ArticlePDF AvailableLiterature Review

A Pathophysiologic Approach to Biomarkers in Acute Respiratory Distress Syndrome

Authors:

Abstract and Figures

Acute respiratory distress syndrome (ARDS) is an acute-onset hypoxic condition with radiographic bilateral lung infiltration. It is characterized by an acute exudative phase combining diffuse alveolar damage and lung edema followed by a later fibroproliferative phase. Despite an improved understanding of ARDS pathobiology, our ability to predict the development of ARDS and risk-stratify patients with the disease remains limited. Biomarkers may help to identify patients at the highest risk of developing ARDS, assess response to therapy, predict outcome, and optimize enrollment in clinical trials. After a short description of ARDS pathobiology, here, we review the scientific evidence that supports the value of various ARDS biomarkers with regard to their major biological roles in ARDS-associated lung injury and/or repair. Ongoing research aims at identifying and characterizing novel biomarkers, in order to highlight relevant mechanistic explorations of lung injury and repair, and to ultimately develop innovative therapeutic approaches for ARDS patients. This review will focus on the pathophysiologic, diagnostic, and therapeutic implications of biomarkers in ARDS and on their utility to ultimately improve patient care.
This content is subject to copyright. Terms and conditions apply.
Review Article
A Pathophysiologic Approach to Biomarkers in Acute
Respiratory Distress Syndrome
Raiko Blondonnet,1,2 Jean-Michel Constantin,1,2
Vincent Sapin,2,3 and Matthieu Jabaudon1,2
1CHU Clermont-Ferrand, Intensive Care Unit, Department of Perioperative Medicine, Estaing University Hospital,
63000 Clermont-Ferrand, France
2Clermont Universit´
e, Universit´
e d’Auvergne, EA 7281, R2D2, 63000 Clermont-Ferrand, France
3Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
Correspondence should be addressed to Matthieu Jabaudon; mjabaudon@chu-clermontferrand.fr
Received  December ; Accepted  January 
Academic Editor: George Perry
Copyright ©  Raiko Blondonnet et al. is is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.
Acute respiratory distress syndrome (ARDS) is an acute-onset hypoxic condition with radiographic bilateral lung inltration. It is
characterized by an acute exudative phase combining diuse alveolar damage and lung edema followed by a later broproliferative
phase. Despite an improved understanding of ARDS pathobiology, our ability to predict the development of ARDS and risk-stratify
patients with the disease remains limited. Biomarkers may help to identify patients at the highest risk of developing ARDS, assess
response to therapy, predict outcome, and optimize enrollment in clinical trials. Aer a short description of ARDS pathobiology,
here, we review the scientic evidence that supports the value of various ARDS biomarkers with regard to their majorbiological roles
in ARDS-associated lung injury and/or repair. Ongoing research aims at identifying and characterizing novel biomarkers, in order
to highlight relevant mechanistic explorations of lung injury and repair, and to ultimately develop innovative therapeutic approaches
for ARDS patients. is review will focus on the pathophysiologic, diagnostic, and therapeutic implications of biomarkers in ARDS
and on their utility to ultimately improve patient care.
1. Introduction
e acute respiratory distress syndrome (ARDS) is a het-
erogeneous syndrome dened by the association of bilateral
radiographic pulmonary opacities, arterial hypoxemia (par-
tial pressure of arterial oxygen (PaO2) to fraction of inspired
oxygen (FiO2)ratio< with a positive end-expiratory
pressure of  cm H2O or more), and exclusion of cardiac
failure as a primary cause []. It is characterized by diuse
alveolar epithelial and lung endothelial injury leading to
increased permeability pulmonary edema and alveolar lling
[]. By denition, ARDS occurs within one week of a known
clinical insult or new or worsening respiratory symptoms, as
a consequence of various risk factors including either direct
(e.g., bacterial or viral pneumonia, gastric aspiration, lung
contusion, toxic inhalation, and near drowning) or indirect
(e.g., sepsis, pancreatitis, severe trauma, massive blood trans-
fusion, and burn) lung injury []. Despite improvements in
intensive care during the last een years, ARDS is still
a frequent (/ inhabitants/year), morbid, and life-
threatening condition, with a mortality rate around % [–
]. In addition, there has been recent recognition of the
clinical and biological heterogeneity within ARDS [–],
thus reecting our incomplete understanding of the biology
of ARDS and hampering the successful clinical translation
of new diagnostic, preventive, and therapeutic strategies
[]. Some investigators have further proposed subdividing
ARDS, for example, on the basis of clinical risk factors
[], by direct versus indirect lung injury [], or by focal
versus nonfocal lung morphology as assessed by CT-scan
[, ]. Characterizing ARDS phenotypes may help to better
understand genetic, genomic, and protein risk factors for
Hindawi Publishing Corporation
Disease Markers
Volume 2016, Article ID 3501373, 20 pages
http://dx.doi.org/10.1155/2016/3501373
Disease Markers
ARDS, predict the syndrome, identify mechanism-dened
subgroups of ARDS, and/or to better target therapy [, ].
e subtype (or phenotype) of a condition is ideally dened
by a distinct functional/pathobiological mechanism, named
endotype, that may explain, at least in part, response to
treatment [].
2. Pathogenesis of ARDS
e pathogenesis of ARDS is characterized by two phases
that may sometimes overlap temporally and spatially []:
exudative and proliferative [] phases. An alveolar-capillary
barrier dysfunction resulting in altered permeability of
epithelial and endothelial alveolar cells characterizes the early
exudative phase. Due to loss of cellular integrity, alveoli
are lled with proteinaceous edema uid that results in
impaired gas exchange. Initially, there is an early exudative
phaseassociatedwithdiusealveolardamage,microvascular
injury with subsequent pulmonary edema, alveolar type 
(AT) epithelial cell necrosis, and inux of inammatory cells
which then release active mediators []. During this early
phase, alveolar inammation is mainly mediated by polymor-
phonuclear neutrophils (PMN) [], but recent ndings also
support a key role for monocytes and macrophages [, ].
Other proinammatory mechanisms are also involved, as
the signicant release of proinammatory cytokines by lungs
cells, inammatory cells, and broblasts.
e association of persistent injury and failure to repair
lung damage in a timely manner mainly contributes to the
pathological broproliferative response during which there
are proliferation of broblasts, hyperplasia of AT cells, and
lung repair. e repair of the injured alveolar epithelium
remains incompletely understood; it involves hyperplasia of
AT ( and may be AT) cel ls, mig r ation along t he basem ent
membrane by AT cells to form a new epithelial barrier, and
complex interactions with ECM and other cells including
alveolar macrophages. In the absence of recovery, processes
leading to brosing alveolitis may occur during a brotic
phase, resulting in some cases in marked changes in lung
structure and function [].
3. Biomarkers of ARDS:
A Pathophysiologic Approach
e discovery and validation of biomarkers of myocardial
injury and ventricular overload such as troponin and brain-
natriuretic peptide (BNP) have transformed the diagnosis,
management, and design of clinical trials in conditions such
as myocardial infarction and congestive heart failure [, ].
In a similar way, identication of plasma biomarkers that may
facilitate diagnosis of ARDS could, at least in theory, improve
clinical care, enhance our understanding of pathophysiology,
and be used to enroll more homogeneous groups of patients
in clinical trials of new therapies, increasing the likelihood of
detecting a treatment eect []. Pathophysiologic changes
canprobablybeusedasaframeworktobetterunderstand
various biomarkers that have been studied in ARDS, includ-
ing the cellular injury pathways that are central to lung injury:
endothelial injury, epithelial injury, proinammatory injury,
coagulation, brosis, and apoptosis [].
Among multiple potential applications, biomarkers may
be used to better identify patients with risk factors of ARDS
who are most likely to develop the syndrome. Subsequently,
they may also be useful to improve risk stratication once
ARDS criteria are present. Biomarkers may also play a
pivotalroleinthedesignoffutureclinicaltrialsthrough
the identication of patients at high risk of poor outcome,
thus decreasing the required sample size needed to show
a therapeutic benet []. More recently, biomarkers have
also been proven useful to evaluate the response to therapy
[, ]. Finally, the study of biomarkers in ARDS plays
a fundamental role in understanding the mechanisms and
pathophysiologyunderlyinglunginjury,thusservingasa
solid basis to develop future therapeutic strategies [].
Wewillnowreviewthebiomarkersthathavebeen
investigated in ARDS, with a focus on biomarkers in groups
that reect their primary function (Figure  and Table ).
3.1. Exudative Phase of ARDS. ARDS is characterized by
an initial exudative phase with diuse alveolar damage
associated with the formation of lung inammatory edema.
Alveolar injury is predominant during this phase, and various
proteins that are specic to lung injury are therefore released
in both the blood and the alveolar compartment, thus serving
as markers of the disease or of its resolution.
3.1.1. Lung Injury
(1) Alveolar Epithelium.ealveolarepithelialliningis
compos e d of AT an d AT  epit helia l c ells an d play s a critic a l
role in barrier function, regulating surfactant production,
and in vectorial transport of alveolar uid. During the acute
phase of ARDS, alveolar epithelial cells undergo neutrophil-
mediated damage and eects of proinammatory cytokines
or of hypoxic injury, thus accounting for the clinical syn-
drome []. Lung-secreted proteins may be found in both the
bronchoalveolar (BAL) uid and the systemic blood because
they can move passively across the epithelial barrier into
serum where they may serve as peripheral indicators of
epithelial damage. Several markers of lung epithelial damage
havebeenstudiedasmarkersofARDS,assupportedbythe
fact that they should be more specic to lung injury than
other markers, for example, inammatory cytokines [].
(a) Alveolar Type 1 Cells.eyareoftwotypes.
RAGE. AT epithelial cells cover –% of the alveolar sur-
faceandcontributetobothalveolaruidclearance(AFC)and
barrier integrity. e receptor for advanced glycation end-
products (RAGE) is a transmembrane pattern-recognition
receptor of the immunoglobulin superfamily that is constitu-
tively expressed at low levels in all cells but abundantly in the
lung. RAGE is primarily located on the basal surface of AT
cells [, ]. Activation of RAGE modulates cell signaling,
culminating in a sustained inammatory response through
various intracellular signaling pathways such as cytokines,
reactive oxygen species (ROS), or proteases and leading to
Disease Markers
Exudative
Epithelium damage
Endothelium damage
Inammatory cascade
IL-6, and IL-8
IL-10
Additional markers: HMGB1, LBP, NO, CRP,
Anti-inammatory: IL-1RA, sTNF-RI/II, and
albumin, and LDH
Pulmonary vascular permeability
EF/PL ratio
Lung matrix alteration
Laminin
Elastin/desmosine
MMPs: 2, 3, 8, 9, and 13
PA- 1
Protein C
Coagulation and brinolysis
phase
ATI cells: sRAGE, HTI56
ATII cells: surfactant, KL-6
Club cells: CC16
Ang-1, Ang-2
ICAM-1
Selectins
VEGF
vWF
Epithelial apoptosis
Fibroproliferative phase
Fas/FasL
VEGF
KGF
HGF
N-PCP-III
Epithelial proliferation
Fibroblast proliferation
Endothelial proliferation
Proinflammatory: IL1-𝛽/TNF-𝛼, IL-18,
rombomodulin
Tissue factor
Cell-free hemoglobin
Club cell
Alveolar type I cell
Alveolar type II cell
Red cell
Alveolar
macrophage
Neutroph il
Capillary Interstitium
Protein-rich
edema uid
dierentiation of
alveolar type II cells
Alveolus
Necrotic alveolar
type I cell
Proliferation and
m
Fibroblast
F : Biomarkers of acute respiratory distress syndrome organized by pathways and phases of lung injury (le: early exudative phase;
right: broproliferative phase).
proinammatory activation of nuclear transcription factor
NF-𝜅B[,].RAGEisimplicatedinARDSasanimportant
pathway to innate immunity and alveolar inammation []
and when the soluble form (sRAGE, for soluble RAGE) is
assayedinplasmaorpulmonaryedemauid,asamarker
of alveolar injury [–]. Full-length RAGE is a transmem-
branereceptor,butitcanalsobefoundassolubleisoforms,
generally referred to as soluble RAGE (sRAGE, comprising
the extracellular domain of RAGE and produced through the
cleavage of full-length RAGE by matrix metalloproteinases)
[, ] and endogenous secretory RAGE (esRAGE, pro-
duced aer alternative splicing) []. Full-length RAGE and
its isoforms are abundantly and constitutively expressed in
the lungs in normal conditions [, –], and sRAGE
is now considered as a promising novel marker of AT cell
injury and a key mediator of alveolar inammation [, ,
]. It is shown that sRAGE expression appears enhanced
during the early stage of ARDS. Our team, with others, has
recently reported in both ARDS patients and a mouse model
of ARDS that the extent of sRAGE elevation in plasma and
alveolar uid correlates with markers of severity assessed by
PaO2/FiO2, lung injury, and alveolar uid clearance (AFC)
[–, ]. A role for RAGE pathway in the regulation of
AFC has been recently described for the rst time [] and is
under active investigation by our team and others [, ].
Interestingly, plasma and BAL sRAGE levels are elevated
during ARDS, independently of any associated severe sepsis
[]. In addition, plasma levels of sRAGE are correlated with
diuse damage as assessed by lung CT-scan and are correlated
with the extent of alveolar damage [, ], suggesting that
sRAGE may serve as a useful biomarker of AT cell injury and
lung damage during ARDS. Plasma levels of sRAGE are also
associated with -day and -day mortality in patients with
ARDS [, , ].
