammatory response syndrome (SIRS), sepsis, or the multi- hypotension in septic shock through a TXA2-mediated mech-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
MICROPARTICLES AND ACUTE LUNG INJURY L371
anism (122). Furthermore, patients with SIRS have elevated matory diseases, MPs may be a biomarker of the disease and
levels of circulating EMPs, increased binding of MPs to PMNs, play important roles by either aggravating or counteracting (or
higher procoagulant activity and higher circulating levels of potentially both) the ongoing disease process. For example,
triggers for MP formation such as plasminogen activator in- interactions between cells such as platelets, PMNs, and endo-
hibitor 1 (PAI-1) compared with healthy controls (126). As we thelial cells are central to the pathology of ALI/ARDS, and
discuss later in greater detail, the effects of MPs are frequently MPs have been demonstrated to critically regulate these inter-
considered to aggravate the pathology in inflammatory and actions and/or modify the phenotypes of the contributing cells
cardiovascular diseases, and increased formation of MPs as by fusion and subsequent transfer of cellular components. In
seen in systemic inflammatory disorders is therefore generally the following, we will summarize recent data in support of an
considered to constitute not only a biomarker, but an important association and possible functional role of MPs in ALI/ARDS
pathophysiological mechanism driving the disease. However, and discuss potential novel interactions between MPs and lung
inflammation and the associated increase in circulating MPs parenchyma or circulating blood cells that are likely pertinent
may also have protective effects, as pointed out by Soriano and to lung injury.
coworkers (158), who demonstrated that increased inflamma-
tion and the presence of EMPs, PMPs, and platelet leukocyte Potential Detrimental Effects of Microparticles in ALI
conjugates are significantly associated with survival in septic
patients. Hence, more detailed and comprehensive experimen- Excessive inflammation, changes in coagulation and fibrin
tal and clinical investigations are in dire need to discern which deposition, and increased permeability of the alveolocapillary
MPs under what conditions exert beneficial or detrimental barrier with resulting protein extravasation and lung edema
effects in inflammatory diseases. formation are characteristic hallmarks of ALI (Fig. 6). In the
Over the past decade, it has become evident that inflamma- following, we highlight how MPs may impact either directly or
tory diseases are closely associated with impaired coagulation indirectly on the pathophysiology underlying these symptoms.
and fibrinolysis, processes that can be regulated through MPs. Inflammation. Inflammatory changes in ALI/ARDS involve
Many but not all MPs express TF, which interacts with factor an abundance of proinflammatory intra- and intercellular sig-
VIIa facilitating activation of factors IX and X, thus leading to naling pathways that include, but are in no way confined to,
generation of thrombin (103). TF MPs are characteristically signaling via arachidonic acid (AA), TXA2, PAI-1, NO, ROS,
found in diseases in which thrombosis is common such as cytokines, and protease-activated receptors (PARs). In this
cancer, sepsis, unstable angina, ALI, sickle cell disease, and section, we highlight data demonstrating that MPs can modu-
thromboembolism (128, 129, 138, 165), giving rise to the late and/or replicate these pathways (114, 120) and thus act as
speculation that they may contribute critically to the disease proinflammatory agents promoting ALI/ARDS.
process. TF is a key player in the activation of the coagulation Over past years, the tight interplay between platelets and
pathway, yet its action is determined not only by its expression PMNs has emerged as an important aspect in the pathogenesis
levels, but also by its accessibility in terms of conformational of ALI (100, 160, 182). Similar to platelets, circulating PMPs
state that is controversial for different cells types or conditions
can interact with PMNs via linkages including glycoprotein
(128). Much of the circulating TF is in fact in an encrypted and,
Ib/Mac-1, ␤2-integrins (CD11b)/Mac-1, or P-selectin/PSGL-1
therefore, inactive state, whereas MPs represent a source of
on PMPs/PMNs, thus causing PMN activation involving se-
decrypted, active TF (127). LPS stimulates monocytes and
cretion of interleukin (IL)-8, oxidative burst, degranulation,
endothelial cells to produce TF that can lead to a coagulopathy
and enhanced leukocyte rolling (79, 99). Likewise, PMPs
that can escalate into disseminated intravascular coagulation
(103). Coincubation of platelets with monocytes and PMNs generated from LPS-stimulated platelets promote endothelial
that produce high amounts of TF following stimulation with activation as exemplified by the finding that they further
proinflammatory agents such as LPS leads to enhanced pre- increase the IL-1␤-induced expression of vascular cell adhe-
sentation of decrypted TF in the form of MPs, thus generating sion molecule-1 (30). Hence, circulating PMPs are not only a
a prothrombotic milieu (127). Even MPs not expressing TF can biomarker, but also, more importantly, an active promoter of
still possess considerable procoagulant properties because they inflammatory disease processes in that they stimulate endothe-
contain anionic phospholipids, including PS, which acts as a lial cells, PMNs, and notably also platelets. The latter was
surface to facilitate assembly of the clotting factor machinery demonstrated in experiments in which PMPs were stimulated
(103, 129, 138). Hence, MPs can be involved in coagulopathies by soluble phospholipase A2; the newly formed AA is then
associated with inflammatory diseases such as sepsis since they rapidly converted by PMPs to TXA2, a classic activator of
possess prothrombotic characteristics with exposed TF and PS platelets (14). In line with this view, ExoU, a Pseudomonas
but conversely may also activate fibrinolysis via changes in aeruginosa cytotoxin with phospholipase A2 activity, increases
plasminogen and metalloproteases (113) or exert anticoagulant both PMP release and TXA2 formation and accordingly re-
properties mediated through protein C and tissue factor path- sulted in lung thrombus formation and increased vascular
way inhibitor (TFPI) (117). In line with the notion that MPs permeability in a murine model of pneumosepsis (101). Over-
may have both pro- and anticoagulant effects, thrombus weight all, a series of proinflammatory effects of PMPs has emerged
was shown to correlate with plasma levels of PMPs and older that is based on the activation of platelets, monocytes, and
MPs in a murine thrombus model, whereas LMPs showed an vascular endothelial cells either directly by MP-derived AA or
inverse correlation (140). by its metabolism to bioactive prostanoids (12, 13, 15). This
Microparticles and acute lung injury. Several studies have recognition substantiates a critical role of PMPs in ALI, since
shown associations of MPs with key structures and cells in TXA2 contributes pivotally to lung vascular leakage and in-
ALI, giving rise to the notion that, similar to systemic inflam- flammation in experimental models of the disease (72).

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
L372 MICROPARTICLES AND ACUTE LUNG INJURY

Fig. 6. Schematic representation of the proposed role of MPs in acute lung injury. I: depiction of normal healthy lung, interstitium and vasculature with associated
basal MP populations (color coded). Acute lung injury (II) involves characteristic pathologies including increased permeability (IIa), coagulation leading to
thrombosis and fibrin deposition (IIb), and inflammation and immunomodulation (IIc). Each of these pathologies is affected in distinct ways by specific MP
populations that are in turn activated by specific stimuli. AA, arachidonic acid; APC, activated protein C; EMP, endothelial MP (blue), EpMP, epithelial MP (light
purple); IL-1␤, interleukin 1␤; INF␥, interferon-␥; LMP, lymphocyte MP (red); LPS, lipopolysaccharide; LXA4, lipoxin A4; MacMP, macrophage MP (pink);
MMP, monocyte MP (purple); MP, microparticle; NMP, polymorphonuclear neutrophil MP (orange); PAI-1, plasminogen activator inhibitor 1; PHA,
phytohemagglutinin; PMN, polymorphonuclear neutrophil; PMP, platelet MP (light green); RMP, red blood cell MP (dark green); ROS, reactive oxygen species;
TFMP, tissue factor expressing microparticle (dark purple); TNF-␣, tumor necrosis factor-␣; TXA2, thromboxane A2.

Platelet-PMN and endothelial-PMN costimulation may not Lastly, inflammatory diseases such as ALI and ARDS are
only be promoted by PMPs, but likewise by PMN micropar- often associated with immune modulation and its complex
ticles (NMPs), since endotoxin (LPS) can stimulate adherent functional consequences including inflammatory damage to
PMNs to release NMPs that generate platelet-activating factor bystander cells (123, 158, 173). Such immune modulation also
(PAF), thus causing activation of platelets (172), endothelium involves platelets that can actively alter innate and adaptive
(149), and other inflammatory cells. In the air spaces, NMPs immune function (94, 152). For example, PMPs can stimulate
may also trigger inflammatory responses and epithelial cell endothelial cells to generate a range of proinflammatory cyto-
damage, as suggested from their abundance in sputum from kines including IL-1, IL-6, IL-8, and monocyte chemoattrac-
patients with cystic fibrosis (136). NMPs can furthermore tant protein-1 (MCP-1) as well as monocyte-derived IL-1,
directly activate endothelial cells to generate proinflammatory TNF-␣, and IL-8 (reviewed in Refs. 20, 120). MPs have also
cytokines such as IL-6, thereby further promoting vascular been directly associated with immunomodulatory effector mol-
inflammation (8, 111, 112). Endothelial cells may in turn ecules such as RMP- and PMP-derived CD40L, IgG, and
likewise release MPs (EMPs) that modulate inflammation and complement found in stored blood products (80). Immuno-
coagulation, and most prominently vascular barrier function, modulatory and proinflammatory effects of MPs may be par-
for which reason they are discussed in greater detail in the ticularly relevant in the context of transfusion-related ALI
section Permeability. (TRALI), because storage of blood products will propel for-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
MICROPARTICLES AND ACUTE LUNG INJURY L373
mation of MPs (25, 33, 144) that will subsequently activate tients who died had a trend toward a higher MP concentration
PMNs (80). Taken together, MPs of diverse cellular origins in lung edema fluid compared with those who survived (P ⫽
including LMPs, PMPs, RMPs, NMPs, and EMPs directly or 0.073), attesting to a detrimental effect of MPs in ALI/ARDS.
indirectly promote inflammatory responses and may thus pro- Similar MPs could be generated in vitro by stimulation of
pel ALI, in that they stimulate cytokine release and promote cultured alveolar epithelial cells with a mix of the proinflam-
PMN activation, migration, and PMN-platelet interaction. matory mediators TNF-␣, IL-1␤, and interferon-␥ (16, 17).