Calfee et al. recently compared biomarker levels in
patients with direct versus indirect ARDS enrolled in a single
center study of  patients and in a secondary analysis of 
ARDS patients drawn from a multicenter randomized con-
trolled trial []: levels of biomarkers of lung epithelial injury
(sRAGE, surfactant protein-D) were signicantly higher in
direct ARDS compared to indirect ARDS.
A recent observational study also supports an ARDS
phenotype based on levels of RAGE ligands and soluble
forms, as elevated sRAGE, high mobility group box- pro-
tein (HMGB), and SA, with decreased esRAGE and
advanced glycation end-products (AGEs), were found to
distinguish patients with ARDS from those without [].
Although these recent ndings warrant further validation in
multicenter studies, monitoring sRAGE levels may be useful
in assessing the response to strategies in ventilator settings
including alveolar recruitment maneuvers in patients with
ARDS [], or in patients without lung injury at risk of post-
operative respiratory complications aer major surgery [].
e predictive value of  single-nucleotide polymorphisms in
RAGE gene (AGER) in the development of ARDS in at-risk
patients is currently under study by our team [].
Disease Markers
T : Biomarkers of acute respiratory distress syndrome organized by phases and pathways of lung injury.
Pathophysiologic feature of ARDS Biomarker References
Exudative phase
Epithelium damage
(i) Alveolar type  cells RAGE [–]
HTI56 [, ]
(ii) Alveolar type  cells Surfactant [, ]
KL- [, ]
(iii) Clara cells CC [, ]
Endothelium damage
Ang-, Ang- [, ]
ICAM- [, –]
Selectins [, ]
VEGF [, ]
vWF [–]
Lung matrix alteration
Laminin [, ]
Elastin/desmosine [, ]
MMPs [–]
Inammatory cascade
(i) Proinammatory
IL-𝛽/TNF-𝛼[, –, ]
IL- [, ]
IL- [–]
IL- [–]
(ii) Anti-inammatory
IL-RA [, ]
sTNF-RI/sTNF-RII [, , ]
IL- [–]
(iii) Additional markers
HMGB [, ]
LBP [, ]
NO [, , ]
CRP []
Albumin []
LDH []
Coagulation and brinolysis
PA- [, , –]
Protein C [, , ]
rombomodulin [, ]
Tissue factor [–]
Cell-free hemoglobin [, , , ]
Pulmonary vascular permeability
EF/PL ratio [, ]
Fibroproliferative phase
Endothelial proliferation
VEGF [, –]
Epithelial proliferation
KGF []
HGF [, , , ]
Epithelial apoptosis
Fas/FasL [, –]
Fibroblast proliferation
N-PCP-III [, , , , ]
Disease Markers
HTI56 . Human type I cell-specic membrane protein (HTI56)
is a -kDa glycosylated lung protein specic to the apical
membrane of human AT cells. HTI56 has biochemical char-
acteristics of an integral membrane protein []. Although
the precise functions of HTI56 remain unknown, HTI56 is
an analog to RTI40, a – kDa integral membrane protein
specic to the apical membrane of rat AT cells []. Patients
with ARDS had higher levels of HTI56 in both lung edema
uid and plasma as compared to patients with hydrostatic
lung edema [], but no study assessing the association
between HTI56 levels and other endpoints in patients with
ARDS (e.g., prognosis) has been published to date.
(b) Alveolar Type 2 Cells.ATcellshaveimportanthomeo-
static functions in the lung, including AFC and production
of alveolar surfactant (involved in lung compliance, keeping
thealveolusopen).ATcellsarealsoknownaskeymediators
of the epithelial repair process [].
Surfactant Proteins. Surfactant has a vital role in maintaining
the integrity of the alveolar-capillary interface. Its essential
function is to decrease surface tension into the alveoli, thus
stabilizing lung volume at low transpulmonary pressures.
Surfactant is composed of approximately % phospholipids,
% other lipids (cholesterol, triacylglycerol, and free fatty
acids), and % proteins. Four surfactant-associated proteins
(SP), designated SP-A, SP-B, SP-C, and SP-D, represent
approximately half of proteins composing surfactant. SP-A
and SP-D easily dissociate from lipids and are hydrosoluble.
eybelongtothelunginnateimmunesystem,thereby
enhancing phagocytosis of bacteria and virus. ey also
exert regulatory eects on AT cells. SP-B and SP-C are
small, extremely hydrophobic proteins that are important in
the formation of the surfactant monolayer in the terminal
airspaces and in the reduction of surface tension, thus
preventing end-expiratory alveolar collapse [].
Early observations in ARDS revealed a loss in surface
tension suggesting a functional loss of the surfactant proteins
[]. In a rst case report of three patients, the ratio
of plasma SP-B/SP-A was inversely associated with both
blood oxygenation and static respiratory system compliance,
suggesting that SP-B breaches the alveolocapillary barrier
more readily than SP-A and may therefore provide a more
sensitive marker of lung injury []. Plasma levels of SP-A
and SP-B are increased in patients with ARDS [] and in at-
risk patients [, ], whereas lower SP-A and SP-B levels
were found in the BAL uid of patients at risk for ARDS
prior to the onset of the clinical syndrome. SP-A and SP-B
levels remained low for as long as  days in patients with
sustained ARDS. Interestingly, this decrease in BAL SP-A and
SP-B does not result simply from dilution of alveolar uids by
plasma entering the alveolar spaces, as SP-D levels remained
stable in parallel []. In a cohort of  patients, reduced
pulmonary edema uid SP-D and elevated plasma SP-A at
the onset of ARDS were associated with poor prognosis [].
Nevertheless, in a study of  patients from the ARDSNet
trial of low versus high end-expiratory pressure in ARDS
(ALVEOLI) as well as in  patients enrolled in a randomized
trial of activated protein C for ARDS, plasma SP-D was not
associated with -day mortality or ventilator-free days [].
KL-6. Krebs von den Lungen- (KL-) is a human MUC
mucin that belongs to the high-molecular-weight glyco-
protein family. Aer the cleavage of S-S bond, KL- can
spread into the pulmonary epithelial lining uid. In the
normal lung, this glycoprotein can be predominantly found
in AT cells, and its expression is enhanced during AT
proliferation, regeneration, or injury, thus representing an
attractive biomarker in ARDS. Plasma KL- is elevated in
ARDSpatientsandcorrelateswithlunginjuryandmortality
[, –]. Plasma levels of SP-D and KL- increase over
time in patients with ARDS and may represent biological
markers of ventilator-associated lung injury because their
increase is attenuated by lung-protective ventilation [].
(c) CC16. Clara cell protein (CC) is a . kDa homod-
imeric protein that is abundantly secreted in airways by the
nonciliated bronchiolar Clara cells. Clara cells are devoted
to the protection of the respiratory tract against toxic
inhaled agents, the repair of damaged epithelium, xenobiotics
detoxication, and the secretion of proteins with important
biological activities. CC is highly expressed in the epithelial
lining uid, with antioxidant/inammatory roles, notably by
modulating the production and/or activity of phospholipase-
A, interferon-𝛾, and tumor necrosis factor-𝛼[]. Available
studies found contradictory, inconclusive ndings during
ARDS. Although higher levels of CC are associated with
lung injury and inammation in some experimental and
clinical studies, patients with ARDS had lower plasma and
pulmonaryedemauidlevelsofCCthanpatientswith
acute cardiogenic pulmonary edema, and no correlation was
found between CC and prognosis. So many conicting
ndings do not currently support the association of CC
with the diagnosis or prognosis of ARDS.
(2) Vascular Endothelium.Vascularendothelialinjuryis
characterized by the disruption of cell components leading
to increased microvascular permeability and alveolar edema.
Endothelial injury is mainly driven by the activation of
inammation and coagulation cascades. e activation of
endothelial cells by circulating mediators leads to increased
expression of cell surface molecules that are important
mediators of leukocyte adhesion and contribute to leuko-
cyte accumulation and transmigration []. Activated lym-
phocytes can also release mediators in microvessels that
increase vascular permeability. Along with these leukocyte
signals, inammatory mediators such as tumor necrosis fac-
tor (TNF), thrombin, and vascular endothelial growth factor
(VEGF) disrupt endothelial-cadherin bonds and contribute
to the vascular leak underlying edema formation in ARDS.
Platelets also contribute to endothelial injury through the
releaseofcytokinesandthroughbrinclotting[].
(a) Angiopoietin. Angiogenic agents, along with VEGF and
angiopoietin- (Ang-), play key roles in vascular develop-
ment. VEGF stimulates the generation of new, immature,
Disease Markers
and leaky blood vessels whereas Ang- enhances angiogene-
sis, inducing vascular maturation, and decreases vascular per-
meability []. e most encouraging data result from recent
studies of angiopoietin- (Ang-), an endothelial protein that
has been studied extensively during sepsis []. Ang- has an
important role as it increases endothelial junction instability,
enhances vascular leak, naturally antagonizes Ang-, and, in
the absence of other angiogenic stimuli, induces vascular
regression and endothelial cell apoptosis. Both Ang- and
Ang- are ligands for the tyrosine kinase receptor Tie-
 [], and a link between Ang- and inammation has
been reported []. erefore, such vascular growth factors
have been proposed as biomarkers for ARDS []. First,
two single-nucleotide polymorphisms within the Ang- gene
(rs and rs) were associated with the risk
of developing ARDS in trauma patients []. In addition,
Agrawal et al. found in a prospective study of  patients
admitted to the intensive care unit (ICU) without ARDS
that higher levels of Ang- were signicantly associated
with increased development of ARDS []. In surgical ICU
patients, levels of Ang- were higher in patients with ARDS
than in those without the syndrome []. A higher Ang-
/Ang- ratio was also an independent predictor of mortality
in ARDS patients [, ], and patients with infection-
relatedARDSwhoseAng-levelsincreasedbetweenday
anddaydoubledtheiroddsofdeath,suggestingthatAng-
kinetics may be particularly valuable by reecting evolving
lung injury []. Finally, in a large study of  patients
enrolled in the ARDSNet uid and catheter treatment trial,
baseline plasma levels of Ang- were associated with -day
mortality in patients with noninfectious ARDS, whereas this
association was not found in patients with infection as their
primaryARDSriskfactor.Basedonasecondaryanalysis
of two large studies, Calfee et al. further demonstrated that
indirect lung injury is characterized by a molecular pheno-
type consistent with more severe lung endothelial injury, as
assessed by plasma Ang-, and less severe epithelial injury
[].
(b) ICAM-1. e soluble intercellular adhesion molecule-
 (sICAM-) is an inducible glycoprotein expressed on
the surface of vascular endothelial cells and other cells
(e.g., hematopoietic cells, AT cells) []. Under physiologic
conditions, sICAM- is not constitutively expressed or is
expressed at low levels in most tissues. During inammation,
andinresponsetostimulisuchasinterferon-𝛾(IFN-𝛾)or
interleukin- (IL-), levels of sICAM- are upregulated [].
Elevated levels have also been found in both plasma and
lung edema uid from patients with ARDS, as compared to
patients with hydrostatic lung edema []. In a multicenter
study, the increase of sICAM- from baseline to day 
was associated with poor clinical outcome [], and in a
prospectivecohortofpatientswithARDS,baselineplasma
levelsofsICAM-werealsoassociatedwithmortality[].
In pediatric patients with ARDS, early elevated plasma levels
of sICAM- were associated with increased risks of death and
of prolonged mechanical ventilation []. In trauma patients,
higher plasma levels of sICAM- at baseline were correlated
with future development of multiple organ dysfunction syn-
drome (MODS) but not with the development of ARDS [–
].
(c) Selectins. Selectins are membrane-associated glycopro-
teins that mediate the adhesion of leukocytes and platelets
to vascular surface. L-selectin is mainly expressed by leuko-
cytes. P-selectin is rapidly redistributed from membranous
secretory granules to the surface of activated platelets and
endothelial cells []. E-selectin is expressed by cytokine-
activated endothelial cells. It has been shown that plasma
levels of such soluble adhesion molecules were markedly
higher in nonsurvivors among critically ill patients and that
they were negatively correlated with lung function (e.g.,
PaO2/FiO2ratio) []. Other studies found that plasma P-
selectin was elevated in patients with ARDS, especially in
those who subsequently died, as compared with patients with
other pulmonary diseases or sepsis but without ARDS [].
Interestingly, patients with ARDS with chronic alcohol con-
sumption had elevated levels of soluble E-selectin in both the
plasma and epithelial uid consistent with altered endothe-
lial and alveolar-capillary function []. More recently, E-
selectin was measured in the plasma levels from  individ-
uals admitted to the emergency department and who were
at-risk for developing ARDS, with higher E-selectin levels
beingassociatedwithbothARDSdevelopmentand-day
mortality []. Circulating soluble E-selectin levels were
elevated in pneumonia patients with ARDS, and plasma levels
decreased along with the treatment of pneumonia [].
(d) VEGF. One of the most extensively studied endothelial
markers in ARDS is VEGF, albeit its value as a biomarker
remains unclear. Vascular endothelial growth factors belong
to the platelet-derived growth factor supergene family. ey
play central roles in the regulation of angiogenesis and
lymphangiogenesis []. Alternative splicing of the VEGF
gene (p.) transcript leads to the generation of several
splice variants, or isoforms, with various sizes []. VEGF-A
is a – kDa glycoprotein acting as the major factor impli-
cated in angiogenesis. It binds to two tyrosine kinase (TK)
receptors, named VEGFR- (Flt-) and VEGFR- (KDR/Flk-
), and regulates endothelial cell proliferation, migration, vas-
cular permeability, secretion, and other endothelial functions
[]. e expression of VEGF in ARDS varies, depending
on the degree of epithelial and endothelial damage. Many
lung cells release VEGF, for example, AT cells, neutrophils,
alveolar macrophages, and activated T cells. us, VEGF
is potentially capable of having an eect on both alveolar
epithelial and endothelial barriers. Interestingly, overexpres-
sion of VEGF induces pulmonary edema in animal models
[]. In a single center study, plasma levels of VEGF were
increased in subjects with ARDS, compared to controls, and
elevated plasma VEGF as measured on day  was associated
with mortality in patients with ARDS []. Nevertheless,
several studies suggest that plasma and alveolar VEGF may
help to predict the development of ARDS and its recovery.
WhereasplasmaVEGFisincreasedinARDSpatients,VEGF
levelsweredecreasedintheBALuidfromARDSpatients,
Disease Markers
as compared to controls [, , ]. In order to better
understand such dierences between plasma and alveolar
expression of VEGF, Ware et al. conducted a study with the
aim to determine whether changes in alveolar levels of VEGF
were specic to ARDS or not []: the authors found that
alveolar levels of VEGF were decreased in both patients with
ARDS and those with hydrostatic edema. e mechanisms
implicated in this alveolar decrease in VEGF during ARDS
might not depend on the degree of lung injury but rather on
the degree of alveolar ooding [].
(e) vWF.Earlystudiesofendothelialmarkersfocusedonvon
Willebrand Factor (vWF), a macromolecular antigen that is
produced predominantly by endothelial cells, and to a lesser
extent by platelets. In the setting of endothelial activation
or injury, vWF is released from preformed stocks into the
circulation[,].VWFhasbeeninvestigatedasabiological
marker of endothelial injury in patients both at-risk for ARDS
and with established ARDS [, ]. In a prospective study
of  ICU patients with sepsis, patients with nonpulmonary
sepsis had higher levels of plasma vWF, with good predictive
and prognostic values for ARDS. Indeed, elevated plasma
levels of vWF had a sensitivity of % and a specicity of
% for the prediction of ARDS development in the setting
of nonpulmonary sepsis []. However, subsequent studies
in patients at-risk for ARDS did not conrm these ndings
[, , ]. In another study of  patients with ARDS
enrolled in the National Heart, Lung, and Blood Institute
ARDS Network trial of lower tidal volume, nonsurvivors
hadhigherplasmalevelsofvWF,comparedtosurvivors
[]. Higher vWF levels were signicantly associated with
fewer organ failure-free days, suggesting that the degree of
endothelial activation and injury is strongly associated with
outcomes in ARDS; nevertheless, ventilator settings had no
impact on vWF levels in this study [].
( f) Lung Extracellular Matrix. e extracellular matrix (ECM)
forms the region of the lung situated between the alveo-
lar epithelium and the vascular endothelium. ECM plays
a mechanical role as it supports and maintains tissular
structures. ECM also represents a complex and dynamic
meshwork inuencing many biological cell functions such as
development, proliferation, and migration []. Collagens
are the main component of ECM, along with glycoproteins
and proteoglycans including hyaluronic acid.
Laminin. Laminins (LM) are ECM proteins with high molec-
ular weights that deposit in basal membranes. Laminins
areinvolvedincellprocessessuchascellularadhesion,
growth, and dierentiation []. Laminin- (LM-) plays an
importantroleincellmigrationandintheremodelingof
epithelial tissue. LM- is activated through its cleavage by
matrix metalloproteinases (MMPs), thus releasing a soluble
LN 𝛾 NH-terminal fragment (GF) that does not deposit
in the ECM and can therefore be detected in the peripheral
blood. In a small single center study, laminin was measured in
the plasma and lung edema uid from  patients with ARDS,
with higher levels found in patients with ARDS as compared
to healthy volunteers []. Interestingly, nonsurvivors had
higher plasma levels of laminin, as measured  days aer
ARDS onset, than survivors, and survivors had decreasing
levels of the marker over time, suggesting that its secretion
is suppressed during ARDS recovery.
Elastin/Desmosine. Elastin is another critical protein of the
ECM that gives the lung its elastic recoil ability. In adults,
elastin, which is expressed by lungs and other tissues, is
usually excreted in the urine. During lung epithelial and
endothelial injury, elastin can be broken down by proteases
such as neutrophil elastase []. Elastin breakdown results in
smaller fragments containing desmosine and isodesmosine
[]. In a large study of  patients with ARDS, those
ventilated with lower tidal volumes had lower urine desmo-
sine levels, a nding that may reect reduced extracellular
matrix breakdown; however, no correlation with mortality
was found in patients with ARDS [].
MMPs. Matrix metalloproteinases (MMPs) are zinc-depend-
ent endopeptidases that are able to degrade almost all
extracellular matrix components. MMP-, a member of the
leukocyte-derived MMPs, contributes to the degradation,
turnover, and remodeling of the extracellular matrix digest-
ingtypeIcollagen[,].MMPsaremajoractorsin
almost all phases of the inammatory response, and their
function is highly regulated. At the tissue level, most impor-
tant inhibitors are the tissue inhibitors of metalloproteinases
(TIMPs). In fulminant inammation, the inhibitory capacity
of TIMPs may be overwhelmed, leading to excessive tissue
damage and adverse outcome []. Previous studies suggest
that MMPs may have an important role in ARDS, although
this role may be either harmful or benecial [, ]. In a
recent study, despite MMP- levels did not predict outcome in
ARDS patients, higher levels of TIMP- were independently
associated with increased -day mortality in a large group
of critically ill, mechanically ventilated patients []. ese
ndings are in contradiction to those from a study in
pediatric ARDS patients in which higher MMP- and active
MMP- levels, as measured  hours aer disease onset, were
associated with longer durations of mechanical ventilation
and fewer ventilator-free days []. Elevated MMP-, MMP-
, and MMP- in the BAL uid from ARDS patients were
associated with patterns of acute inammation but with poor
outcome []. Interestingly, MMP- and MMP- may be
protective against lung injury by cleaving transmembrane
receptor RAGE into sRAGE, thus regulating RAGE activation
by its ligands [, ].
3.1.2. Inammatory Cascades. During ARDS, inammatory
responses can either be related to an ongoing primary
infectious stimulus such as pneumonia or to systemic inam-
mation, such as in sepsis or in pancreatitis []. e inam-
matory cascade involves inammatory cells and the release
of inammatory mediators, as driven by a complex network
of cytokines. A comparison between blood and alveolar
cytokines suggests that most inammatory mediators orig-
inate from the lung []. Alarmins, or damage-associated
molecular patterns (DAMPs), are released by dead cells or
local inammatory cells (e.g., alveolar macrophages). ey
Disease Markers
activate and recruit immune cells via binding to dierent
receptors, such as TLR, IL- receptor (IL-R), or RAGE,
thereby initiating and perpetuating multiple proinamma-
tory pathways [, ].
Regulation of the inammatory response is a complex
process that requires interplay between several immune
mediators []. Both pro- and anti-inammatory biomark-
ers have been studied in ARDS.
(1) Proinammatory Cytokines
(a) IL-1𝛽and TNF-𝛼.IL-𝛽and TNF-𝛼are the most biolog-
ically potent cytokines secreted by activated macrophages in
theearlyphaseofARDS.eycausethereleaseofavariety
of proinammatory chemokines such as monocyte chemo-
tactic protein- (MCP-), macrophage inammatory protein-
𝛼(MIP-𝛼), IL-, and IL- with subsequent recruitment
of inammatory cells into the air spaces, alteration of the
endothelial-epithelial barrier permeability, and impairment
of uid transport leading to alveolar edema []. TNF-𝛼also
promotes lung edema indirectly, through the production of
reactive oxygen species (ROS) and a decreased expression
of epithelial sodium (ENaC) and Na+-K+-ATPase channels
[]. Finally, TNF-𝛼,apotentchemoattractantforbrob-
lasts, is a promoter of lung brosis in experimental studies
[, ]. Interestingly, the ratio of BAL to serum levels
of both TNF-𝛼and IL-𝛽is typically high, suggesting that
such cytokines may originate from the lung in the setting
of ARDS []. Persistent elevation of plasma and BAL IL-
𝛽is associated with worse outcome [, ]. Both TNF-
𝛼and IL-𝛽areelevatedintheplasmaandBALuidfrom
patients at risk of and with ARDS [, ] and associated
with mortality [].
(b) IL-18. Inammasomes are intracellular macromolecular
complexes that serve as platforms for the activation of the
proinammatory enzyme caspase-, which in turn cleaves
pro-IL-𝛽and pro-IL- into IL-𝛽and IL- []. ese
inammasome-activated cytokines play central roles in the
propagation of the acute inammatory response. IL- and
caspase-playcriticalrolesinthedevelopmentoflung
injury, and higher levels of IL- are correlated with disease
severity and mortality in patients with ARDS []. Among
 patients with acute respiratory failure, those with ARDS
had signicantly higher serum levels of IL-, and serum IL-
 was signicantly higher in nonsurvivors [].
(c) IL-6. IL- is produced by a wide range of cells including
monocytes/macrophages, endothelial cells, broblasts, and
smooth muscle cells in response to stimulation by endotoxin,
IL-𝛽,andTNF-𝛼[]. IL- is one of the most important
mediators of fever and is critical for B-cell dierentiation and
maturation with secretion of immunoglobulins, cytotoxic T
cell dierentiation, macrophage and monocyte function, and
production of acute phase proteins. Although IL- activates
both proinammatory and anti-inammatory mechanisms,
IL- primarily correlates with a proinammatory prole
during the early phase of ARDS. Plasma IL- increases early
in patients at risk of developing ARDS []. IL- is elevated in
both plasma and BAL uid during ARDS [] and correlates
with mortality [, ].
(d) IL-8. IL- is a proinammatory cytokine with a role in
neutrophil/monocyte chemotaxis and neutrophil apoptosis
inhibition. High plasma and BAL levels of IL- are found early
duringARDSandpredictoutcome[].However,previous
studies did not support such ndings [, ]. In a recent
monocenter study of  patients, only baseline IL- (among
 other biomarkers) was associated with the development of
multiorgan failure, even aer adjustment for other relevant
variables[].Also,severalstudieshaveevaluatedtherole
of the anti-IL- autoantibody/IL- immune complexes in
ARDS, a pathway that could lead to the identication of novel
biomarkers and therapeutic targets [, –].
In a recent study, Calfee et al. used latent class analysis
to integrate both clinical and biological data to identify
two ARDS endotypes in an analysis of , patients from
two ARDSNet trials (ARMA and ALVEOLI) []. A rst
endotype was categorized by more severe inammation, as
assessed by both IL- and IL- levels, and worse clinical
outcomes, whereas a second endotype had less inammation,
lessshock,andbetterclinicaloutcomes.Basedonthedata
from the ALVEOLI trial, a “proinammatory” endotype
was associated with higher mortality and better response to
higher levels of positive end-expiratory pressure [].
(2) Anti-Inammatory Cytokines.einammatoryresponse
is also strongly inuenced by anti-inammatory systems,
including nonspecic (e.g., -macroglobulin, IL-) and
specic (e.g., IL- receptor antagonist (IL-RA) antagonists,
soluble IL- receptor II (sIL-RII), soluble TNF receptor
I (sTNF-RI), and soluble TNF receptor II (sTNF-RII)) of
proinammatory cytokines.
(a) IL-1RA. Circulating IL-RA levels are increased but do
not predict the development of ARDS in at-risk patients
[]. Studies of gene expression in alveolar macrophages
and circulating leukocytes from healthy control subjects and
patients with ARDS revealed that sIL-RII may be valuable as
a biomarker because of increased levels in both the lung and
circulation during ARDS [].
(b) sTNF-RI/sTNF-RII. Soluble TNF-𝛼receptors (sTNF-R)
I and II can bind TNF and compete with its binding to
the cellular receptor, thus reducing its bioavailability. Soluble
TNF-RI and TNF-II are associated with morbidity and
mortality in patients with ARDS [], and a strategy of low
tidal volume ventilation is associated with decreased sTNF-
RI levels []. Trauma-associated ARDS diers clinically and
biologically from ARDS due to other clinical disorders, with
lower levels of sTNF-RI patients with trauma as a primary
cause of ARDS [, ].
(c) IL-10. Interleukin- (IL-) is an anti-inammatory
cytokine that is produced by several cells including B lym-
phocytes, monocytes, and alveolar macrophages []. Aside
from inhibiting the production of IL- and TNF-𝛼,IL-
Disease Markers
upregulates TNF receptors [] and stimulates the produc-
tion of the naturally occurring IL-RA and the releas e of sTNF
receptors []. IL- inhibits the production of proinam-
matory mediators by alveolar macrophages involved during
ARDS []. ARDS patients have lower plasma and BAL
levels of IL- than at-risk patients who did not develop the
syndrome []. Higher baseline IL- levels were associated
with higher morbidity and mortality [].
(3) Additional Markers. Other markers with potential clinical
importance in ARDS-associated inammation have been
identied as putative biomarkers during ARDS.
High mobility group box nuclear protein  (HMGB) is a
DNA nuclear binding protein that is secreted by immune cells
including monocytes and macrophages. HMGB increases
early aer severe trauma and correlates with systemic inam-
matory response and development of ARDS []. Alveolar
andplasmalevelsofHMGB(asmeasuredinthearterialor
central venous blood) are elevated in patients with ARDS and
associated with outcome []. In  patients with ARDS,
plasma levels of HMGB were also higher in nonsurvivors
and correlated with levels of sRAGE [].
Lipopolysaccharide binding protein (LBP) is an acute
phase protein that is correlated with lung inammation
during ARDS []. More recently, it has also been demon-
stratedthatserumlevelsofLBPwerestronglyassociatedwith
increased mortality and the development of ARDS in patients
with severe sepsis [].