Coagulation. Severe infections lead to activation of the Whereas both ARDS and non-ARDS controls in Bastarache’s
coagulation cascade and altered fibrinolysis, with the lung study were severely sick, BALF analyses from healthy pigs
being a key recipient of fibrin and clots during ALI and sepsis with volume-controlled ventilation set to maintain normocap-
(165). The hypercoagulable and hypofibrinolytic changes seen nia (37.5– 41 mmHg PaCO2) with 5 cmH2O positive end-
in ALI/ARDS contribute pivotally to the pathology of the expiratory pressure revealed PMPs that were positive for fi-
disease (170, 171) and involve characteristic changes in medi- brinogen, vWF, and P-selectin (124). This finding gains rele-
ators of coagulation and fibrinolysis, including TF, thrombo- vance in light of the fact that mechanical ventilation, in
modulin, TFPI, PAI-1, PS, and von Willebrand factor (vWF). particular at tidal volumes ⬎6 ml/kg body wt, will exacerbate
TF is considered to play an important role in the pathophys- ALI and promote ventilator-induced lung injury. PMPs in
iology of lung injury, because it can activate PAR1 and PAR2, BALF were evident after only 1 h of mechanical ventilation,
which are involved in inflammatory and procoagulatory re- yet, unfortunately, actual tidal volumes applied were not re-
sponses causing fibrin deposition in the lung (82, 165). Impor- ported in this study so that it is unclear whether this effect was
tantly, TF is expressed not only on activated mononuclear attributable to overventilation or occurs even under conditions
cells, endothelial cells, alveolar epithelium, or respiratory mac- of protective ventilation. Notably, presence of PMPs was
rophages, but likewise on many MPs (11, 165). The procoagu- likewise detected in human tracheal mucus obtained from five
lant activity of PMP membranes exceeds that of their parent humans at extubation after surgery, suggesting that this finding
platelet membranes by up to 100-fold (116, 156). In vascular may reflect a more general effect of mechanical ventilation
homeostasis, the effects of TF are counteracted by TFPI, which and/or surgical trauma.
blocks the TF-induced stimulation of the coagulation cascade Similar to TF expressing MPs, a procoagulant environment
by reversibly inhibiting factor Xa and thrombin. Severe infec- in ALI may be likewise promoted by PMPs. However, the
tions such as sepsis that constitute typical predispositions for degree to which circulating PMPs are actually elevated in
ALI are characterized by an imbalance between elevated TF patients with ALI or ARDS remains to be elucidated. Earlier
and lowered TFPI levels causing enhanced coagulation and work by George and coworkers (60) did not detect an increase
inflammation (165). The relevance of this imbalance is high- in blood PMPs in patients with ARDS from various causes,
lighted by observations from humans (56) and experimental whereas postcardiac bypass patients exhibited an increased
models of ALI in which blockade of TF by antibodies (175) or plasma concentration of PMPs. Yet it is conceivable that the
TFPI (48) reduced lung damage and improved pulmonary radiolabeled GPIIb antibody technique used to assay PMPs in
function. MMPs can attenuate the expression of TFPI and this study did not screen for the entire population of PMPs in
thrombomodulin, an endothelium-derived anticoagulant factor, the ARDS cohort. Beyond TF expression, MPs expose large
thus increasing thrombogenicity in vitro (4), whereas TF is amounts of PS on their outer membranes, thus creating an ideal
abundantly expressed on MPs under inflammatory conditions surface for activation of the coagulation pathway (148). In
(91). In healthy human volunteers, MP-borne TF procoagulant addition, MPs generate and release other prothrombotic factors
activity increased eightfold within 4 h after infusion of LPS including thrombin (80) or vWF multimers that can promote
(11). Hence, infectious or inflammatory stressors may contrib- and stabilize platelet aggregation (20). In summary, there is
ute to and/or aggravate ALI in part by the formation and ample evidence from in vitro animal and human studies dem-
release of TF studded MPs that in the presence of an attenuated onstrating the formation of MPs with procoagulant activity
anticoagulatory response will generate the characteristic pro- under inflammatory conditions and in ALI in both the vascular
thrombotic milieu associated with ALI, especially in consider- and alveolar compartment, where they are likely to contribute
ation of the previously discussed fact that TF on MPs is often pivotally to the hypercoagulable and hypofibrinolytic state
decrypted and active. Notably, TF activity on MPs in ALI/ characteristic for ALI/ARDS.
ARDS is not necessarily confined to the vascular compartment Permeability. The increased permeability of capillary and
(16), but, in line with the clinical presentation of fibrin clots in alveolar barriers in ALI leads to the extravasation of high
the distal air spaces, may similarly apply to the alveolar molecular weight protein and inflammatory cells into the air
compartment, where increased TF production by alveolar ep- spaces of the lung, thus impairing respiratory mechanics,
ithelial cells has been documented (18, 165). In pulmonary attenuating gas exchange, causing increased shunting and ven-
edema fluid from mechanically ventilated (⬍7 ml/kg tidal tilation perfusion mismatch, and contributing to the consolida-
volume) patients with ARDS, Bastarache and colleagues (16) tion of lung tissue during the subsequent fibroproliferative
detected higher concentrations of epithelial cell marker recep- phase of ALI/ARDS. Several biolipid mediators such as TXA2,
tor for advanced glycation end products (RAGE) and TF- phospholipids, and sphingolipids have been implicated in the
enriched MPs in bronchoalveolar lavage fluid (BALF) com- pathogenesis of permeability-type lung edema and are notably
pared with patients with hydrostatic lung edema whose MPs also linked directly or indirectly to the formation and function
also expressed less TF and RAGE. It is possible that ARDS of MPs (62, 104).
leads to either larger MPs that express more RAGE receptors As discussed before, PMPs can promote the formation of
or perhaps a numeric increase in RAGE-positive MPs com- TXA2 from AA (14). This finding gains relevance in the
pared with ARDS-negative hydrostatic edema controls. Pa- context of permeability-type edema because TXA2 inhibitors

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
L374 MICROPARTICLES AND ACUTE LUNG INJURY

can reduce lung vascular permeability in animal models of ALI modulated by the bioavailability of NO, and once formed MPs
(72, 182). In line with this view, increased levels of circulating can attenuate local NO production, in particular with respect to
PMPs were associated with elevated TXA2 plasma concentra- endothelial cells (20), which in turn may promote ALI and
tions and lung vascular permeability in a mouse model of barrier permeability. Accordingly, EMPs derived from PAI-1-
bacterial pneumosepsis with histological evidence of thrombus stimulated endothelial cells can induce direct ALI in mice and
formation in the lungs (101). In sepsis, the increased MP- stimulate the release of IL-1␤ and TNF-␣, leading to increased
associated formation of TXA2 may serve to maintain a basal PMN recruitment to the lung (31). In Brown Norway rats,
vasopressor effect (120, 122), yet this beneficial effect does not intravenous infusion of EMPs at pathophysiologically relevant
translate to the low pressure system of the lung where in- concentrations induced characteristic signs of pulmonary
creased TXA2 formation will primarily promote lung edema. edema, lung PMN infiltration, and increased lung vascular
In addition to eicosanoids, phospho- and sphingolipid me- permeability (44). Increased lung capillary permeability was
diators such as PAF and ceramide have been proposed to likewise evident in C57BL/6 mice when EMPs were infused as
contribute critically to vascular hyperpermeability in ALI (62). either primary or secondary hit (44). EMPs infusion into
PAF can be expressed on the surface of NMPs and promote C57BL/6 mice likewise led to increased inflammatory cytokine
platelet aggregation and association with PMNs (172), whereas release and lung PMN recruitment (31) and exacerbated LPS-
ceramide has been implicated in MP formation due to its induced lung injury. Notably, the aggravation of LPS-induced
involvement in lipid rafts (155). Acid sphingomyelinase lung injury was particularly prominent when EMPs were in-
(ASM), which catalyzes the conversion of sphingomyelin to fused prior to rather than simultaneous with LPS (31), indicat-
ceramide, triggers the release of brain glial MPs, an effect that ing that EMPs can both prime the lung for subsequent injurious
is blocked by ASM inhibitors or in astrocytes obtained from stimuli or exacerbate lung damage in previously challenged
ASM-deficient (Smpd1⫺/⫺) mice (23). Although it remains to lung tissue.
be shown whether a similar ASM-mediated mechanism con- Clinical implications. The recognition of a potential detri-
tributes to MP formation in other nonglial cell types, this mental effect of MPs in the pathophysiology of ALI/ARDS
finding raises the intriguing notion that the demonstrated det- opens up new therapeutic perspectives, in that removal of MPs
rimental effects of ASM/ceramide signaling in ALI (87) relate and/or inhibition of MP functions may present promising
to the increased formation of MPs. strategies for future treatment interventions (32, 184). Based on
Endothelial function. Apart from biolipid mediators, various a similar rationale, removal of PMPs by a membrane plasma
MPs may directly impact endothelial function, thereby criti- separator was recently tested in eight patients undergoing
cally regulating vascular barrier properties as well as promot- apheresis, yet although considerable amounts of PMPs were
ing the hyperinflammatory and hypercoagulatory responses removed by filtration total numbers of circulating PMPs could
discussed in previous sections. EMPs can attenuate NO release not be diminished by this intervention, potentially because of a
from cultured endothelial cells and induce a corresponding stimulation of PMP formation by the plasmapheresis procedure
decrease in eNOS phosphorylation at Ser1179 and a decrease per se (66). Compared with mechanical removal, pharmaco-
in hsp90 association consistent with the notion of EMP-in- logical attenuation of circulating MP levels may not only be
duced endothelial dysfunction (44). In line with this view, feasible but may have inadvertently already been introduced
EMP treatment impaired endothelium-mediated vasodilation in into the treatment of inflammatory diseases, since glucocorti-
both mouse and human ex vivo vessel preparations (44). coid treatment has been shown to significantly reduce circu-
Likewise, EMPs attenuate vasorelaxation in rat aortas in a lating PMP levels in patients with polymyositis and dermato-
dose-dependent fashion involving impaired NO signaling and myositis (154). Yet at the current stage the significance of
endothelial function (29). These findings gain relevance in the immunosuppression or immunomodulation by glucocorticoids
context of ALI in view of the frequently barrier-protective (85, in the treatment of ALI/ARDS remains unclear owing to mixed
179), anti-inflammatory, and anticoagulatory effects of basal clinical results (173), and a thorough analysis of the effects of
endothelial NO production in the lung. Endothelial dysfunction steroid administration on circulating MP levels and subpopu-
and impaired aortic vasorelaxation were likewise inducible in lations is yet lacking.