Nitric oxide (NO) is a marker of oxidative stress that
has also been investigated as a marker of ARDS. In patients
with persistent ARDS, higher levels of nitric oxide and
of its end-products (e.g., nitrotyrosine) are associated with
mortality []. In contrast, higher urine NO levels were
strongly associated with better clinical outcomes including
mortality and ventilator-free days in patients enrolled in the
ARDSNet low tidal volume trial []. Extracellular citrulline,
theeectiveprecursorofNO,islowerintheplasmafrom
patients with severe sepsis and lower plasma citrulline is
associated with the presence of ARDS []. Mechanisms
involved in the regulation of lung injury by NO-dependent
pathways remain unknown.
Although C-reactive protein (CRP) is widely considered
asamarkerofsystemicinammation,higherlevelsofCRP
areassociatedwithbetteroutcomeamongpatientswith
ARDS []. Nevertheless, a recent study found that albumin
levels, rather than CRP, may help to predict and monitor the
severity and course of ARDS in febrile critically ill patients
with ARDS or at risk for the syndrome []. In the same
study, levels of lactate dehydrogenase (LDH) predicted -
day mortality but were not correlated with severity [].
3.1.3. Coagulation and Fibrinolysis. During early ARDS, acti-
vation of the inammatory cascades results in the activation
of the coagulation system, which in turn can inuence
inammatory responses by aecting the expression of various
cytokines such as IL-, IL-, and IL-. Activation of coagula-
tion pathways induces migration of inammatory cells into
alveoli through the endothelial and epithelial barriers and
generates thrombin formation. In addition, proinammatory
events may also inhibit brinolysis and induce platelet acti-
vation [].
Extravascular brin deposition, when localized predomi-
nantly in the alveolar compartment, is found in several acute
inammatory lung diseases, and enhanced alveolar procoag-
ulant activity is reported in ARDS patients []. Fibrin depo-
sition may be benecial for gas exchange by sealing leakage
sites when lung capillary endothelial and epithelial barriers
are disrupted. Nevertheless, brin alveolar deposition may
be harmful since it can lead to activation of neutrophils
and broblasts, endothelial injury, loss of surfactant activity
favoring alveolar collapse, impaired alveolar uid clearance,
and thrombotic obstruction of the microcirculation [].
(1) PAI-1. e balance between activation of coagulation
and activation of brinolysis is an important determinant
of the amount and duration of brin deposition during
lung injury. Plasminogen activator (PA) and plasminogen
activator inhibitor- (PAI-) regulate brinolysis through the
conversion of plasminogen to plasmin, a brinolytic enzyme.
PA- is a major endogenous inhibitor of PA. Both PA and PA-
are secreted by various cells including macrophages, brob-
lasts, and lung endothelial and epithelial cells []. During
ARDS, alveolar epithelial cells and activated macrophages
overexpress PAI-, thus contributing to decreased alveolar
brinolytic activity. Nevertheless, the value of PAI- as
a biomarker in ARDS remains controversial. PAI- levels
were higher in patients with ARDS than in patients with
hydrostatic lung edema [], and higher PAI- levels are
associated with mortality and higher durations of mechanical
ventilation in patients with ARDS [, ]. In a large Finnish
study, PAI- levels were not correlated with mortality or
development of ARDS in critically ill patients under mechan-
ical ventilation, but low baseline plasma levels of the soluble
urokinase plasminogen activator receptor (suPAR) were pre-
dictiveofsurvival[].Inasecondaryanalysisoftwolarge
randomized controlled trials, PAI- was associated with lung
injury (as dened as decreased oxygenation index) but not
with mortality []. Nevertheless, in another study of patients
from the ARDSNet ARMA study, higher levels of PAI-
were independently associated with higher mortality and
clinical outcomes, including organ failure []. However,
this association between PAI- levels and the development of
multiorgan failure was not conrmed in a recent study of 
ARDS patients [].
(2) Protein C. Protein C system is an important endogenous
regulator of coagulation and brinolysis. Protein C is synthe-
sized by the liver and circulates as an inactive compound.
It is transformed to its active form on cell surface by the
thrombomodulin- (TM-) thrombin complex [, ]. e
endothelial cell protein C receptor (EPCR) is another cell
surfaceproteinthatcanfurtherenhanceproteinactivation
by binding the TM-thrombin complex []. In addition
to suppressing thrombin formation, activated protein C has
anti-inammatory properties such as decreasing the levels
of proinammatory cytokines []. Protein C can improve
endothelial permeability and exert antiapoptotic eects via
 Disease Markers
p pathways []. Activated protein C can also inactivate
PAI-, thus promoting brinolysis []. Plasma protein C
was signicantly lower in patients with ARDS as compared
to controls, and it was associated with worse clinical out-
comes, including higher hospital mortality, shorter duration
of unassisted ventilation, and increased risk of multiple organ
failure []. In a larger cohort of patients with early ARDS,
lowplasmalevelsofproteinCwereagainassociatedwith
mortality and adverse clinical outcomes []. Decreased
levels of protein C during ARDS suggest a link between
hypercoagulability and mortality.
(3) rombomodulin. rombomodulin (TM) is a mul-
tidomain transmembrane-bound glycoprotein found on the
surface of endothelial cell. Its main role is to neutralize the
procoagulant eects of thrombin and accelerate activation
of protein C. In addition to its membrane-bound form, TM
also exists as a circulating soluble isoform in the plasma.
In patients with ARDS, levels of soluble thrombomodulin
(sTM) are higher in the pulmonary edema uid than in
plasma [], suggesting an alveolar source, but no correlation
was found between plasma sTM and the development of
ARDS, yet higher levels of sTM were observed in patients
at high risk for ARDS. In patients with established ARDS,
higher plasma and alveolar levels of sTM were correlated
with severity of illness and multiple organ failure []. In
alargeranalysisofpatients,elevatedlevelsofplasma
sTM were associated with increased mortality thus possibly
reecting an increased degree of inammation and both lung
and systemic endothelial damage [].
(4) Tissue Factor (TF). Tissue factor (TF) is a  kDa trans-
membrane glycoprotein that is the most potent stimulator of
the extrinsic coagulation cascade. TF initiates the coagulation
cascade by binding and allosterically activating coagulation
factor VIIa. e resulting TF-VIIa complex binds the sub-
strate coagulation factor X via multiple interactions along
an extended interface to produce the TF-VIIa-X complex.
is complex leads eventually to thrombin formation and
brin deposition []. Levels of TF in lung edema uid are
higher than plasma levels in patients with ARDS, supporting
a lung origin for TF in this setting, and both plasma and
alveolar levels of TF are higher in ARDS patients as compared
to patients with hydrostatic edema []. Notably, patients
withsepsis-inducedARDSmayhavehigherlevelsofTFas
compared to patients without ARDS [].
(5) Cell-Free Hb. Levels of cell-free hemoglobin (Hb) are
higher in the air space of ARDS patients as compared
to critically ill patients with hydrostatic lung edema [].
Instillation of red blood cells or cell-free hemoglobin causes
lung injury in rats [] and intra-alveolar hemorrhage is
associated with high levels of intra-alveolar cell-free Hb,
more severe lung injury, and increased lipid peroxidation
in the lung from mice with tissue factor deciency [].
Precise cellular and molecular mechanisms by which cell-
free hemoglobin in the air space could mediate or potenti-
ate ARDS are currently under investigation []. Cell-free
Hbcouldactivatechemokinereleasewithinthelung,and
decompartmentalization of hemoglobin is likely to provide a
signicant proinammatory stimulus in the setting of diuse
alveolar damage and hemorrhage during ARDS [].
3.1.4. EF/PL Protein Ratio. e pathophysiology of ARDS
includes disruption of several physical barriers including
endothelial and epithelial cell layers, the basement mem-
brane, and the extracellular matrix, resulting in increased pul-
monary microvascular permeability. e pulmonary edema
uid-to-plasma protein (EF/PL) ratio is a rapid, safe, and
noninvasive measure of alveolar-capillary membrane per-
meability. e EF/PL ratio was rst proposed as a tool to
determine the etiology of acute pulmonary edema []. More
recently,inalargestudyofcriticallyillpatients,Wareetal.
demonstrated that the EF/PL ratio had an excellent discrimi-
native value in distinguishing ARDS from hydrostatic edema
and was strongly associated with clinical outcomes. Using a
cuto of ., the EF/PL ratio had a sensitivity of % and a
specicity of % for the diagnosis of ARDS [].
3.2. Fibroproliferative Phase of ARDS. In some patients,
important and persistent accumulation of macrophages,
brocytes, broblasts, and myobroblasts in the alveolar
compartment leads to excessive deposition of ECM com-
ponents including bronectin and collagen types I and III,
among other proteins. An imbalance between probrotic and
antibrotic mediators may subsequently drive this bropro-
liferative response []. Growth factors play a major role
in the resolution of ARDS []. Lung endothelial repair is
promoted by vascular endothelial growth factor (VEGF).
A variety of growth factors promote repair of the alveo-
lar epithelium including keratinocyte growth factor (KGF),
hepatocyte growth factor (HGF), broblast growth factor
(FGF), and transforming growth factor-𝛼(TGF-𝛼)[].Two
major pathways with opposite eects involve growth factors
during ARDS: tyrosine kinase receptor mediation (e.g., KGF,
HGF, FGF, and VEGF) and serine-threonine kinase receptors
such as TGF-𝛽,whichtendtohaveopposedeectonthe
upregulation that occurs when the tyrosine kinase receptor
pathway is involved [, , ].
3.2.1. Endothelial Proliferation. Novel evidence points to a
potentialroleofVEGFinpromotingrepairofthealveolar-
capillary membrane during recovery from ARDS, and under-
standingtheroleofVEGFinthisdiseaseprocesscould
be crucial for developing new therapeutic strategies [,
]. In the lung, VEGF is produced primarily by epithelial
cells; it increases microvascular permeability [] but has an
important role also during the repair phase by stimulating
endothelial cell proliferation and survival [, ]. e
levels of VEGF are increased in plasma from patients with
ARDS but are decreased in BAL uid, compared to healthy
controls; subsequently, BAL levels of VEGF increase during
the resolution of lung injury [, , ].
3.2.2. Epithelial Proliferation and Apoptosis. KGF, also known
as FGF-, is a potent mitogenic factor for alveolar epithelial
cells that is primarily produced by broblasts and other
Disease Markers 
cellssuchasTlymphocytes.KGFregulatestransepithelial
transport of sodium by stimulating the epithelial channel
Na+-K+-ATPase in alveolar epithelial cells [].
HGF is a nonspecic mitogen secreted by broblasts,
alveolar macrophages, endothelial cells, and epithelial cells.
Several animal and human studies suggest that KGF and HGF
could protect the alveolar space against injury and could
facilitate the repair of alveolar structures aer injury [, ,
].KGFlevelscouldbemeasuredintheBALfrompatients
with ARDS but not in the BAL from those without ARDS; in
addition, BAL KGF was associated with poor prognosis [].
Elevated HGF levels were also associated with outcome [].
In  patients with ARDS and  patients with hydrostatic
lung edema, HGF and KGF were proven biologically active
in the edema uid of patients with ARDS, and higher levels
ofHGFwereassociatedwithmortalityinthesepatients[].
Apoptosisofalveolarepithelialcellsisamajorphe-
nomenon in the initiation and perpetuation of lung injury
[]. e Fas/FasL system plays an important role in the
regulation of cell life and death through its ability to initiate
apoptosis []. is system combines the cell membrane
surface receptor Fas (CD) and its natural ligand FasL
(CDL). Membrane-bound FasL mediates lymphocyte-
dependent cytotoxicity, clonal deletion of alloreactive T cells,
and activation-induced suicide of T cells []. Its soluble
form (sFasL) results from cleavage of membrane FasL by
MMPs and induces apoptosis in susceptible cells [].
Apoptosis is induced when membrane-bound or soluble FasL
binds to Fas-bearing cells. By contrast, apoptosis is inhibited
when soluble Fas binds to either membrane-bound FasL or
sFasL thus preventing FasL from interacting with membrane-
bound Fas receptors []. In patients with ARDS, sFasL was
detectable in the lung before and aer the onset of clinically
dened ARDS, and nonsurvivors had signicantly higher
BAL levels of sFasL on day  as compared with survivors
[]. Both soluble Fas and soluble FasL were associated with
outcome and higher in the lung edema uid from patients
with ARDS, compared to control patients with hydrostatic
pulmonary edema []. Nevertheless, recent ndings sug-
gested limited role for Fas/FasL system and apoptosis in
airway epithelial cell death during ARDS [].
3.2.3. Fibroblast Proliferation. Pulmonary broblasts pro-
duce procollagen III peptide (PCP-III), that is, a precursor
of collagen. e NT part of procollagen III, resulting from
the enzymatic cleavage of procollagen by specic proteases in
the extracellular space, is considered as a marker of collagen
synthesis. Alveolar levels of N-PCP-III are higher in ARDS
patient, as compared with controls []. e elevation of
N-PCP-III in pulmonary edema uid begins within the rst
 h of ARDS, that is, during the acute phase of increased
endothelial and epithelial permeability to protein, suggesting
that brosing alveolitis could begin very early in the course of
clinical ARDS []. In another study, high levels of N-PCP-
III were early predictors of poor outcome [, ]. More
recently, Forel et al. measured alveolar N-PCP-III in patients
with nonresolving ARDS, thus identifying patients who had
developed lung broproliferation []. Unfortunately, it
is still unknown whether N-PCP-III measurements could
be useful in selecting patients who would benet from
glucocorticoid therapy, among others, to reduce the ARDS-
associated lung brosis [].
4. Perspectives
4.1. Combining Biomarkers. Despite advances in the identi-
cation of biomarker candidate and better understanding of
ARDS pathogenesis, no single clinical or biological marker
reliably predicts clinical outcomes in ARDS. e combination
of clinical and biological marker is attractive in order to
improve the sensitivity and/or the specicity of the test,
especially through a recent approach aimed at measuring
 biological markers that reect endothelial and epithelial
injury, inammation, and coagulation: vWF, SP-D, TNF-R,
IL-, IL-, ICAM-, protein C, and PAI- in  patients
enrolled in the the ARDSNet trial of low versus high positive
end-expiratory pressure []. Clinical predictors predicted
mortality with an area under the ROC curve (AUC) of
., whereas a combination of these  biomarkers and the
clinical predictors had an AUC of .. e best performing
biomarkers were the neutrophil chemotactic factor IL- and
SP-D, a product of AT cells [], supporting the concept
that acute inammation and alveolar epithelial injury are
important pathogenetic pathways in human ARDS. More
recently,apanelofbiomarkersoflungepithelialinjury
and inammation (SP-D, sRAGE, IL-, CC, and IL-)
provided excellent discrimination for diagnosis of ARDS
in patients with severe sepsis []. erefore, and beyond
their better diagnostic and prognostic values, the use of such
biomarker panels may be useful for selecting patients for
clinical trials that are designed to reduce lung epithelial injury
[]. Nevertheless, whether a therapeutic strategy based on
biomarker measurements would benet patient outcome has
never been investigated.
4.2. Lung Imaging as an ARDS Biomarker. Studies of lung
imaging during ARDS have revealed that adequate ventilator
settingsmayvaryamongpatientswiththesamesyndrome.
In a large study, gas and tissue distribution in the lungs of
ARDS patients were assessed using computed tomography
(CT) and compared to those of healthy volunteers []. Lung
morphology in ARDS is characterized by marked excess of
lung tissue associated with a major decrease in aerated lung
regions and in functional residual capacity. Some patients
with ARDS exhibit preserved aeration of the upper lobes
despitethepresenceofanoverallexcessoflungtissue(focal
ARDS), as opposed to other patients with more diuse loss
of aeration and excessive lung tissue (“diuse” or “nonfocal”
ARDS) [].
Lung morphology may inuence the response to pos-
itive end-expiratory pressure (PEEP), recruitment maneu-
vers (RM), prone position, and patient outcome [, ].
In a prospective study of nineteen patients with ARDS,
Constantin et al. found that lung morphology at zero end-
expiratory pressure could predict the response to a RM with
continuous positive airway pressure of  cm H2Ofor
 Disease Markers
seconds. Nonfocal morphology was associated with higher
lung recruitability and PaO2/FiO2was signicantly increased
bytheRM[,].Incontrast,patientswithfocallung
morphology were at risk of signicant hyperination during
the RM, with no improvement of arterial oxygenation.
It has also been hypothesized that the eects of PEEP
may depend on lung morphology. Puybasset et al. assessed
the responses to PEEP among patients with focal or nonfocal
ARDS []. e regional distribution of intrapulmonary
gas and lung tissue inuences the eects of PEEP in ARDS
patients: maximal alveolar recruitment, without evidence
of overdistension, was observed in patients with nonfocal
ARDS. Nevertheless, PEEP induced mild alveolar recruit-
ment in patients with focal ARDS, along with overdistension
of previously aerated lung regions.
Interestingly, phenotyping patients with ARDS based
on their lung morphology might be possible by measuring
plasma sRAGE with commercially available kits, even though
these ndings need further validation [, ]. However,
RAGE pathway is a promising candidate for subphenotyping
patients with ARDS, as it is believed to play a major role
in the mechanisms leading to AFC and their regulation
[]. Recent ndings that support a relationship between
impairedAFCandlungmorphologymaythereforellagap
in the full recognition of an ARDS phenotype based on lung
morphologythatcouldbelinkedtoanendotypeofimpaired
AFC and activated RAGE pathway [, , , , , ].
4.3. Biomarkers in ARDS: Can ey Improve Patient Care?
Biomarkers are broadly used in critically ill patients,
especially during inammatory and/or infectious diseases.
Biomarkers have been commonly dened as characteristics
that are objectively measured and evaluated as indicators
of normal biological processes, pathogenic processes, or
pharmacologic responses to therapeutic interventions [,
].Biomarkers provide a powerful approach to understand
a disease with multiple applications in observational and
analytic epidemiology, randomized clinical trials, screening,
and diagnosis or prognosis [].
Nevertheless, there are important technical attributes for
a relevant biomarker. First, the marker must be present in
peripheral body tissue and/or uid (e.g., blood, urine, saliva,
breath, or cerebrospinal uid); second, it must be easy to
detect or quantify in assays that are both aordable and
robust; and, third, its regulation should be associated as
specically as possible with damage of a particular tissue,
preferably in a quantiable manner. Prior to the widespread
use of a marker of interest, it is essential that validation and
conrmation of candidate biomarkers by robust statistical
methods are performed during biomarker discovery [].
Sensitivity and specicity are common quality parameters
for biomarkers. Sensitivity describes the probability of a
positive test in cases and specicity describes probability of
negative test in controls. An association between sensitivity
and specicity is represented in the ROC curve by graphing
sensitivity versus  specicity. Area under the ROC curve
(AUROC) is therefore a measure of performance of a marker.
ere is no absolute cuto value of AUROC for robustness of
a marker, but a minimum of . is required and values greater
than . are good particularly in a heterogeneous critically ill
patient population [, ].
To summarize, an ideal biomarker should indicate a
clear relationship with the pathophysiologic event, needs
to be reliable, reproducible, disease specic, and sensitive,
and should be sampled by simple methods and relatively
inexpensive, with little or no diurnal variation. During
ARDS, no single marker has been validated with all these
criteria to date, yet we believe that sRAGE may fulll all
prerequisitesofabiomarkerofARDS.First,plasmasRAGE
has good diagnostic and prognostic values [, , ,
, ]. Second, it is very well correlated with lung injury
severity and specic pathophysiologic features of ARDS, for
example, alveolar uid clearance and lung morphology [,
–, , ]. Previous studies of the predictive value of
early levels of sRAGE for the development of ARDS in a
general population of patients admitted to the emergency
department were negative [], but current research focuses
on both the kinetics of sRAGE and esRAGE and RAGE gene
polymorphisms as predictors of the development of ARDS
in at-risk critically ill patients []. Finally, there is some
evidence suggesting that monitoring sRAGE could inform,
at least partially, on therapeutic responses in patients with or
without ARDS [, , ]. If these data are conrmed by
future studies, such ndings would denitely help to reinforce
sRAGE a real biomarker of ARDS.
5. Conclusion
Biomarker research provides an important translational link
to our understanding of lung pathobiology. rough the
identication and testing of candidate biomarkers, we have
gained insight into the pathogenic importance of endothelial
and epithelial injury and have started to unravel the complex
pathways that contribute to endothelial and epithelial cell
dysfunction, inammation, brosis, and apoptosis in ARDS.
In addition, biomarker studies may help us to explore the
cellular and molecular mechanisms of various therapeutic
strategies for ARDS, and to better understand the potential
proinammatory eects of mechanical ventilation. Given
the clinical heterogeneity of patients with ARDS and the
complexity of the underlying pathobiology, it is unlikely that
a single biomarker will emerge for ARDS, as cardiac-specic
troponin did for myocardial infarction, but the develop-
ment of small biomarker panels reecting each important
lung injury pathway would provide valuable predictive and
prognostic information for both clinicians and investigators.
While biomarkers are currently not recommended for use
in clinical practice in ARDS, biomarker discovery may
hold signicant promise in order to develop and apply
targeted therapies, and to identify candidates for enrollment
in patient-tailored clinical trials of novel therapies for ARDS.
Disclosure
e funders had no inuence on the study design, conduct,
andanalysisoronthepreparationofthispaper.
Disease Markers 
Conflict of Interests
No conict of interests, other sources of nancial support,
corporate involvement, patent holdings, and so forth are to
be declared for all authors.
Authors’ Contribution
Raiko Blondonnet was involved in the conception and design
of the review, in writing the paper, and in its revision prior
to submission. Jean-Michel Constantin was involved in the
conception and design of the review, in writing the paper,
and in its revision prior to submission. Vincent Sapin was
involved in the conception and design of the review, in
writing the paper, and in its revision prior to submission.
Matthieu Jabaudon takes responsibility for the content of
the paper and was involved in the conception and design of
the review, in writing the paper, and in its revision prior to
submission.
Acknowledgments
is work was supported by grants from the Auvergne
Regional Council (“Programme Nouveau Chercheur de la
R´
egion Auvergne”),thefrenchAgence Nationale de la
Recherche, and the Direction G´
en´
eraledelOredeSoins
(“Programme de Recherche Translationnelle en Sant´
e”ANR-
-PRTS-).
References
[] V.M.Ranieri,G.D.Rubenfeld,P.T.ompsonetal.,“Acuteres-
piratory distress syndrome: the Berlin denition,e Journal
of the American Medical Association,vol.,no.,pp.
, .
[] L.B.WareandM.A.Matthay,“eacuterespiratorydistress
syndrome,e New England Journal of Medicine,vol.,no.
, pp. –, .
[] C. Brun-Buisson, C. Minelli, G. Bertolini et al., “Epidemiology
and outcome of acute lung injury in European intensive care
units,IntensiveCareMedicine,vol.,no.,pp.,.
[] G.D.Rubenfeld,E.Caldwell,E.Peabodyetal.,“Incidenceand
outcomes of acute lung injury,e New England Journal of
Medicine,vol.,no.,pp.,.
[]D.W.Dowdy,M.P.Eid,C.R.Dennisonetal.,“Qualityof
life aer acute respiratory distress syndrome: a meta-analysis,
Intensive Care Medicine, vol. , no. , pp. –, .
[] C.S.Calfee,M.D.Eisner,L.B.Wareetal.,“Trauma-associated
lung injury diers clinically and biologically from acute lung
injury due to other clinical disorders,Critical Care Medicine,
vol.,no.,pp.,.
[] C.S.Calfee,D.R.Janz,G.R.Bernardetal.,Distinctmolecular
phenotypes of direct versus indirect ARDS in single and multi-
center studies,Chest,vol.,no.,pp.,.
[] P. Tejera, N. J. Meyer, F. Chen et al., “Distinct and replicable
genetic risk factors for acute respiratory distress syndrome
of pulmonary or extrapulmonary origin,Journal of Medical
Genetics,vol.,no.,pp.,.
[]M.A.Matthay,G.A.Zimmerman,C.Esmonetal.,“Future
research directions in acute lung injury: summary of a National
Heart, Lung, and Blood Institute Working Group,American
Journal of Respiratory and Critical Care Medicine,vol.,no.
, pp.  – ,   .
[] J. P. Reilly, S. Bellamy, M. G. S. Shashaty et al., “Heterogeneous
phenotypes of acute respiratory distress syndrome aer major
trauma,Annals of the American oracic Society,vol.,no.,
pp. –, .
[] L. Puybasset, P. Cluzel, N. Chao et al., “A computed tomography
scan assessment of regional lung volume in acute lung injury,
American Journal of Respiratory and Critical Care Medicine,vol.
, no. , pp. –, .
[] J.-M. Constantin, S. Grasso, G. Chanques et al., “Lung morphol-
ogy predicts response to recruitment maneuver in patients with
acute respiratory distress syndrome,Critical Care Medicine,
vol. , no. , pp. –, .
[] C.S.Calfee,K.Delucchi,P.E.Parsons,B.T.ompson,L.B.
Ware, and M. A. Matthay, “Subphenotypes in acute respiratory
distress syndrome: latent class analysis of data from two
randomised controlled trials, e Lancet Respiratory Medicine,
vol. , no. , pp. –, .
[] R.Marshall,G.Bellingan,andG.Laurent,“eacuterespira-
tory distress syndrome: brosis in the fast lane,orax,vol.,
no.,pp.,.
[]R.B.Henderson,J.A.R.Hobbs,M.Mathies,andN.Hogg,
“Rapid recruitment of inammatory monocytes is independent
of neutrophil migration,” Blood,vol.,no.,pp.,
.
[] H. M. Marriott and D. H. Dockrell, “e role of the macrophage
in lung disease mediated by bacteria,Experimental Lung
Research,vol.,no.,pp.,.
[] L. J. M. Cross and M. A. Matthay, “Biomarkers in acute lung
injury: insights into the pathogenesis of acute lung injury,
Critical Care Clinics,vol.,no.,pp.,.
[] H. K. Gaggin and J. L. Januzzi, “Biomarkers and diagnostics in
heart failure,” Biochimica et Biophysica Acta (BBA)—Molecular
Basis of Disease, vol. , no. , pp. –, .
[] E. Christenson and R. H. Christenson, “e role of cardiac
biomarkers in the diagnosis and management of patients
presenting with suspected acute coronary syndrome,Annals of
Laboratory Medicine,vol.,no.,pp.,.
[] L. B. Ware, T. Koyama, Z. Zhao et al., “Biomarkers of lung
epithelial injury and inammation distinguish severe sepsis
patients with acute respiratory distress syndrome,Critical Care,
vol. , no. , article R, .
[] M. A. Matthay, L. B. Ware, and G. A. Zimmerman, “e
acute respiratory distress syndrome,e Journal of Clinical
Investigation,vol.,no.,pp.,.
[] M. Shirasawa, N. Fujiwara, S. Hirabayashi et al., “Receptor for
advanced glycation end-products is a marker of type I lung
alveolar cells,Genes to Cells,vol.,no.,pp.,.