mice by LMPs, which caused overexpression of caveolin-1 and Notably, agonists of peroxisome proliferator-activated re-
concomitant inhibition of eNOS (106). Endothelial NO pro- ceptor (PPAR)-␥ such as rosiglitazone, which have been intro-
duction is also inhibited by smooth muscle cell MPs via a duced as antidiabetic drugs but also show efficiency in lung
␤3-integrin-mediated mechanism (49). The relevance of im- diseases such as pulmonary hypertension (67, 68), may present
paired endothelial NO formation in the context of ALI and an alternative approach to target deleterious MP functions in
inflammation is highlighted by the fact that IL-8-dependent inflammatory diseases. MMPs increase macrophage and mono-
PMN migration and endothelial transmigration in vitro are cyte superoxide anion production, cytokine release, and NF-␬B
enhanced by the NO synthase inhibitor NG-nitro-L-arginine activation via a PPAR-␥-regulated mechanism that can be
methyl ester (L-NAME), and likewise by NMPs generated by inhibited by PPAR-␥ agonists (5). PPAR-␥ agonists have also
L-NAME treatment, but not by D-NAME or untreated PMNs been demonstrated to inhibit LMP-mediated vascular dysfunc-
(125). Conversely, NO may also regulate MP formation al- tion and increased release of proinflammatory proteins from
though in different ways depending on cell type and/or exper- isolated murine aortae (120) and the upregulation of proinflam-
imental conditions, as highlighted by the finding that L-NAME matory cytokines including IL-8 and monocyte chemotactic
induced NMP formation from PMNs (125), whereas macro- protein-1 by monocyte MPs in cultured human lung epithelial
phage MP formation in response to Toll-like receptor agonists cells (83). PPAR-␥ expression has been shown to be reduced in
such as LPS is reduced by inhibition of inducible nitric oxide ALI (97) whereas agonists of PPAR-␣ or -␥ can attenuate
synthase (iNOS) (59). Taken together, MP formation can be experimental ALI (9, 98, 150). Although PPAR agonists may

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
MICROPARTICLES AND ACUTE LUNG INJURY L375
thus have therapeutic potential in the context of ALI/ARDS,
their effects on circulating MP levels and MP function in this
disease remain to be determined.
Several reviews of MPs have highlighted the interesting
strategy to modulate PMP formation by statin or polyunsatu-
rated fatty acid (PUFA) therapy (42, 91). Although PUFAs
have been pursued as therapeutic interventions in ARDS with
reported benefits in terms of decreased permeability and PMN
recruitment (157), statin treatment remains controversial yet is
actively pursued as potential therapy for ALI/ARDS (26, 86).
Because randomized clinical trials are currently underway, it
will be of interest to see whether potential benefits of statin
therapy in ARDS are likewise associated with reduced levels or
altered profiles of circulating MPs. Fig. 7. Beneficial and detrimental potential of MP as highlighted by their
Importantly, attenuation of MP formation may also provide effects on endothelial cells. MPs influence endothelial cells in different ways
(both beneficial and detrimental) depending on their cell origin and the specific
an attractive strategy to reduce complications associated with stimulating trigger leading to their formation. Highlighted are examples of
the administration of stored blood products, including TRALI. divergent MP effects on nitric oxide (NO) release, proreactive oxidative
RMPs typically form after 10 days of storage, whereas PMPs species (ROS) production, expression of inflammatory enzymes such as
accumulate earlier within days, and levels of both can be cyclooxygenase-2 (COX-2) or procoagulant factors such as tissue factor (TF),
significantly reduced if packed cells are leukoreduced or in the or angiogenic responses including proliferation and migration. Reproduced
from Benameur et al. (20) with kind permission from Pharmacological
presence of storage additives such as glucose, pyruvate, ino- Reports.
sine, adenine, and phosphate (80). Storage of platelets with
PGE1, theophylline, or aprotinin as well as pharmacological
inhibition of ␣IIb␤3-integrin signaling have been proposed to released by mesenchymal stem cells (MSCs) and may thus, at
reduce ex vivo PMP production (25, 33) and may thus help to least in part, account for the paracrine effects by which MSCs
reduce PMN activation and sequestration in the lungs subse- have emerged as a promising new therapeutic strategy in ALI
quent to transfusion of stored blood products. Compounds such (63, 92, 93, 107). MSCs secrete MPs that are enriched in
as cyclosporin A and Orai 1 inhibitors have been proposed as pre-miRNAs (35) and, in a rat renal injury model, could inhibit
a means of decreasing levels of EMPs and PMPs, respectively apoptosis and promote proliferation of kidney epithelial cells
(5, 10, 21, 32). Further work is required to examine the effects by paracrine effects that are in part mediated via intercellular
of eliminating or altering selected MP populations generated in transfer of mRNA and miRNA (58). MSCs have also recently
stored blood products to test whether such interventions may been shown to donate mitochondria to lung parenchymal cells
positively impact the mortality and morbidity of transfused in ALI by a microvesicular mechanism that could imply MPs
experimental animals and ultimately patients. (73); yet the relevance of mitochondrial transfer for the effects
of MSCs has been challenged by others (38). Further investi-
Potential Beneficial Effects of Microparticles in ALI gations are needed to characterize the contributions of MSC-
derived MPs in the benefits these cells appear to offer in the
MPs are not necessarily always associated with progressive setting of lung injury. In the following, we will discuss mech-
organ dysfunction and disease outcome. As recently discussed anisms that may potentially contribute to the beneficial effects
in seminal reviews by Morel and coworkers (120) and Dignat- of non-stem-cell-derived MPs in ALI focusing on the same
George and Boulanger (46), MPs are by no means always pathophysiological characteristics of the disease as previously
detrimental but may frequently exert beneficial effects, in that done for the detrimental role of MPs.
they exhibit anti-inflammatory and anticoagulant potential and Inflammation. Although we previously emphasized the role
can improve endothelial and vascular function under patholog- of TXA2 in the context of the delivery and release of AA and
ical conditions (Fig. 7). Experimental evidence for the benefi- AA metabolites by MPs, it must be taken into consideration
cial effects of MPs comprise the promotion of myocardial that not all eicosanoids are proinflammatory and increase
tissue repair postinfarction by PMPs, the stimulation of matrix barrier permeability. Notably, PMPs contain lipoxygenase 12
degradation and new blood vessel formation by EMPs through that, when transferred to mast cells, promotes production of the
activation of matrix metalloproteases, or the triggering of anti-inflammatory leukotriene lipoxin A4 (LXA4) (161). In a
angiogenic endothelial responses and the reconstitution of murine model of experimental colitis, injection of PMPs and
endothelial function and NO generation associated with re- LXA4 attenuated inflammatory responses, whereas lipoxygen-
duced ROS production by LMPs carrying the developmental ase 12 protein-deficient MPs or mast cell depletion reversed
morphogen sonic hedgehog (3, 20, 114). These findings exem- this beneficial effect (161). Since LXA4 analogs have been
plify the seemingly paradoxical beneficial effects that have shown to attenuate ALI in animal models (47, 76), MP-
been documented for a variety of MPs under different patho- mediated generation of LXA4 might prove beneficial in ALI.
logical conditions, a notion that gains particular relevance in Although certain MPs formed in ALI may have the ability to
the context of ALI by the clinical observation that higher levels cause detrimental immune modulation as described in previous
of circulating LMPs or EMPs, respectively, are associated with sections, others may confer beneficial immune functions capa-
increased survival in both sepsis and ARDS (65, 158). The ble of enhancing survival or reducing morbidity. NMPs can
potential role of beneficial MPs gains additional momentum in stimulate secretion of transforming growth factor-␤, which
light of the fact that considerable numbers of MPs can also be blocks macrophage activity, and can express annexin A1,

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
L376 MICROPARTICLES AND ACUTE LUNG INJURY

which acts as an anti-inflammatory protein blocking adhesion Va (95). Importantly, APC bound to EPCR in microparticulate
of PMNs to endothelial cells (40, 57). In addition, MPs gen- form retains anticoagulant activity (131), and has been shown
erated from KATO-III human gastric cancer cells can interact to exert both antiapoptotic and barrier-protective effects (130).
with monocytes and change their cytokine release profile from Permeability. APC may also exert important barrier-protec-
proinflammatory mediators such as TNF-␣ to anti-inflamma- tive effects, in that EPCR ligation promotes transactivation of
tory cytokines such as IL-10 (83). Although the relevance of sphingosine 1-phosphate receptor 1 (S1P1) via phosphatidyl-
such effects in the context of ALI/ARDS remains to be eluci- inositol 3-kinase (PI3K)-dependent phosphorylation (51).
dated, these findings underscore the anti-inflammatory poten- Hence, APC-bearing MPs may limit vascular permeability via the
tial harbored by many MPs. A recent analysis of ARDS well-documented barrier-enhancing signaling cascade down-
patients demonstrated elevations in both LMPs and NMPs stream of S1P1 (169). This notion is supported by experimental
within BAL samples of ARDS patients compared with spon- data demonstrating that EPCR/APC-bearing EMPs reduce endo-
taneously breathing controls (65), whereas levels of PMPs did thelial permeability in vitro via a mechanism that involves PAR-1,
not differ among ARDS patients compared with simplified PI3K, and notably also the vascular endothelial growth factor
acute physiology score (SAPS II) and sequential organ failure receptor 2 (130).
assessment (SOFA) score matched ventilated intensive care Endothelial function. Although MPs are frequently associ-
patient controls without ARDS and spontaneously breathing ated with endothelial and vascular dysfunction (vide supra),
patients being bronchoscopically investigated for suspected several studies by the group of Ramarosin Andriantsitohaina
chronic lung diseases. Notably, survivors of ARDS had statis- and Maria Carmen Martinez (3, 114, 121) have demonstrated
tically elevated levels of LMPs compared with the ventilated that the endothelium may also receive beneficial signals from
and spontaneously breathing control groups. This unexpected MPs carrying sonic hedgehog, resulting in correction of endo-
finding raises the possibility that LMPs may not only be a thelial injury, reconstitution of NO release, and reduced ROS
biomarker of ARDS and/or potentially promote inflammatory production. The role of NO in this context, however, is com-
signaling and parenchymal damage but may at times also be plex because it can exert both attenuating and aggravating
protective and reflect a “healthy” level of immune function. effects on inflammatory, coagulatory, or permeability changes
This physiological effect may be absent in severe inflammatory depending on its location, concentrations, and local microen-
diseases such as ARDS, causing “immune paralysis” or a vironment (85). Although a recent meta-analysis did not sup-
relative anergy of immune function that may have ramifica- port the rationale for inhaled NO as an effective treatment
tions on the host that directly impact mortality (123). strategy in ALI/ARDS (2), reconstitution of endogenous NO
Coagulation. Whereas the high expression levels of TF in production and an intact endothelial and vascular function may
MPs are generally considered to be procoagulant and barrier nonetheless provide important benefits that could positively
disruptive, they may also have anticoagulant and barrier- impact on clinical outcome in ALI/ARDS.
protective properties through PAR1-mediated transactivation Clinical implications. Taken together, MPs possess a variety
of PAR2 signaling pathways (82). In addition, MPs can express of mechanisms by which they may exert anti-inflammatory,
a variety of anticoagulant factors including thrombomodulin, anticoagulatory, and barrier-enhancing effects under healthy or
TFPI, endothelial protein C receptor (EPCR), and protein S diseased conditions. Although our understanding of these prop-
(120). Thus the potential involvement of MPs in anticoagulant erties of MPs in general, and in the context of ALI/ARDS in
pathways may present both an intrinsic protective mechanism particular, is still in its infancy, the potential to exploit this
and a possible future therapeutic target in lung injury. Notably, intrinsic property for therapeutic purposes poses a challenging
LMPs from hepatitis C patients have been shown to cause yet promising path to pursue.
increased fibrinolysis after fusion with hepatic stellate cells,
thus decreasing fibrin deposition (84). Synopsis and Implications
Activated protein C (APC) proteolytically inactivates key
coagulation factors including factor Va and factor VIIIa. Re- There has been little in the way of effective innovations and
combinant APC has been shown to reduce ALI and increase interventions in the treatment of ALI/ARDS beyond the intro-
alveolar ventilation in experimental animal models (71, 110, duction of protective ventilation (1) and restrictive fluid man-
167) and individual clinical case reports (81). In sepsis pa- agement strategies (176). ALI/ARDS is commonly associated
tients, treatment with APC has been reported to show a trend with increased formation of MPs from various cellular origins
toward increased survival that is notably associated with MP found in both the distal air spaces and circulating blood. These
formation and concomitant anti-inflammatory effects (132). MPs are small vesicles with an intact lipid bilayer that may
However, a recent Cochrane review did not recommend its use contain membrane and cytosolic proteins, lipids, organelles,
because of a failure to improve survival and an increased and genetic material from their parent cells that can function
bleeding risk, which has led to a decrease in its use as a therapy autonomously or transfer their contents to other cells. Although
for severe sepsis (105). Interestingly nevertheless in the con- most studies have looked at MPs as potential biomarkers for
text of this review, APC induces the release of MP-associated disease and progression of illness, MPs bear the potential to
EPCR that can bind APC and exert anticoagulatory (131) and convey both detrimental as well as beneficial effects with
potentially also anti-inflammatory and barrier-protective ef- respect to the excessive inflammatory, coagulatory, and per-
fects (120). EPCR (131) is a member of the major histocom- meability responses characteristic for ALI/ARDS (Fig. 6).
patibility complex/CD1 superfamily that promotes protein C Further investigations into MPs’ actions are required to better
activation at the cell surface (159), yet upon metalloprotease- understand their role in the pathophysiology of acute lung
mediated cleavage circulates in a soluble form (sEPCR) (88) disease and to utilize their potential for therapeutic interven-
that neutralizes APC so that it can no longer inactivate factor tions. Such concepts may include strategies to prevent the

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
MICROPARTICLES AND ACUTE LUNG INJURY L377
formation of MPs by altering flip-flop scramblase enzyme respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 297:
activities (116) or therapeutic manipulation of MP populations L1035–L1041, 2009.
17. Bastarache JA, Sebag SC, Grove BS, Ware LB. Interferon-gamma and
by pharmacological interventions (138). Even more attractive tumor necrosis factor-alpha act synergistically to up-regulate tissue factor
is the potential perspective to use specifically engineered “de- in alveolar epithelial cells. Exp Lung Res 37: 509 –517, 2011.
signer” MPs as vectors to move genes, proteins, or specific 18. Bastarache JA, Wang L, Geiser T, Wang Z, Albertine KH, Matthay
cellular functions between cells (20). Clearly, MPs add another MA, Ware LB. The alveolar epithelium can initiate the extrinsic coag-
layer of complexity to the already multifaceted mechanisms ulation cascade through expression of tissue factor. Thorax 62: 608 –616,
2007.
involved in ALI/ARDS, but in parallel they may offer hope of 19. Bastarache JA, Ware LB, Bernard GR. The role of the coagulation
obtainable and specific sites for future pharmacological manip- cascade in the continuum of sepsis and acute lung injury and acute
ulation. respiratory distress syndrome. Semin Respir Crit Care Med 27: 365–376,
2006.
ACKNOWLEDGMENTS 20. Benameur T, Andriantsitohaina R, Martinez MC. Therapeutic poten-
tial of plasma membrane-derived microparticles. Pharmacol Rep 61:
Illustrations in Figs. 4, 5, and 6 are courtesy of Stefania Spano.
49 –57, 2009.
REFERENCES 21. Bergmeier W, Oh-Hora M, McCarl CA, Roden RC, Bray PF, Feske
S. R93W mutation in Orai1 causes impaired calcium influx in platelets.
1. Acute Respiratory Distress Syndrome Network. Ventilation with Blood 113: 675–678, 2009.
lower tidal volumes as compared with traditional tidal volumes for acute 22. Beyer C, Pisetsky DS. The role of microparticles in the pathogenesis of
lung injury and the acute respiratory distress syndrome. N Engl J Med rheumatic diseases. Nat Rev Rheumatol 6: 21–29, 2010.
342: 1301–1308, 2000. 23. Bianco F, Perrotta C, Novellino L, Francolini M, Riganti L, Menna
2. Afshari A, Brok J, Moller AM, Wetterslev J. Inhaled nitric oxide for E, Saglietti L, Schuchman EH, Furlan R, Clementi E, Matteoli M,
acute respiratory distress syndrome and acute lung injury in adults and Verderio C. Acid sphingomyelinase activity triggers microparticle re-
children: a systematic review with meta-analysis and trial sequential lease from glial cells. EMBO J 28: 1043–1054, 2009.
analysis. Anesth Analg 112: 1411–1421, 2011. 24. Biro E, Akkerman JW, Hoek FJ, Gorter G, Pronk LM, Sturk A,
3. Agouni A, Mostefai HA, Porro C, Carusio N, Favre J, Richard V, Nieuwland R. The phospholipid composition and cholesterol content of
Henrion D, Martinez MC, Andriantsitohaina R. Sonic hedgehog platelet-derived microparticles: a comparison with platelet membrane
carried by microparticles corrects endothelial injury through nitric oxide fractions. J Thromb Haemost 3: 2754 –2763, 2005.
release. FASEB J 21: 2735–2741, 2007. 25. Bode AP, Orton SM, Frye MJ, Udis BJ. Vesiculation of platelets
4. Aharon A, Tamari T, Brenner B. Monocyte-derived microparticles and during in vitro aging. Blood 77: 887–895, 1991.
exosomes induce procoagulant and apoptotic effects on endothelial cells. 26. Bosma KJ, Taneja R, Lewis JF. Pharmacotherapy for prevention and
Thromb Haemost 100: 878 –885, 2008. treatment of acute respiratory distress syndrome: current and experimen-
5. Al-Massarani G, Vacher-Coponat H, Paul P, Arnaud L, Loundou A, tal approaches. Drugs 70: 1255–1282, 2010.
Robert S, Moal V, Berland Y, Dignat-George F, Camoin-Jau L. 27. Boulanger CM. Microparticles, vascular function and hypertension.
Kidney transplantation decreases the level and procoagulant activity of Curr Opin Nephrol Hypertens 19: 177–180, 2010.
circulating microparticles. Am J Transplant 9: 550 –557, 2009. 28. Breimo ES, Osterud B. Generation of tissue factor-rich microparticles
6. Aliotta JM, Pereira M, Johnson KW, de Paz N, Dooner MS, Puente in an ex vivo whole blood model. Blood Coagul Fibrinolysis 16:
N, Ayala C, Brilliant K, Berz D, Lee D, Ramratnam B, McMillan PN, 399 –405, 2005.
Hixson DC, Josic D, Quesenberry PJ. Microvesicle entry into marrow 29. Brodsky SV, Zhang F, Nasjletti A, Goligorsky MS. Endothelium-
cells mediates tissue-specific changes in mRNA by direct delivery of derived microparticles impair endothelial function in vitro. Am J Physiol
mRNA and induction of transcription. Exp Hematol 38: 233–245, 2010. Heart Circ Physiol 286: H1910 –H1915, 2004.
7. Amabile N, Rautou PE, Tedgui A, Boulanger CM. Microparticles: key 30. Brown GT, McIntyre TM. Lipopolysaccharide signaling without a
protagonists in cardiovascular disorders. Semin Thromb Hemost 36: nucleus: kinase cascades stimulate platelet shedding of proinflammatory
907–916, 2010. IL-1beta-rich microparticles. J Immunol 186: 5489 –5496, 2011.