[] M. Neeper, A. M. Schmidt, J. Brett et al., “Cloning and
expression of a cell surface receptor for advanced glycosylation
end products of proteins,” e Journal of Biological Chemistry,
vol. , no. , pp. –, .
[] M. Jabaudon, E. Futier, L. Roszyk, V. Sapin, B. Pereira, and
J. Constantin, “Association between intraoperative ventilator
settings and plasma levels of soluble receptor for advanced
glycation end-products in patients without pre-existing lung
injury,Respirology, vol. , no. , pp. –, .
[] L. G. Dobbs, R. F. Gonzalez, L. Allen, and D. K. Froh, “HTI,
an integral membrane protein specic to human alveolar type
 Disease Markers
Icells,Journal of Histochemistry & Cytochemistry,vol.,pp.
–, .
[] L. G. Dobbs, M. C. Williams, and R. Gonzalez, “Monoclonal
antibodies specic to apical surfaces of rat alveolar type I cells
bind to surfaces of cultured, but not freshly isolated, type II
cells,Molecular Cell Research, vol. , no. , pp. –, .
[] I. W. Cheng, L. B. Ware, K. E. Greene, T. J. Nuckton, M. D.
Eisner, and M. A. Matthay, “Prognostic value of surfactant
proteins A and D in patients with acute lung injury,Critical
Care Medicine,vol.,no.,pp.,.
[] A. Agrawal, H. Zhuo, S. Brady et al., “Pathogenetic and
predictive value of biomarkers in patients with ALI and lower
severity of illness: results from two clinical trials,American
Journal of Physiology—Lung Cellular and Molecular Physiology,
vol. , no. , pp. L–L, .
[] H. Sato, M. E. J. Callister, S. Mumby et al., “KL- levels are
elevated in plasma from patients with acute respiratory distress
syndrome,European Respiratory Journal,vol.,no.,pp.
, .
[] R. M. Determann, A. A. N. M. Royakkers, J. J. Haitsma et al.,
“Plasma levels of surfactant protein D and KL- for evaluation
of lung injury in critically ill mechanically ventilated patients,
BMC Pulmonary Medicine,vol.,article,.
[] F. Broeckaert and A. Bernard, “Clara cell secretory protein
(CC): characteristics and perspectives as lung peripheral
biomarker,Clinical and Experimental Allergy,vol.,no.,pp.
–, .
[] R. Cartin-Ceba, R. D. Hubmayr, R. Qin et al., “Predictive value
of plasma biomarkers for mortality and organ failure devel-
opment in patients with acute respiratory distress syndrome,
Journal of Critical Care,vol.,no.,pp..e.e,.
[] L. Eklund and P. Saharinen, “Angiopoietin signaling in the
vasculature,ExperimentalCell Research,vol.,no.,pp.
, .
[] C. S. Calfee, D. Gallagher, J. Abbott, B. T. ompson, and
M. A. Matthay, “Plasma angiopoietin- in clinical acute lung
injury: prognostic and pathogenetic signicance,Critical Care
Medicine,vol.,no.,pp.,.
[] M. L. Dustin, R. Rothlein, A. K. Bhan, C. A. Dinarello, and T.
A. Springer, “Induction by IL  and interferon-gamma: tissue
distribution, biochemistry, and function of a natural adherence
molecule (ICAM-),e Journal of Immunology,vol.,no.,
pp.,.
[] R. P. McEver, “Selectins: lectins that initiate cell adhesion under
ow,Current Opinion in Cell Biology,vol.,no.,pp.,
.
[] D. Osaka, Y. Shibata, K. Kanouchi et al., “Soluble endothelial
selectin in acute lung injury complicated by severe pneumonia,
International Journal of Medical Sciences,vol.,no.,pp.
, .
[] M. Shibuya, “Vascular endothelial growth factor and its receptor
system: physiological functions in angiogenesis and pathologi-
cal roles in various diseases,JournalofBiochemistry,vol.,
no. , pp. –, .
[] L.B.Ware,R.J.Kaner,R.G.Crystaletal.,“VEGFlevelsinthe
alveolar compartment do not distinguish between ARDS and
hydrostatic pulmonary oedama,European Respiratory Journal,
vol.,no.,pp.,.
[] L. B. Ware, M. D. Eisner, B. T. ompson, P. E. Parsons, and M.
A. Matthay, “Signicance of von Willebrand factor in septic and
nonseptic patients with acute lung injury,American Journal of
Respiratory and Critical Care Medicine,vol.,no.,pp.
, .
[] A. J. Reininger, “Function of von Willebrand factor in
haemostasis and thrombosis,Haemophilia,vol.,supplement
, pp. –, .
[] M. S. Bajaj and S. M. Tricomi, “Plasma levels of the three
endothelial-specic proteins von Willebrand factor, tissue fac-
tor pathway inhibitor, and thrombomodulin do not predict the
development of acute respiratory distress syndrome,Intensive
Care Medicine,vol.,no.,pp.,.
[] J. Tzu and M. P. Marinkovich, “Bridging structure with function:
structural, regulatory, and developmental role of laminins,
International Journal of Biochemistry and Cell Biology,vol.,
no. , pp. –, .
[] K. Torii, K.-I. Iida, Y. Miyazaki et al., “Higher concentrations
of matrix metalloproteinases in bronchoalveolar lavage uid of
patients with adult respiratory distress syndrome,American
Journal of Respiratory and Critical Care Medicine,vol.,no.
,pp.,.
[] E. G. Cleary and M. A. Gibson, “Elastic tissue, elastin and
elastin associated microbrils,” in Extracellular Matrix,vol.,
pp. –, Harwood Academic Publishers, Amsterdam, e
Netherlands, .
[] D. E. McClintock, B. Starcher, M. D. Eisner et al., “Higher urine
desmosine levels are associated with mortality in patients with
acute lung injury,e American Journal of Physiology—Lung
Cellular and Molec ular Physiology,vol.,no.,pp.LL,
.
[] J. H¨
astbacka, R. Linko, T. Tervahartiala et al., “Serum MMP-
 and TIMP- in critically ill patients with acute respiratory
failure: TIMP- is associated with increased -day mortality,
Anesthesia & Analgesia, vol. , no. , pp. –, .
[] A. H. Hergrueter, K. Nguyen, and C. A. Owen, “Matrix met-
alloproteinases: all the RAGE in the acute respiratory distress
syndrome,e American Journal of Physiology—Lung Cellular
and Molecular Physiology,vol.,no.,pp.LL,.
[] N. Yamakawa, T. Uchida, M. A. Matthay, and K. Makita,
“Proteolytic release of the receptor for advanced glycation end
products from in vitro and in situ alveolar epithelial cells,e
American Journal of Physiology— Lung Cellular and Molecular
Physiology,vol.,no.,pp.LL,.
[] T. Strowig, J. Henao-Mejia, E. Elinav, and R. Flavell, “Inam-
masomes in health and disease,” Nature,vol.,no.,pp.
–, .
[] H. Makabe, M. Kojika, G. Takahashi et al., “Interleukin- levels
reect the long-term prognosis of acute lung injury and acute
respiratory distress syndrome,Journal of Anesthesia,vol.,no.
,pp.,.
[] J. Cohen, “e immunopathogenesis of sepsis,Nature,vol.,
no.,pp.,.
[] D. Bouros, M. G. Alexandrakis, K. M. Antoniou et al., “e
clinical signicance of serum and bronchoalveolar lavage
inammatory cytokines in patients at risk for Acute Respirator y
Distress Syndrome,BMC Pulmonary Medicine,vol.,article,
.
[] P. E. Parsons, M. A. Matthay, L. B. Ware, and M. D. Eisner,
“Elevated plasma levels of soluble TNF receptors are associated
with morbidity and mortality in patients with acute lung
injury,e American Journal of Physiology—Lung Cellular and
Molecular Physiology, vol. , no. , pp. L–L, .
[] G.U.Meduri,S.Headley,G.Kohleretal.,“Persistentelevation
of inammatory cytokines predicts a poor outcome in ARDS:
Disease Markers 
plasma IL-𝛽and IL- levels are consistent and ecient predic-
tors of outcome over time,Chest, vol. , no. , pp. –,
.
[] P. E. Parsons, M. Moss, J. L. Vannice, E. E. Moore, F. A.
Moore, and J. E. Repine, “Circulating IL-ra and IL- levels
are increased but do not predict the development of acute
respiratory distress syndrome in at-risk patients,American
JournalofRespiratoryandCriticalCareMedicine,vol.,no.
, pp. –, .
[]M.A.Kovach,K.A.Stringer,R.Buntingetal.,“Microarray
analysis identies IL- receptor type  as a novel candidate
biomarker in patients with acute respiratory distress syndrome,
Respiratory Research,vol.,article,.
[] R. G. Brower, P. N. Lanken, N. MacIntyre et al., “Higher versus
lower positive end-expiratory pressures in patients with the
acute respiratory distress syndrome,e New England Journal
of Medicine, vol. , no. , pp. –, .
[] P. E. Parsons, M. D. Eisner, B. T. ompson et al., “Lower tidal
volume ventilation and plasma cytokine markers of inamma-
tion in patients with acute lung injury,Critical Care Medicine,
vol.,no.,pp.,.
[] D. F. Fiorentino, A. Zlotnik, T. R. Mosmann, M. Howard, and
A. O’Garra, “IL- inhibits cytokine production by activated
macrophages,JournalofImmunology,vol.,no.,pp.
, .
[] L. Armstrong and A. B. Millar, “Relative production of tumour
necrosis factor and interleukin  in adult respiratory distress
syndrome,orax,vol.,no.,pp.,.
[] M. J. Cohen, K. Brohi, C. S. Calfee et al., “Early release of
high mobility group box nuclear protein  aer severe trauma
in humans: role of injury severity and tissue hypoperfusion,
Critical Care,vol.,no.,articleR,.
[] T.Nakamura,E.Sato,N.Fujiwara,Y.Kawagoe,S.Maeda,andS.-
I. Yamagishi, “Increased levels of soluble receptor for advanced
glycation end products (sRAGE) and high mobility group box
 (HMGB) are associated with death in patients with acute
respiratory distress syndrome,Clinical Biochemistry,vol.,
no. -, pp. –, .
[] T. R. Martin, G. D. Rubenfeld, J. T. Ruzinski et al., “Relationship
between soluble CD, lipopolysaccharide binding protein,
and the alveolar inammatory response in patients with acute
respiratory distress syndrome,American Journal of Respiratory
and Critical Care Medicine,vol.,no.,pp.,.
[] J.Villar,L.P
´
erez-M´
endez, E. Espinosa et al., “Serum lipopol-
ysaccharide binding protein levels predict severity of lung
injury and mortality in patients with severe sepsis,PLoS ONE,
vol. , no. , Article ID e, .
[] C. Sittipunt, K. P. Steinberg, J. T. Ruzinski et al., “Nitric oxide
and nitrotyrosine in the lungs of patients with acute respiratory
distress syndrome,AmericanJournalofRespiratoryandCritical
Care Medicine,vol.,no.,pp.,.
[] L.B.Ware,J.A.Magarik,N.Wickershametal.,“Lowplasma
citrulline levels are associated with acute respiratory distress
syndrome in patients with severe sepsis,Critical Care,vol.,
article R, .
[]E.K.Bajwa,U.A.Khan,J.L.Januzzi,M.N.Gong,B.T.
ompson, and D. C. Christiani, “Plasma C-reactive protein
levelsareassociatedwithimprovedoutcomeinARDS,Chest,
vol. , no. , pp. –, .
[] S. H. Hoeboer, H. M. Oudemans-van Straaten, and A. B. J.
Groeneveld, “Albumin rather than C-reactive protein may be
valuable in predicting and monitoring the severity and course
of acute respiratory distress syndrome in critically ill patients
with or at risk for the syndrome aer new onset fever,BMC
Pulmonary Medicine, vol. , article , .
[] I. R. Doyle, C. Hermans, A. Bernard, T. E. Nicholas, and
A. D. Bersten, “Clearance of clara cell secretory protein 
(CC) and surfactant proteins A and B from blood in acute
respiratory failure,American Journal of Respirator yand Critical
Care Medicine,vol.,no.,pp.,.
[] L. B. Ware, J. A. Bastarache, and L. Wang, “Coagulation
and brinolysis in human acute lung injury—new therapeutic
targets?” Keio Journal of Medicine,vol.,no.,pp.,
.
[]P.Prabhakaran,L.B.Ware,K.E.White,M.T.Cross,M.A.
Matthay, and M. A. Olman, “Elevated levels of plasminogen
activator inhibitor- in pulmonary edema uid are associated
with mortality in acute lung injury,American Journal of
Physiology—Lung Cellular and Molecular Physiology,vol.,
no. , pp. L–L, .
[]L.B.Ware,X.Fang,andM.A.Matthay,“ProteinCand
thrombomodulin in human acute lung injury,e American
Journal of Physiology—Lung Cellular and Molecular Physiology,
vol. , no. , pp. L–L, .
[] A. Sapru, C. S. Calfee, K. D. Liu et al., “Plasma soluble
thrombomodulin levels are associated with mortality in the
acute respiratory distress syndrome,Intensive Care Medicine,
vol. , pp. –, .
[] A.Koutsi,A.Papapanagiotou,andA.G.Papavassiliou,“rom-
bomodulin: from haemostasis to inammation and tumouri-
genesis,InternationalJournalofBiochemistryandCellBiology,
vol. , no. , pp. –, .
[] E. M. Conway, “rombomodul in and its role in inammation,”
Seminars in Immunopathology,vol.,no.,pp.,.