8. Angelillo-Scherrer A. Leukocyte-derived microparticles in vascular
31. Buesing KL, Densmore JC, Kaul S, Pritchard KA Jr, Jarzembowski
homeostasis. Circ Res 110: 356 –369, 2012.
JA, Gourlay DM, Oldham KT. Endothelial microparticles induce
9. Aoki Y, Maeno T, Aoyagi K, Ueno M, Aoki F, Aoki N, Nakagawa J,
inflammation in acute lung injury. J Surg Res 166: 32–39, 2011.
Sando Y, Shimizu Y, Suga T, Arai M, Kurabayashi M. Pioglitazone,
32. Camaioni C, Gustapane M, Cialdella P, Della Bona R, Biasucci LM.
a peroxisome proliferator-activated receptor gamma ligand, suppresses
Microparticles and microRNAs: new players in the complex field of
bleomycin-induced acute lung injury and fibrosis. Respiration 77: 311–
coagulation. Intern Emerg Med 2011, Oct. 11. [Epub ahead of print]
319, 2009.
10. Arachiche A, Kerbiriou-Nabias D, Garcin I, Letellier T, Dachary- 33. Cauwenberghs S, Feijge MA, Harper AG, Sage SO, Curvers J,
Prigent J. Rapid procoagulant phosphatidylserine exposure relies on Heemskerk JW. Shedding of procoagulant microparticles from unstimu-
high cytosolic calcium rather than on mitochondrial depolarization. lated platelets by integrin-mediated destabilization of actin cytoskeleton.
Arterioscler Thromb Vasc Biol 29: 1883–1889, 2009. FEBS Lett 580: 5313–5320, 2006.
11. Aras O, Shet A, Bach RR, Hysjulien JL, Slungaard A, Hebbel RP, 34. Chaar V, Romana M, Tripette J, Broquere C, Huisse MG, Hue O,
Escolar G, Jilma B, Key NS. Induction of microparticle- and cell- Hardy-Dessources MD, Connes P. Effect of strenuous physical exercise
associated intravascular tissue factor in human endotoxemia. Blood 103: on circulating cell-derived microparticles. Clin Hemorheol Microcirc 47:
4545–4553, 2004. 15–25, 2011.
12. Barry OP, FitzGerald GA. Mechanisms of cellular activation by plate- 35. Chen TS, Lai RC, Lee MM, Choo AB, Lee CN, Lim SK. Mesenchy-
let microparticles. Thromb Haemost 82: 794 –800, 1999. mal stem cell secretes microparticles enriched in pre-microRNAs. Nu-
13. Barry OP, Kazanietz MG, Pratico D, FitzGerald GA. Arachidonic cleic Acids Res 38: 215–224, 2010.
acid in platelet microparticles up-regulates cyclooxygenase-2-dependent 36. Chironi GN, Boulanger CM, Simon A, Dignat-George F, Freyssinet
prostaglandin formation via a protein kinase C/mitogen-activated protein JM, Tedgui A. Endothelial microparticles in diseases. Cell Tissue Res
kinase-dependent pathway. J Biol Chem 274: 7545–7556, 1999. 335: 143–151, 2009.
14. Barry OP, Pratico D, Lawson JA, FitzGerald GA. Transcellular 37. Chung J, Suzuki H, Tabuchi N, Sato K, Shibamiya A, Koyama T.
activation of platelets and endothelial cells by bioactive lipids in platelet Identification of tissue factor and platelet-derived particles on leukocytes
microparticles. J Clin Invest 99: 2118 –2127, 1997. during cardiopulmonary bypass by flow cytometry and immunoelectron
15. Barry OP, Pratico D, Savani RC, FitzGerald GA. Modulation of microscopy. Thromb Haemost 98: 368 –374, 2007.
monocyte-endothelial cell interactions by platelet microparticles. J Clin 38. Colletti EJ, Airey JA, Liu W, Simmons PJ, Zanjani ED, Porada CD,
Invest 102: 136 –144, 1998. Almeida-Porada G. Generation of tissue-specific cells from MSC does
16. Bastarache JA, Fremont RD, Kropski JA, Bossert FR, Ware LB. not require fusion or donor-to-host mitochondrial/membrane transfer.
Procoagulant alveolar microparticles in the lungs of patients with acute Stem Cell Res 2: 125–138, 2009.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
L378 MICROPARTICLES AND ACUTE LUNG INJURY

39. Collino F, Deregibus MC, Bruno S, Sterpone L, Aghemo G, Viltono resting and activated platelets and platelet membrane microparticles in
L, Tetta C, Camussi G. Microvesicles derived from adult human bone normal subjects, and observations in patients during adult respiratory
marrow and tissue specific mesenchymal stem cells shuttle selected distress syndrome and cardiac surgery. J Clin Invest 78: 340 –348, 1986.
pattern of miRNAs. PLoS One 5: e11803, 2010. 61. Ghosh K, Shetty S. Blood coagulation in falciparum malaria—a review.
40. Dalli J, Norling LV, Renshaw D, Cooper D, Leung KY, Perretti M. Parasitol Res 102: 571–576, 2008.
Annexin 1 mediates the rapid anti-inflammatory effects of neutrophil- 62. Goggel R, Winoto-Morbach S, Vielhaber G, Imai Y, Lindner K,
derived microparticles. Blood 112: 2512–2519, 2008. Brade L, Brade H, Ehlers S, Slutsky AS, Schutze S, Gulbins E, Uhlig
41. Dasgupta SK, Le A, Chavakis T, Rumbaut RE, Thiagarajan P. S. PAF-mediated pulmonary edema: a new role for acid sphingomyeli-
Developmental endothelial locus-1 (del-1) mediates clearance of platelet nase and ceramide. Nat Med 10: 155–160, 2004.
microparticles by the endothelium. Circulation 125: 1664 –1672, 2012. 63. Gotts JE, Matthay MA. Mesenchymal stem cells and acute lung injury.
42. Davizon P, Munday AD, Lopez JA. Tissue factor, lipid rafts, and Crit Care Clin 27: 719 –733, 2011.
microparticles. Semin Thromb Hemost 36: 857–864, 2010. 64. Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus
43. Dean WL, Lee MJ, Cummins TD, Schultz DJ, Powell DW. Proteomic MC, Tetta C, Bussolati B, Camussi G. Microvesicles released from
and functional characterisation of platelet microparticle size classes. human renal cancer stem cells stimulate angiogenesis and formation of
Thromb Haemost 102: 711–718, 2009. lung premetastatic niche. Cancer Res 71: 5346 –5356, 2011.
44. Densmore JC, Signorino PR, Ou J, Hatoum OA, Rowe JJ, Shi Y, 65. Guervilly C, Lacroix R, Forel JM, Roch A, Camoin-Jau L, Papazian
Kaul S, Jones DW, Sabina RE, Pritchard KA Jr, Guice KS, Oldham L, Dignat-George F. High levels of circulating leukocyte microparticles
KT. Endothelium-derived microparticles induce endothelial dysfunction are associated with better outcome in acute respiratory distress syndrome.
and acute lung injury. Shock 26: 464 –471, 2006. Crit Care 15: R31, 2011.
45. Deregibus MC, Cantaluppi V, Calogero R, Lo Iacono M, Tetta C, 66. Hanafusa N, Satonaka H, Doi K, Yatomi Y, Noiri E, Fujita T.
Biancone L, Bruno S, Bussolati B, Camussi G. Endothelial progenitor Platelet-derived microparticles are removed by a membrane plasma
cell derived microvesicles activate an angiogenic program in endothelial separator. ASAIO J 56: 323–325, 2010.
cells by a horizontal transfer of mRNA. Blood 110: 2440 –2448, 2007. 67. Hansmann G, de Jesus Perez VA, Alastalo TP, Alvira CM, Guign-
46. Dignat-George F, Boulanger CM. The many faces of endothelial abert C, Bekker JM, Schellong S, Urashima T, Wang L, Morrell
microparticles. Arterioscler Thromb Vasc Biol 31: 27–33, 2011. NW, Rabinovitch M. An antiproliferative BMP-2/PPARgamma/apoE
47. El Kebir D, Jozsef L, Pan W, Wang L, Petasis NA, Serhan CN, Filep axis in human and murine SMCs and its role in pulmonary hypertension.
JG. 15-epi-lipoxin A4 inhibits myeloperoxidase signaling and enhances J Clin Invest 118: 1846 –1857, 2008.
resolution of acute lung injury. Am J Respir Crit Care Med 180: 68. Hansmann G, Wagner RA, Schellong S, Perez VA, Urashima T,
311–319, 2009. Wang L, Sheikh AY, Suen RS, Stewart DJ, Rabinovitch M. Pulmo-
48. Enkhbaatar P, Okajima K, Murakami K, Uchiba M, Okabe H, nary arterial hypertension is linked to insulin resistance and reversed by
Okabe K, Yamaguchi Y. Recombinant tissue factor pathway inhibitor peroxisome proliferator-activated receptor-gamma activation. Circula-
reduces lipopolysaccharide-induced pulmonary vascular injury by inhib- tion 115: 1275–1284, 2007.
iting leukocyte activation. Am J Respir Crit Care Med 162: 1752–1759,
69. Hess C, Sadallah S, Hefti A, Landmann R, Schifferli JA. Ectosomes
2000.
released by human neutrophils are specialized functional units. J Immu-
49. Essayagh S, Brisset AC, Terrisse AD, Dupouy D, Tellier L, Navarro
nol 163: 4564 –4573, 1999.
C, Arnal JF, Sie P. Microparticles from apoptotic vascular smooth
70. Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, Xiao T,
muscle cells induce endothelial dysfunction, a phenomenon prevented by
Schafer J, Lee ML, Schmittgen TD, Nana-Sinkam SP, Jarjoura D,
beta3-integrin antagonists. Thromb Haemost 94: 853–858, 2005.
Marsh CB. Detection of microRNA expression in human peripheral
50. Faille D, El-Assaad F, Mitchell AJ, Alessi MC, Chimini G, Fusai T,
blood microvesicles. PLoS One 3: e3694, 2008.
Grau GE, Combes V. Endocytosis and intracellular processing of
71. Husari AW, Khayat A, Awdeh H, Hatoum H, Nasser M, Mroueh
platelet microparticles by brain endothelial cells. J Cell Mol Med 2011
SM, Zaatari G, El-Sabban M, Dbaibo GS. Activated protein C atten-
Aug 29. doi: 10.1111/j.1582-4934.2011.01434.x. [Epub ahead of print]
51. Finigan JH, Dudek SM, Singleton PA, Chiang ET, Jacobson JR, uates acute lung injury and apoptosis in a hyperoxic animal model. Shock
Camp SM, Ye SQ, Garcia JG. Activated protein C mediates novel lung 33: 467–472, 2010.
endothelial barrier enhancement: role of sphingosine 1-phosphate recep- 72. Ishitsuka Y, Moriuchi H, Isohama Y, Tokunaga H, Hatamoto K,
tor transactivation. J Biol Chem 280: 17286 –17293, 2005. Kurita S, Irikura M, Iyama K, Irie T. A selective thromboxane A2
52. Fink K, Feldbrugge L, Schwarz M, Bourgeois N, Helbing T, Bode C, (TXA2) synthase inhibitor, ozagrel, attenuates lung injury and decreases
Schwab T, Busch HJ. Circulating annexin V positive microparticles in monocyte chemoattractant protein-1 and interleukin-8 mRNA expression
patients after successful cardiopulmonary resuscitation. Crit Care 15: in oleic acid-induced lung injury in guinea pigs. J Pharm Sci 111:
R251, 2011. 211–215, 2009.
53. Flaumenhaft R. Formation and fate of platelet microparticles. Blood 73. Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K,
Cells Mol Dis 36: 182–187, 2006. Rowlands DJ, Quadri SK, Bhattacharya S, Bhattacharya J. Mito-
54. Flaumenhaft R, Mairuhu AT, Italiano JE. Platelet- and megakaryo- chondrial transfer from bone-marrow-derived stromal cells to pulmonary
cyte-derived microparticles. Semin Thromb Hemost 36: 881–887, 2010. alveoli protects against acute lung injury. Nat Med 18: 759 –765, 2012.
55. Forest A, Pautas E, Ray P, Bonnet D, Verny M, Amabile N, Bou- 74. Italiano JE Jr, Mairuhu AT, Flaumenhaft R. Clinical relevance of
langer C, Riou B, Tedgui A, Mallat Z, Boddaert J. Circulating microparticles from platelets and megakaryocytes. Curr Opin Hematol
microparticles and procoagulant activity in elderly patients. J Gerontol A 17: 578 –584, 2010.
Biol Sci Med Sci 65: 414 –420, 2010. 75. Jin M, Drwal G, Bourgeois T, Saltz J, Wu HM. Distinct proteome
56. Gando S, Kameue T, Matsuda N, Hayakawa M, Morimoto Y, features of plasma microparticles. Proteomics 5: 1940 –1952, 2005.
Ishitani T, Kemmotsu O. Imbalances between the levels of tissue factor 76. Jin SW, Zhang L, Lian QQ, Liu D, Wu P, Yao SL, Ye DY.
and tissue factor pathway inhibitor in ARDS patients. Thromb Res 109: Posttreatment with aspirin-triggered lipoxin A4 analog attenuates lipopo-
119 –124, 2003. lysaccharide-induced acute lung injury in mice: the role of heme oxy-
57. Gasser O, Schifferli JA. Activated polymorphonuclear neutrophils dis- genase-1. Anesth Analg 104: 369 –377, 2007.
seminate anti-inflammatory microparticles by ectocytosis. Blood 104: 77. Jy W, Horstman LL, Ahn YS. Microparticle size and its relation to
2543–2548, 2004. composition, functional activity, and clinical significance. Semin Thromb
58. Gatti S, Bruno S, Deregibus MC, Sordi A, Cantaluppi V, Tetta C, Hemost 36: 876 –880, 2010.
Camussi G. Microvesicles derived from human adult mesenchymal stem 78. Jy W, Horstman LL, Jimenez JJ, Ahn YS, Biro E, Nieuwland R,
cells protect against ischaemia-reperfusion-induced acute and chronic Sturk A, Dignat-George F, Sabatier F, Camoin-Jau L, Sampol J,
kidney injury. Nephrol Dial Transplant 26: 1474 –1483, 2011. Hugel B, Zobairi F, Freyssinet JM, Nomura S, Shet AS, Key NS,
59. Gauley J, Pisetsky DS. The release of microparticles by RAW 264.7 Hebbel RP. Measuring circulating cell-derived microparticles. J Thromb
macrophage cells stimulated with TLR ligands. J Leukoc Biol 87: Haemost 2: 1842–1851, 2004.
1115–1123, 2010. 79. Jy W, Mao WW, Horstman L, Tao J, Ahn YS. Platelet microparticles
60. George JN, Pickett EB, Saucerman S, McEver RP, Kunicki TJ, bind, activate and aggregate neutrophils in vitro. Blood Cells Mol Dis 21:
Kieffer N, Newman PJ. Platelet surface glycoproteins. Studies on 217–231; discussion 231a, 1995.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
MICROPARTICLES AND ACUTE LUNG INJURY L379
80. Jy W, Ricci M, Shariatmadar S, Gomez-Marin O, Horstman LH, 103. Mackman N. Role of tissue factor in hemostasis and thrombosis. Blood
Ahn YS. Microparticles in stored red blood cells as potential mediators Cells Mol Dis 36: 104 –107, 2006.
of transfusion complications. Transfusion 51: 886 –893, 2011. 104. Maniatis NA, Kotanidou A, Catravas JD, Orfanos SE. Endothelial
81. Kallet RH, Jasmer RM, Pittet JF. Alveolar dead-space response to pathomechanisms in acute lung injury. Vascul Pharmacol 49: 119 –133,
activated protein C in acute respiratory distress syndrome. Respir Care 2008.
55: 617–622, 2010. 105. Marti-Carvajal AJ, Sola I, Lathyris D, Cardona AF. Human recom-
82. Kaneider NC, Leger AJ, Agarwal A, Nguyen N, Perides G, Derian C, binant activated protein C for severe sepsis. Cochrane Database Syst Rev
Covic L, Kuliopulos A. ‘Role reversal’ for the receptor PAR1 in 4: CD004388, 2011.
sepsis-induced vascular damage. Nat Immunol 8: 1303–1312, 2007. 106. Martin S, Tesse A, Hugel B, Martinez MC, Morel O, Freyssinet JM,
83. Koppler B, Cohen C, Schlondorff D, Mack M. Differential mecha- Andriantsitohaina R. Shed membrane particles from T lymphocytes
nisms of microparticle transfer to B cells and monocytes: anti-inflam- impair endothelial function and regulate endothelial protein expression.
matory propertiesof microparticles. Eur J Immunol 36: 648 –660, 2006. Circulation 109: 1653–1659, 2004.
84. Kornek M, Popov Y, Libermann TA, Afdhal NH, Schuppan D. 107. Matthay MA, Goolaerts A, Howard JP, Lee JW. Mesenchymal stem
Human T cell microparticles circulate in blood of hepatitis patients and cells for acute lung injury: preclinical evidence. Crit Care Med 38:
induce fibrolytic activation of hepatic stellate cells. Hepatology 53: S569 –S573, 2010.
230 –242, 2011. 108. Matthay MA, Zimmerman GA. Acute lung injury and the acute
85. Kuebler WM. The Janus-faced regulation of endothelial permeability by respiratory distress syndrome: four decades of inquiry into pathogenesis
cyclic GMP. Am J Physiol Lung Cell Mol Physiol 301: L157–L160, and rational management. Am J Respir Cell Mol Biol 33: 319 –327, 2005.
2011. 109. Mause SF, Weber C. Microparticles: protagonists of a novel commu-
86. Kuebler WM. Statins STAT for (over)ventilated patients? Crit Care 14: nication network for intercellular information exchange. Circ Res 107:
1014, 2010. 1047–1057, 2010.
87. Kuebler WM, Yang Y, Samapati R, Uhlig S. Vascular barrier regula- 110. Maybauer MO, Maybauer DM, Fraser JF, Szabo C, Westphal M,
tion by PAF, ceramide, caveolae, and NO—an intricate signaling net- Kiss L, Horvath EM, Nakano Y, Herndon DN, Traber LD, Traber
work with discrepant effects in the pulmonary and systemic vasculature. DL. Recombinant human activated protein C attenuates cardiovascular
Cell Physiol Biochem 26: 29 –40, 2010. and microcirculatory dysfunction in acute lung injury and septic shock.
88. Kurosawa S, Stearns-Kurosawa DJ, Hidari N, Esmon CT. Identifi- Crit Care 14: R217, 2010.
cation of functional endothelial protein C receptor in human plasma. J 111. Mesri M, Altieri DC. Endothelial cell activation by leukocyte micro-
Clin Invest 100: 411–418, 1997. particles. J Immunol 161: 4382–4387, 1998.
89. Lacroix R, Robert S, Poncelet P, Dignat-George F. Overcoming 112. Mesri M, Altieri DC. Leukocyte microparticles stimulate endothelial
limitations of microparticle measurement by flow cytometry. Semin cell cytokine release and tissue factor induction in a JNK1 signaling
Thromb Hemost 36: 807–818, 2010. pathway. J Biol Chem 274: 23111–23118, 1999.
90. Lacroix R, Robert S, Poncelet P, Kasthuri RS, Key NS, Dignat- 113. Meziani F, Delabranche X, Asfar P, Toti F. Bench-to-bedside review:
George F. Standardization of platelet-derived microparticle enumeration circulating microparticles—a new player in sepsis? Crit Care 14: 236,
by flow cytometry with calibrated beads: results of the International 2010.