[] W. Ruf and M. Riewald, “Tissue factor-dependent coagulation
protease signaling in acute lung injury,Critical Care Medicine,
vol. , no. , pp. S–S, .
[] M. Xue, Z. Sun, M. Shao et al., “Diagnostic and prognostic utility
of tissue factor for severe sepsis and sepsis-induced acute lung
injury,Journal of Translational Medicine,vol.,article,
.
[] J.A.Bastarache,S.C.Sebag,J.K.Cluneetal.,“Lowlevelsof
tissue factor lead to alveolar haemorrhage, potentiating murine
acute lung injury and oxidative stress,orax,vol.,no.,
pp. –, .
[]S.Mumby,L.Ramakrishnan,T.W.Evans,M.J.D.Griths,
and G. J. Quinlan, “Methemoglobin-induced signaling and
chemokine responses in human alveolar epithelial cells,e
American Journal of Physiology—Lung Cellular and Molecular
Physiology,vol.,no.,pp.LL,.
[] A. Fein, R. F. Grossman, J. G. Jones et al., “e value of edema
uid protein measurement in patients with pulmonary edema,
e American Journal of Medicine,vol.,no.,pp.,.
[]L.B.Ware,R.D.Fremont,J.A.Bastarache,C.S.Calfee,
and M. A. Matthay, “Determining the aetiology of pulmonary
oedema by the oedema uid-to-plasma protein ratio,European
Respiratory Journal,vol.,no.,pp.,.
[] D.R.ickett,L.Armstrong,S.J.Christie,andA.B.Millar,
“Vascular endothelial growth factor may contribute to increased
vascular permeability in acute respiratory distress syndrome,
American Journal of Respiratory and Critical Care Medicine,vol.
, no. , pp. –, .
 Disease Markers
[] Y. Abadie, F. Bregeon, L. Papazian et al., “Decreased VEGF
concentration in lung tissue and vascular injury during ARDS,
European Respiratory Journal,vol.,no.,pp.,.
[] T. J. Desai and W. V. Cardoso, “Growth factors in lung develop-
ment and disease: friends or foe?” Respiratory Research,vol.,
article , .
[] B.Maitre,S.Boussat,D.Jeanetal.,“Vascularendothelialgrowth
factor synthesis in the acute phase of experimental and clinical
lung injury,European Respiratory Journal,vol.,no.,pp.
, .
[] Z. Borok, S. I. Danto, L. L. Dimen, X.-L. Zhang, and R.
L. Lubman, “Na+-K+-ATPase expression in alveolar epithelial
cells: upregulation of active ion transport by KGF,American
Journal of Physiology—Lung Cellular and Molecular Physiology,
vol. , no. , pp. L–L, .
[]L.B.WareandM.A.Matthay,“Keratinocyteandhepatocyte
growth factors in the lung: Roles in lung development, inam-
mation, and repair,e American Journal of Physiology—Lung
Cellular and Molecular Physiology,vol.,no.,pp.L
L, .
[]V.Galani,E.Tatsaki,M.Baietal.,“eroleofapoptosisin
the pathophysiology of Acute Respiratory Distress Syndrome
(ARDS): an up-to-date cell-specic review, Pathology Research
and Practice,vol.,no.,pp.,.
[] R.C.Pires-Neto,M.M.B.Morales,T.Lancasetal.,“Expression
of acute-phase cytokines, surfactant proteins, and epithelial
apoptosis in small airways of human acute respiratory distress
syndrome,Journal of Critical Care, vol. , no. , pp. .e–
.e, .
[] D. Talmor, T. Sarge, A. Legedza et al., “Cytokine release
following recruitment maneuvers,Chest,vol.,no.,pp.
–, .
[]J.M.Walter,J.Wilson,andL.B.Ware,“Biomarkersinacute
respiratory distress syndrome: from pathobiology to improving
patient care,Expert Review of Respiratory Medicine,vol.,no.
, pp. –, .
[] C. S. Calfee, K. Delucchi, P. E. Parsons, B. T. ompson, L. B.
Ware, and M. A. Matthay, “Latent class models identify two
subphenotypes in respiratory distress syndrome with dieren-
tial response to positive end-expiratory pressure,” Annals of the
American oracic Society,vol.,supplement,p.S,.
[] V. Newman, R. F. Gonzalez, M. A. Matthay, and L. G. Dobbs, “A
novel alveolar type I cell-specic biochemical marker of human
acute lung injury,American Journal of Respiratory and Critical
Care Medicine,vol.,no.,pp.,.
[] A. M. Schmidt, S. D. Yan, S. F.Yan, and D. M. Stern, “e biology
of the receptor for advanced glycation end products and its
ligands,Biochimica et Biophysica Acta (BBA)—Molecular Cell
Research, vol. , no. -, pp. –, .
[] T. Uchida, M. Shirasawa, L. B. Ware et al., “Receptor for
advanced glycation end-products is a marker of type I cell
injury in acute lung injury,American Journal of Respiratory and
Critical Care Medicine,vol.,no.,pp.,.
[] M.T.Kuipers,T.vanderPoll,M.J.Schultz,andC.W.Wieland,
“Bench-to-bedside review: damage-associated molecular pat-
terns in the onset of ventilator-induced lung injury,Critical
Care,vol.,no.,article,.
[] T. Uchida, M. Shirasawa, L. B. Ware et al., “Receptor for
advanced glycation end-products is a marker of type I cell
injury in acute lung injury,American Journal of Respiratory and
Critical Care Medicine,vol.,no.,pp.,.
[] C. S. Calfee, L. B. Ware, M. D. Eisner et al., “Plasma receptor for
advanced glycation end products and clinical outcomes in acute
lung injury,orax, vol. , no. , pp. –, .
[] M.Jabaudon,E.Futier,L.Roszyketal.,“Solubleformofthe
receptor for advanced glycation end products is a marker of
acute lung injury but not of severes epsis in critically ill patients,
Critical Care Medicine, vol. , no. , pp. –, .
[] M. Jabaudon, R. Blondonnet, L. Roszyk et al., “Soluble RAGE
predicts impaired alveolar uid clearance in acute respiratory
distress syndrome,AmericanJournalofRespiratoryandCritical
Care Medicine,vol.,no.,.
[] M. D. Johnson, J. H. Widdicombe, L. Allen, P. Barbry, and L.
G. Dobbs, “Alveolar epithelial I cells contain transport proteins
and transport sodium, supporting an active role for type I cells
in regulation of lung liquid homeostasis,” Proceedings of the
National Academy of Sciences of the United States of America,
vol. , no. , pp. –, .
[] L. E. Hanford, J. J. Enghild, Z. Valnickova et al., “Purication
and characterization of mouse soluble receptor for advanced
glycation end products (sRAGE),e Journal of Biological
Chemistry,vol.,no.,pp.,.
[] C.Cheng,K.Tsuneyama,R.Kominamietal.,“Expressionpro-
ling of endogenous secretory receptor for advanced glycation
end products in human organs,Modern Pathology,vol.,no.
, pp. –, .
[] J. Brett, A. M. Schmidt, S. D. Yan et al., “Survey of the
distribution of a newly characterized receptor for advanced
glycation end products in tissues,e American Journal of
Patholog y,vol.,no.,pp.,.
[]L.E.Hanford,C.L.Fattman,L.M.Schaefer,J.J.Enghild,
Z. Valnickova, and T. D. Oury, “Regulation of receptor for
advanced glycation end products during bleomycin-induced
lung injury,American Journal of Respiratory Cell and Molecular
Biology,vol.,no.,pp.SS,.
[]J.M.Englert,L.E.Hanford,N.Kaminskietal.,“Arolefor
the receptor for advanced glycation end products in idiopathic
pulmonary brosis,e American Journal of Pathology,vol.,
no.,pp.,.
[] A. M. Schmidt, S. D. Yan, S. F. Yan, and D. M. Stern, “e
multiligand receptor RAGE as a progression factor amplifying
immune and inammatory responses,e Journal of Clinical
Investigation,vol.,no.,pp.,.
[] M. Jabaudon, R. Blondonnet, L. Roszyk et al., “Soluble forms
and ligands of the re ceptor for advanced glycation end-products
in patients with acute respiratory distress syndrome: an obser-
vational prospective study,PLoS ONE,vol.,no.,ArticleID
e, .
[] C. A. Downs, L. H. Kreiner, N. M. Johnson, L. A. Brown, and
M. N. Helms, “Receptor for advanced glycation end-products
regulates lung uid balance via protein kinase C-gp91phox
signaling to epithelial sodium channels,American Journal of
Respiratory Cell and Molecular Biology,vol.,no.,pp.,
.
[] R. Briot, J. A. Frank, T. Uchida, J. W. Lee, C. S. Calfee, and M. A.
Matthay, “Elevated levels of the receptor for advanced glycation
end products, a marker of alveolar epithelial type i cell injury,
predict impaired alveolar uid clearance in isolated perfused
human lungs,Chest,vol.,no.,pp.,.
[] J.-M. Constantin, “Impact of Inammation Biomarkers
on the Acute Respiratory Distress Syndrome (ARDS)
Denition,” (NCT), https://clinicaltrials.gov/ct/show/
NCT?term=constantin+clermont&rank=.
Disease Markers 
[] M. Jabaudon, N. Hamroun, L. Roszyk et al., “Eects of a recruit-
ment maneuver on plasma levels of soluble RAGE in patients
with diuse acute respiratory distress syndrome: a prospective
randomized crossover study,Intensive Care Medicine,vol.,
no.,pp.,.
[] M. Jabaudon, “Predictive Values of Plasma Soluble RAGE Levels
and RAGE Polymorphisms for the Onset of Acute Respira-
tory Distress Syndrome in Critically Ill Patients (PrediRAGE
Study),” in: ClinicalTrials.gov, , http://clinicaltrials.gov/
show/NCT.
[] J. Bhattacharya and M. A. Matthay, “Regulation and repair of the
alveolar-capillary barrier in acute lung injury,Annual Review of
Physiology,vol.,pp.,.
[] I. Frerking, A. G¨
unther,W.Seeger,andU.Pison,“Pulmonary
surfactant: functions, abnormalities and therapeutic options,
Intensive Care Medicine,vol.,no.,pp.,.
[]D.G.Ashbaugh,D.B.Bigelow,T.L.Petty,andB.E.Levine,
Acute respiratory distress in adults,e Lancet, vol. , no. ,
pp.,.
[] I. R. Doyle, A. D. Bersten, and T. E. Nicholas, “Surfactant
proteins-A and -B are elevated in plasma of patients with acute
respiratory failure,American Journal of Respirator yand Critical
Care Medicine,vol.,no.,pp.,.
[]K.E.Greene,S.Ye,R.J.Mason,andP.E.Parsons,“Serum
surfactant protein-A levels predict development of ARDS in at-
risk patients,Chest, vol. , supplement , pp. S–S, .
[]A.D.Bersten,T.Hunt,T.E.Nicholas,andI.R.Doyle,
“Elevated plasma surfactant protein-B predicts development
of acute respiratory distress syndrome in patients with acute
respiratory failure,American Journal of Respirator yand Critical
Care Medicine,vol.,no.,pp.,.
[] A. Ishizaka, T. Matsuda, K. H. Albertine et al., “Elevation of KL-
, a lung epithelial cell marker, in plasma and epithelial lining
uid in acute respiratory distress syndrome,e American
Journal of Physiology—Lung Cellular and Molecular Physiology,
vol. , no. , pp. L–L, .
[] N. Nathani, G. D. Perkins, W. Tunniclie, N. Murphy, M. Manji,
andD.R.ickett,“KerbsvonLungrenantigenisamarkerof
alveolar inammation but not of infection in patients with acute
respiratory distress syndrome,Critical Care,vol.,articleR,
.
[] T.Kondo,N.Hattori,N.Ishikawaetal.,“KL-concentration
in pulmonary epithelial lining uid is a useful prognostic
indicator in patients with acute respiratory distress syndrome,
Respiratory Research, vol. , article , .
[] R.Lucas,A.D.Verin,S.M.Black,andJ.D.Catravas,“Regulators
of endothelial and epithelial barrier integrity and function in
acute lung injury,Biochemical Pharmacology,vol.,no.,pp.
–, .
[] A. Binnie, J. L. Tsang, and C. C. dos Santos, “Biomarkers
in acute respiratory distress syndrome,Current Opinion in
Critical Care,vol.,no.,pp.,.
[] S.Tsigkos,M.Koutsilieris,andA.Papapetropoulos,“Angiopoi-
etins in angiogenesis and beyond,Expert Opinion on Investiga-
tional Drugs,vol.,no.,pp.,.
[] U. Fiedler, Y. Reiss, M. Scharpfenecker et al., “Angiopoietin-
sensitizes endothelial cells to TNF-𝛼and has a crucial role in
the induction of inammation,Nature Medicine,vol.,no.,
pp. –, .
[] V. Bhandari and J. A. Elias, “e role of angiopoietin  in
hyperoxia-induced acute lung injury,Cell Cycle,vol.,no.,
pp.,.
[] N. J. Meyer, M. Li, R. Feng et al., “ANGPT genetic variant
is associated with trauma-associated acute lung injury and
altered plasma angiopoietin- isoform ratio,American Journal
of Respiratory and Critical Care Medicine,vol.,no.,pp.
–, .
[] A. Agrawal, M. A. Matthay, K. N. Kangelaris et al., “Plasma
angiopoietin- predicts the onset of acute lung injury in criti-
cally ill patients,American Journal of Respiratory and Critical
Care Medicine,vol.,no.,pp.,.
[] T. Wada, S. Jesmin, S. Gando et al., “e role of angiogenic
factors and their soluble receptors in acute lung injury (ALI)/
acute respiratory distress syndrome (ARDS) associated with
critical illness,Journal of Inammation,vol.,article,.