Society on Thrombosis and Haemostasis SSC Collaborative workshop. J 114. Meziani F, Tesse A, Andriantsitohaina R. Microparticles are vectors of
Thromb Haemost 8: 2571–2574, 2010. paradoxical information in vascular cells including the endothelium: role
91. Lechner D, Weltermann A. Circulating tissue factor-exposing micro- in health and diseases. Pharmacol Rep 60: 75–84, 2008.
particles. Thromb Res 122, Suppl 1: S47–S54, 2008. 115. Miguet L, Pacaud K, Felden C, Hugel B, Martinez MC, Freyssinet
92. Lee JW, Fang X, Krasnodembskaya A, Howard JP, Matthay MA. JM, Herbrecht R, Potier N, van Dorsselaer A, Mauvieux L. Pro-
Concise review: Mesenchymal stem cells for acute lung injury: role of teomic analysis of malignant lymphocyte membrane microparticles using
paracrine soluble factors. Stem Cells 29: 913–919, 2011. double ionization coverage optimization. Proteomics 6: 153–171, 2006.
93. Lee JW, Gupta N, Serikov V, Matthay MA. Potential application of 116. Morel O, Jesel L, Freyssinet JM, Toti F. Cellular mechanisms under-
mesenchymal stem cells in acute lung injury. Expert Opin Biol Ther 9: lying the formation of circulating microparticles. Arterioscler Thromb
1259 –1270, 2009. Vasc Biol 31: 15–26, 2011.
94. Li Z, Yang F, Dunn S, Gross AK, Smyth SS. Platelets as immune 117. Morel O, Morel N, Freyssinet JM, Toti F. Platelet microparticles and
mediators: their role in host defense responses and sepsis. Thromb Res vascular cells interactions: a checkpoint between the haemostatic and
127: 184 –188, 2011. thrombotic responses. Platelets 19: 9 –23, 2008.
95. Liaw PC, Neuenschwander PF, Smirnov MD, Esmon CT. Mecha- 118. Morel O, Pereira B, Averous G, Faure A, Jesel L, Germain P,
nisms by which soluble endothelial cell protein C receptor modulates Grunebaum L, Ohlmann P, Freyssinet JM, Bareiss P, Toti F. In-
protein C and activated protein C function. J Biol Chem 275: 5447–5452, creased levels of procoagulant tissue factor-bearing microparticles within
2000. the occluded coronary artery of patients with ST-segment elevation
96. Little KM, Smalley DM, Harthun NL, Ley K. The plasma micropar- myocardial infarction: role of endothelial damage and leukocyte activa-
ticle proteome. Semin Thromb Hemost 36: 845–856, 2010. tion. Atherosclerosis 204: 636 –641, 2009.
97. Liu D, Zeng BX, Shang Y. Decreased expression of peroxisome pro- 119. Morel O, Toti F, Jesel L, Freyssinet JM. Mechanisms of microparticle
liferator-activated receptor gamma in endotoxin-induced acute lung in- generation: on the trail of the mitochondrion! Semin Thromb Hemost 36:
jury. Physiol Res 55: 291–299, 2006. 833–844, 2010.
98. Liu D, Zeng BX, Zhang SH, Wang YL, Zeng L, Geng ZL, Zhang SF. 120. Morel O, Toti F, Morel N, Freyssinet JM. Microparticles in endothelial
Rosiglitazone, a peroxisome proliferator-activated receptor-gamma ago- cell and vascular homeostasis: are they really noxious? Haematologica
nist, reduces acute lung injury in endotoxemic rats. Crit Care Med 33: 94: 313–317, 2009.
2309 –2316, 2005. 121. Mostefai HA, Agouni A, Carusio N, Mastronardi ML, Heymes C,
99. Lo SC, Hung CY, Lin DT, Peng HC, Huang TF. Involvement of Henrion D, Andriantsitohaina R, Martinez MC. Phosphatidylinositol
platelet glycoprotein Ib in platelet microparticle mediated neutrophil 3-kinase and xanthine oxidase regulate nitric oxide and reactive oxygen
activation. J Biomed Sci 13: 787–796, 2006. species productions by apoptotic lymphocyte microparticles in endothe-
100. Looney MR, Nguyen JX, Hu Y, Van Ziffle JA, Lowell CA, Matthay lial cells. J Immunol 180: 5028 –5035, 2008.
MA. Platelet depletion and aspirin treatment protect mice in a two-event 122. Mostefai HA, Meziani F, Mastronardi ML, Agouni A, Heymes C,
model of transfusion-related acute lung injury. J Clin Invest 119: 3450 – Sargentini C, Asfar P, Martinez MC, Andriantsitohaina R. Circulat-
3461, 2009. ing microparticles from patients with septic shock exert protective role in
101. Machado GB, de Assis MC, Leao R, Saliba AM, Silva MC, Suassuna vascular function. Am J Respir Crit Care Med 178: 1148 –1155, 2008.
JH, de Oliveira AV, Plotkowski MC. ExoU-induced vascular hyper- 123. Munford RS, Pugin J. Normal responses to injury prevent systemic
permeability and platelet activation in the course of experimental Pseu- inflammation and can be immunosuppressive. Am J Respir Crit Care
domonas aeruginosa pneumosepsis. Shock 33: 315–321, 2010. Med 163: 316 –321, 2001.
102. MacKenzie A, Wilson HL, Kiss-Toth E, Dower SK, North RA, 124. Mutschler DK, Larsson AO, Basu S, Nordgren A, Eriksson MB.
Surprenant A. Rapid secretion of interleukin-1beta by microvesicle Effects of mechanical ventilation on platelet microparticles in bronchoal-
shedding. Immunity 15: 825–835, 2001. veolar lavage fluid. Thromb Res 108: 215–220, 2002.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
L380 MICROPARTICLES AND ACUTE LUNG INJURY

125. Nolan S, Dixon R, Norman K, Hellewell P, Ridger V. Nitric oxide mRNA in normal pregnancy and pre-eclampsia. Placenta 29: 942–949,
regulates neutrophil migration through microparticle formation. Am J 2008.
Pathol 172: 265–273, 2008. 147. Robert S, Poncelet P, Lacroix R, Arnaud L, Giraudo L, Hauchard A,
126. Ogura H, Tanaka H, Koh T, Fujita K, Fujimi S, Nakamori Y, Sampol J, Dignat-George F. Standardization of platelet-derived micro-
Hosotsubo H, Kuwagata Y, Shimazu T, Sugimoto H. Enhanced particle counting using calibrated beads and a Cytomics FC500 routine
production of endothelial microparticles with increased binding to leu- flow cytometer: a first step towards multicenter studies? J Thromb
kocytes in patients with severe systemic inflammatory response syn- Haemost 7: 190 –197, 2009.
drome. J Trauma 56: 823–830; discussion 830 – 821, 2004. 148. Rosing J, Speijer H, Zwaal RF. Prothrombin activation on phospholipid
127. Osterud B. The role of platelets in decrypting monocyte tissue factor. membranes with positive electrostatic potential. Biochemistry 27: 8 –11,
Dis Mon 49: 7–13, 2003. 1988.
128. Osterud B. Tissue factor expression in blood cells. Thromb Res 125, 149. Samapati R, Yang Y, Yin J, Stoerger C, Arenz C, Dietrich A,
Suppl 1: S31–S34, 2010. Gudermann T, Adam D, Wu S, Freichel M, Flockerzi V, Uhlig S,
129. Owens AP 3rd, Mackman N. Microparticles in hemostasis and throm- Kuebler WM. Lung endothelial Ca2⫹ and permeability response to
bosis. Circ Res 108: 1284 –1297, 2011. platelet-activating factor is mediated by acid sphingomyelinase and
130. Perez-Casal M, Downey C, Cutillas-Moreno B, Zuzel M, Fukudome transient receptor potential classical 6. Am J Respir Crit Care Med 185:
K, Toh CH. Microparticle-associated endothelial protein C receptor and 160 –170, 2012.
the induction of cytoprotective and anti-inflammatory effects. Haemato-
150. Schaefer MB, Pose A, Ott J, Hecker M, Behnk A, Schulz R, Weiss-
logica 94: 387–394, 2009.
mann N, Gunther A, Seeger W, Mayer K. Peroxisome proliferator-
131. Perez-Casal M, Downey C, Fukudome K, Marx G, Toh CH. Acti-
activated receptor-alpha reduces inflammation and vascular leakage in a
vated protein C induces the release of microparticle-associated endothe-
murine model of acute lung injury. Eur Respir J 32: 1344 –1353, 2008.
lial protein C receptor. Blood 105: 1515–1522, 2005.
151. Schwarz MI, Albert RK. “Imitators” of the ARDS: implications for
132. Perez-Casal M, Thompson V, Downey C, Welters I, Wyncoll D,
Thachil J, Toh CH. The clinical and functional relevance of micropar- diagnosis and treatment. Chest 125: 1530 –1535, 2004.
ticles induced by activated protein C treatment in sepsis. Crit Care 15: 152. Semple JW, Italiano JE Jr, Freedman J. Platelets and the immune
R195, 2011. continuum. Nat Rev Immunol 11: 264 –274, 2011.
133. Phua J, Badia JR, Adhikari NK, Friedrich JO, Fowler RA, Singh 153. Shet AS. Characterizing blood microparticles: technical aspects and
JM, Scales DC, Stather DR, Li A, Jones A, Gattas DJ, Hallett D, challenges. Vasc Health Risk Manag 4: 769 –774, 2008.
Tomlinson G, Stewart TE, Ferguson ND. Has mortality from acute 154. Shirafuji T, Hamaguchi H, Higuchi M, Kanda F. Measurement of
respiratory distress syndrome decreased over time? A systematic review. platelet-derived microparticle levels using an enzyme-linked immunosor-
Am J Respir Crit Care Med 179: 220 –227, 2009. bent assay in polymyositis and dermatomyositis patients. Muscle Nerve
134. Piccin A, Murphy WG, Smith OP. Circulating microparticles: patho- 39: 586 –590, 2009.
physiology and clinical implications. Blood Rev 21: 157–171, 2007. 155. Simak J, Gelderman MP. Cell membrane microparticles in blood and
135. Pirro M, Schillaci G, Bagaglia F, Menecali C, Paltriccia R, Manna- blood products: potentially pathogenic agents and diagnostic markers.
rino MR, Capanni M, Velardi A, Mannarino E. Microparticles de- Transfus Med Rev 20: 1–26, 2006.
rived from endothelial progenitor cells in patients at different cardiovas- 156. Sinauridze EI, Kireev DA, Popenko NY, Pichugin AV, Panteleev
cular risk. Atherosclerosis 197: 757–767, 2008. MA, Krymskaya OV, Ataullakhanov FI. Platelet microparticle mem-
136. Porro C, Lepore S, Trotta T, Castellani S, Ratclif L, Battaglino A, Di branes have 50- to 100-fold higher specific procoagulant activity than
Gioia S, Martinez MC, Conese M, Maffione AB. Isolation and char- activated platelets. Thromb Haemost 97: 425–434, 2007.
acterization of microparticles in sputum from cystic fibrosis patients. 157. Singer P, Shapiro H. Enteral omega-3 in acute respiratory distress
Respir Res 11: 94, 2010. syndrome. Curr Opin Clin Nutr Metab Care 12: 123–128, 2009.
137. Prokopi M, Pula G, Mayr U, Devue C, Gallagher J, Xiao Q, Bou- 158. Soriano AO, Jy W, Chirinos JA, Valdivia MA, Velasquez HS,
langer CM, Westwood N, Urbich C, Willeit J, Steiner M, Breuss J, Jimenez JJ, Horstman LL, Kett DH, Schein RM, Ahn YS. Levels of
Xu Q, Kiechl S, Mayr M. Proteomic analysis reveals presence of endothelial and platelet microparticles and their interactions with leuko-
platelet microparticles in endothelial progenitor cell cultures. Blood 114: cytes negatively correlate with organ dysfunction and predict mortality in
723–732, 2009. severe sepsis. Crit Care Med 33: 2540 –2546, 2005.
138. Puddu P, Puddu GM, Cravero E, Muscari S, Muscari A. The 159. Stearns-Kurosawa DJ, Kurosawa S, Mollica JS, Ferrell GL, Esmon
involvement of circulating microparticles in inflammation, coagulation CT. The endothelial cell protein C receptor augments protein C activa-
and cardiovascular diseases. Can J Cardiol 26: 140 –145, 2010. tion by the thrombin-thrombomodulin complex. Proc Natl Acad Sci USA
139. Quesenberry PJ, Aliotta JM. Cellular phenotype switching and mi- 93: 10212–10216, 1996.
crovesicles. Adv Drug Deliv Rev 62: 1141–1148, 2010. 160. Tabuchi A, Kuebler WM. Endothelium-platelet interactions in inflam-
140. Ramacciotti E, Hawley AE, Farris DM, Ballard NE, Wrobleski SK, matory lung disease. Vascul Pharmacol 49: 141–150, 2008.
Myers DD Jr, Henke PK, Wakefield TW. Leukocyte- and platelet- 161. Tang K, Liu J, Yang Z, Zhang B, Zhang H, Huang C, Ma J, Shen
derived microparticles correlate with thrombus weight and tissue factor GX, Ye D, Huang B. Microparticles mediate enzyme transfer from
activity in an experimental mouse model of venous thrombosis. Thromb
platelets to mast cells: a new pathway for lipoxin A4 biosynthesis.
Haemost 101: 748 –754, 2009.
Biochem Biophys Res Commun 400: 432–436, 2010.
141. Rand ML, Wang H, Bang KW, Packham MA, Freedman J. Rapid
162. Terrisse AD, Puech N, Allart S, Gourdy P, Xuereb JM, Payrastre B,
clearance of procoagulant platelet-derived microparticles from the circu-
Sie P. Internalization of microparticles by endothelial cells promotes
lation of rabbits. J Thromb Haemost 4: 1621–1623, 2006.
142. Rank A, Nieuwland R, Crispin A, Grutzner S, Iberer M, Toth B, platelet/endothelial cell interaction under flow. J Thromb Haemost 8:
Pihusch R. Clearance of platelet microparticles in vivo. Platelets 22: 2810 –2819, 2010.
111–116, 2011. 163. Toth B, Nikolajek K, Rank A, Nieuwland R, Lohse P, Pihusch V,
143. Rank A, Nieuwland R, Delker R, Kohler A, Toth B, Pihusch V, Friese K, Thaler CJ. Gender-specific and menstrual cycle dependent
Wilkowski R, Pihusch R. Cellular origin of platelet-derived micropar- differences in circulating microparticles. Platelets 18: 515–521, 2007.
ticles in vivo. Thromb Res 126: e255–e259, 2010. 164. Ullal AJ, Pisetsky DS. The release of microparticles by Jurkat leukemia
144. Rank A, Nieuwland R, Liebhardt S, Iberer M, Grutzner S, Toth B, T cells treated with staurosporine and related kinase inhibitors to induce
Pihusch R. Apheresis platelet concentrates contain platelet-derived and apoptosis. Apoptosis 15: 586 –596, 2010.
endothelial cell-derived microparticles. Vox Sang 100: 179 –186, 2011. 165. van der Poll T. Tissue factor as an initiator of coagulation and inflam-
145. Rauch U, Bonderman D, Bohrmann B, Badimon JJ, Himber J, mation in the lung. Crit Care 12, Suppl 6: S3, 2008.
Riederer MA, Nemerson Y. Transfer of tissue factor from leukocytes to 166. Vickers KC, Remaley AT. Lipid-based carriers of microRNAs and
platelets is mediated by CD15 and tissue factor. Blood 96: 170 –175, intercellular communication. Curr Opin Lipidol 23: 91–97, 2012.
2000. 167. Waerhaug K, Kuzkov VV, Kuklin VN, Mortensen R, Nordhus KC,
146. Reddy A, Zhong XY, Rusterholz C, Hahn S, Holzgreve W, Redman Kirov MY, Bjertnaes LJ. Inhaled aerosolised recombinant human
CW, Sargent IL. The effect of labour and placental separation on the activated protein C ameliorates endotoxin-induced lung injury in anaes-
shedding of syncytiotrophoblast microparticles, cell-free DNA and thetised sheep. Crit Care 13: R51, 2009.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org


Review
MICROPARTICLES AND ACUTE LUNG INJURY L381
168. Wang C, Mody M, Herst R, Sher G, Freedman J. Flow cytometric 178. Xiao HY, Matsubayashi H, Bonderman DP, Bonderman PW, Reid T,
analysis of platelet function in stored platelet concentrates. Transfus Sci Miraglia CC, Gao DY. Generation of annexin V-positive platelets and
20: 129 –139, 1999. shedding of microparticles with stimulus-dependent procoagulant activ-
169. Wang L, Dudek SM. Regulation of vascular permeability by sphin- ity during storage of platelets at 4 degrees C. Transfusion 40: 420 –427,
gosine 1-phosphate. Microvasc Res 77: 39 –45, 2009. 2000.
170. Ware LB. Pathophysiology of acute lung injury and the acute respiratory 179. Yang Y, Yin J, Baumgartner W, Samapati R, Solymosi EA, Reppien
distress syndrome. Semin Respir Crit Care Med 27: 337–349, 2006. E, Kuebler WM, Uhlig S. Platelet-activating factor reduces endothelial
171. Ware LB, Matthay MA. The acute respiratory distress syndrome. N nitric oxide production: role of acid sphingomyelinase. Eur Respir J 36:
Engl J Med 342: 1334 –1349, 2000. 417–427, 2010.
172. Watanabe J, Marathe GK, Neilsen PO, Weyrich AS, Harrison KA, 180. Yuan A, Farber EL, Rapoport AL, Tejada D, Deniskin R, Akhmedov
Murphy RC, Zimmerman GA, McIntyre TM. Endotoxins stimulate NB, Farber DB. Transfer of microRNAs by embryonic stem cell
neutrophil adhesion followed by synthesis and release of platelet-acti- microvesicles. PLoS One 4: e4722, 2009.
vating factor in microparticles. J Biol Chem 278: 33161–33168, 2003. 181. Yuana Y, Oosterkamp TH, Bahatyrova S, Ashcroft B, Garcia Ro-
173. Webster NR, Galley HF. Immunomodulation in the critically ill. Br J driguez P, Bertina RM, Osanto S. Atomic force microscopy: a novel
Anaesth 103: 70 –81, 2009. approach to the detection of nanosized blood microparticles. J Thromb
174. Weerheim AM, Kolb AM, Sturk A, Nieuwland R. Phospholipid Haemost 8: 315–323, 2010.
composition of cell-derived microparticles determined by one-dimen- 182. Zarbock A, Singbartl K, Ley K. Complete reversal of acid-induced
sional high-performance thin-layer chromatography. Anal Biochem 302: acute lung injury by blocking of platelet-neutrophil aggregation. J Clin
191–198, 2002. Invest 116: 3211–3219, 2006.
175. Welty-Wolf KE, Carraway MS, Ortel TL, Ghio AJ, Idell S, Egan J, 183. Zhou T, Garcia JG, Zhang W. Integrating microRNAs into a system
Zhu X, Jiao JA, Wong HC, Piantadosi CA. Blockade of tissue biology approach to acute lung injury. Transl Res 157: 180 –190, 2011.
factor-factor X binding attenuates sepsis-induced respiratory and renal 184. Zimmerman GA. Thinking small, but with big league consequences:
failure. Am J Physiol Lung Cell Mol Physiol 290: L21–L31, 2006. procoagulant microparticles in the alveolar space. Am J Physiol Lung
176. Wiedemann HP, Wheeler AP, Bernard GR, Thompson BT, Hayden Cell Mol Physiol 297: L1033–L1034, 2009.
D, deBoisblanc B, Connors AF Jr, Hite RD, Harabin AL. Comparison 185. Zwicker JI. Impedance-based flow cytometry for the measurement of
of two fluid-management strategies in acute lung injury. N Engl J Med microparticles. Semin Thromb Hemost 36: 819 –823, 2010.
354: 2564 –2575, 2006. 186. Zwicker JI, Lacroix R, Dignat-George F, Furie BC, Furie B. Mea-
177. Wolf P. The nature and significance of platelet products in human surement of platelet microparticles. Methods Mol Biol 788: 127–139,
plasma. Br J Haematol 13: 269 –288, 1967. 2012.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00354.2011 • www.ajplung.org

You might also like