[] T. Ong, D. E. McClintock, R. H. Kallet, L. B. Ware, M.
A. Matthay, and K. D. Liu, “Ratio of angiopoietin- to
angiopoietin- as a predictor of mortality in acute lung injury
patients,Critical Care Medicine,vol.,no.,pp.,
.
[] K. A. Roebuck and A. Finnegan, “Regulation of intercellular
adhesion molecule- (CD) gene expression,Journal of Leu ko-
cyte Biology, vol. , no. , pp. –, .
[] E. R. Conner, L. B. Ware, G. Modin, and M. A. Matthay,
“Elevated pulmonary edema uid concentrations of soluble
intercellular adhesion molecule- in patients with acute lung
injury: biological and clinical signicance,Chest,vol.,pp.
S–S, .
[] C.S.Calfee,M.D.Eisner,P.E.Parsonsetal.,“Solubleinter-
cellular adhesion molecule- and clinical outcomes in patients
with acute lung injury,IntensiveCareMedicine,vol.,no.,
pp. –, .
[] P.Agouridakis,D.Kyriakou,M.G.Alexandrakisetal.,“e
predictive role of serum and bronchoalveolar lavage cytokines
and adhesion molecules for acute respiratory distress syndrome
development and outcome,Respiratory Research,vol.,article
, .
[] H. R. Flori, L. B. Ware, D. Glidden, and M. A. Matthay, “Early
elevation of plasma soluble intercellular adhesion molecule- in
pediatric acute lung injury identies patients at increased risk of
death and prolonged mechanical ventilation,Pediatric Critical
Care Medicine,vol.,no.,pp.,.
[] A. Sousa, F. Raposo, S. Fonseca et al., “Measurement of
cytokines and adhesion molecules in the rst  hours aer
severe trauma: association with severity and outcome,Disease
Markers,vol.,ArticleID,pages,.
[] S. Gando, T. Kameue, N. Matsuda et al., “Combined activation
of coagulation and inammation has an important role in
multiple organ dysfunction and poor outcome aer severe
trauma,rombosis and Haemostasis,vol.,no.,pp.
, .
[] K. Xing, S. Murthy, W. C. Liles, and J. M. Singh, “Clinical utility
of biomarkers of endothelial activation in sepsis-a systematic
review,Critical Care,vol.,articleR,.
[] J.Boldt,M.Wollbr
¨
uck, D. Kuhn, L. C. Linke, and G. Hempel-
mann, “Do plasma levels of circulating soluble adhesion
molecules dier between surviving and nonsurviving critically
ill patients?” Chest,vol.,no.,pp.,.
[] F. Sakamaki, A. Ishizaka, M. Handa et al., “Soluble form of P-
selectin in plasma is elevated in acute lung injury,American
JournalofRespiratoryandCriticalCareMedicine,vol.,no.,
pp.,.
[] E.L.Burnham,M.Moss,F.Harris,andL.A.S.Brown,“Elevated
plasma and lung endothelial selectin levels in patients with
 Disease Markers
acute respiratory distress syndrome and a history of chronic
alcohol abuse,Critical Care Medicine,vol.,no.,pp.
, .
[] K. Okajima, N. Harada, G. Sakurai et al., “Rapid assay for plasma
soluble E-selectin predicts the development of acute respiratory
distress syndrome in patients with systemic inammatory
response syndrome,Tra nsla tion al Res e arc h,vol.,no.,pp.
–, .
[] A.R.L.MedfordandA.B.Millar,“Vascularendothelialgrowth
factor (VEGF) in acute lung injury (ALI) and acute respiratory
distress syndrome (ARDS): paradox or paradigm?” orax,vol.
,no.,pp.,.
[] M. Shibuya, “Dierential roles of vascular endothelial growth
factor receptor- and receptor- in angiogenesis,Journal of
Biochemistry and Molecular Biology,vol.,no.,pp.,
.
[] R.J.Kaner,J.V.Ladetto,R.Singh,N.Fukuda,M.A.Matthay,
and R. G. Crystal, “Lung overexpression of the vascular
endothelial growth factor gene induces pulmonary edema,
American Journal of Respiratory Cell and Molecular Biology,vol.
, no. , pp. –, .
[] L. Azamrei, S. Gurzu, R. Solomon et al., “Vascular endothelial
growth factor: a possible mediator of endothelial activation in
acute respiratory distress syndrome,Minerva Anestesiologica,
vol.,no.,pp.,.
[]D.R.ickett,L.Armstrong,andA.B.Millar,“Arolefor
vascular endothelial growth factor in acute and resolving lung
injury,American Journal of Respiratory and Critical Care
Medicine,vol.,no.,pp.,.
[] A. C. A. Carvalho, S. M. Bellman, V. J. Saullo, D. Quinn, and W.
M. Zapol, “Altered factor VIII in acute respiratory failure,” e
New England Journal of Medicine, vol. , no. , pp. –,
.
[] D. B. Rubin, J. P. Wiener-Kronish, J. F. Murray et al., “Elevated
vonWillebrandfactorantigenisanearlyplasmapredictorof
acute lung injury in nonpulmonary sepsis syndrome,” Journal
of Clinical Investigation,vol.,no.,pp.,.
[] M. Moss, L. Ackerson, M. K. Gillespie, F. A. Moore, E. E.
Moore, and P. E. Parsons, “Von willebrand factor antigen levels
are not predictive for the adult respiratory distress syndrome,
American Journal of Respiratory and Critical Care Medicine,vol.
, no. , pp. –, .
[] A. K. Sabharwal, S. P. Bajaj, A. Ameri et al., “Tissue factor
pathway inhibitor and von Willebrand factor antigen levels in
adult respiratory distress syndrome and in a primate model
of sepsis,” American Journal of Respiratory and Critical Care
Medicine,vol.,no.I,pp.,.
[] P. R. M. Rocco, C. Dos Santos, and P. Pelosi, “Lung parenchyma
remodeling in acute respiratory distress syndrome,Minerva
Anestesiologica,vol.,no.,pp.,.
[] B. C. Starcher, “Lung elastin and matrix,Chest,vol.,no.,
supplement , pp. S–S, .
[] G. M. Albaiceta, A. Gutierrez-Fern´
andez, E. Garc´
ıa-Prieto et al.,
Absence or inhibition of matrix metalloproteinase- decreases
ventilator-induced lung injury,American Journal of Respirator y
Cell and Molecular Biology,vol.,no.,pp.,.
[] S. E. G. Fligiel, T. Standiford, H. M. Fligiel et al., “Matrix
metalloproteinases and matrix metalloproteinase inhibitors in
acute lung injury,Human Pathology,vol.,no.,pp.,
.
[] M. Y. F. Kong, Y. Li, R. Oster, A. Gaggar, and J. P. Clancy,
“Early elevation of matrix metalloproteinase- and - in pedi-
atric ARDS is associated with an increased risk of prolonged
mechanical ventilation,PLoS ONE,vol.,no.,ArticleID
e, .
[] A. Gonz´
alez-L´
opez and G. M. Albaiceta, “Repair aer acute
lung injury: molecular mechanisms and therapeutic opportu-
nities,Critical Care,vol.,no.,article,.
[] M. Bhatia and S. Moochhala, “Role of inammatory mediators
in the pathophysiology of acute respiratory distress syndrome,
Journal of Patholog y,vol.,no.,pp.,.
[]L.A.DadaandJ.I.Sznajder,“Hypoxicinhibitionofalveolar
uid reabsorption,” in Hypoxia and the Circulation, pp. –,
Springer US, .
[]A.E.PostlethwaiteandJ.M.Seyer,“Stimulationofbroblast
chemotaxis by human recombinant tumor necrosis factor 𝛼
(TNF-𝛼)andasyntheticTNF-𝛼- peptide,e Journal of
Experimental Medicine,vol.,no.,pp.,.
[] P. F. Piguet, M. A. Collart, G. E. Grau, A.-P. Sappino, and P. Vas-
salli, “Requirement of tumour necrosis factor for development
of silica-induced pulmonary brosis,Nature,vol.,no.,
pp. –, .
[] W.Y.Park,R.B.Goodman,K.P.Steinbergetal.,“Cytokine
balance in the lungs of patients with acute respiratory distress
syndrome,American Journal of Respiratory and Critical Care
Medicine,vol.,no.,pp.,.
[] J. Pugin, B. Ricou, K. P. Steinberg, P. M. Suter, and T. R. Martin,
“Proinammatory activity in bronchoalveolar lavage uids
from patients with ARDS, a prominent role for interleukin-,
American Journal of Respiratory and Critical Care Medicine,vol.
, no. , pp. –, .
[]G.U.Meduri,G.Kohler,S.Headley,E.Tolley,F.Stentz,
and A. Postlethwaite, “Inammatory cytokines in the BAL of
patients with ARDS. Persistent elevation over time predicts
poor outcome,” Chest,vol.,no.,pp.,.
[] R.Roten,M.Markert,F.Feihl,M.-D.Schaller,M.-C.Tagan,and
C. Perret, “Plasma levels of tumor necrosis factor in the adult
respiratory distress syndrome,American Review of Respiratory
Disease,vol.,no.,pp.,.
[] T. Dolinay, Y. S. Kim, J. Howrylak et al., “Inammasome-
regulated cytokines are critical mediators of acute lung injury,
American Journal of Respiratory and Critical Care Medicine,vol.
, no. , pp. –, .
[] D. McClintock, H. Zhuo, N. Wickersham, M. A. Matthay, and
L. B. Ware, “Biomarkers of inammation, coagulation and
brinolysis predict mortality in acute lung injury,Critical Care,
vol. , no. , article R, .
[] R. P. Baughman, K. L. Gunther, M. C. Rashkin, D. A. Keeton,
and E. N. Pattishall, “Changes in the inammatory response of
the lung during acute respiratory distress syndrome: prognostic
indicators,American Journal of Respiratory and Critical Care
Medicine,vol.,no.,pp.,.
[] H. Sch¨
utte,J.Lohmeyer,S.Rosseauetal.,“Bronchoalveolar
and systemic cytokine proles in patients with ARDS, severe
pneumonia and cardiogenic pulmonary oedema,” European
Respiratory Journal, vol. , no. , pp. –, .
[] A. Kurdowska, E. J. Miller, J. M. Noble et al., “Anti-IL-
autoantibodies in alveolar uid from patients with the adult
respiratory distress syndrome,e Journal of Immunology,vol.
, no. , pp. –, .
[]A.Kurdowska,J.M.Noble,I.S.Grant,C.R.Robertson,C.
Haslett, and S. C. Donnelly, “Anti-interleukin- autoantibodies
Disease Markers 
in patients at risk for acute respiratory distress syndrome,
Critical Care Medicine,vol.,no.,pp.,.
[]A.Krupa,H.Kato,M.A.Matthay,andA.K.Kurdowska,
“Proinammatory activity of anti-IL- autoantibody:IL- com-
plexes in alveolar edema uid from patients with acute lung
injury,e American Journal of Physiology—Lung Cellular and
Molecular Physiology, vol. , no. , pp. L–L, .
[] R. Fudala, A. Krupa, M. A. Matthay, T. C. Allen, and A. K.
Kurdowska, “Anti-IL- autoantibody:IL- immune complexes
suppress spontaneous apoptosis of neutrophils,” American Jour-
nal of Physiology—Lung Cellular and Molecular Physiology,vol.
, no. , pp. L–L, .
[] M.A.MatthayandL.B.Ware,“Resolutionofalveolaredemain
acute respiratory distress syndrome. Physiology and biology,
American Journal of Respiratory and Critical Care Medicine,vol.
, no. , pp. –, .
[] P. Ralph, I. Nakoinz, A. Sampson-Johannes et al., “IL-, T
lymphocyte inhibitor of human blood cell production of IL-
and tumor necrosis factor,e Journal of Immunology,vol.,
no. , pp. –, .
[] M. Seitz, P. Loetscher, B. Dewald, H. Towbin, H. Gallati, and
M. Baggiolini, “Interleukin- dierentially regulates cytokine
inhibitor and chemokine release from blood mononuclear cells
and broblasts,EuropeanJournalofImmunology,vol.,no.,
pp. –, .
[] C.-J.Lo,M.Fu,andH.G.Cryer,“Interleukininhibitsalveolar
macrophage production of inammatory mediators involved
in adult respiratory distress syndrome,Journal of Surgical
Research, vol. , no. , pp. –, .
[]A.Sapru,J.L.Wiemels,J.S.Witte,L.B.Ware,andM.A.
Matthay, “Acute lung injury and the coagulation pathway:
potential role of gene polymorphisms in the protein C and
brinolytic pathways,IntensiveCareMedicine,vol.,no.,pp.
–, .
[] V. Jalkanen, R. Yang, R. Linko et al., “SuPAR and PAI- in
critically ill, mechanically ventilated patients,Intensive Care
Medicine,vol.,no.,pp.,.
[] L. B. Ware, M. A. Matthay, P. E. Parsons et al., “Pathogenetic and
prognostic signicance of alteredcoagulation and brinolysis in
acute lung injury/acute respiratory distress syndrome,Critical
Care Medicine,vol.,no.,pp.,.
[] C.T.Esmon,“InammationandtheactivatedproteinCantico-
agulant pathway,Seminars in rombosis and Hemostasis,vol.
, supplement , pp. –, .
[] M. F. Nold, C. A. Nold-Petry, D. Fischer et al., “Activated
protein C downregulates p mitogen-activated protein kinase
and improves clinical parameters in an in-vivo model of septic
shock,rombosis and Haemostasis,vol.,no.,pp.,
.
[] T.Cheng,D.Liu,J.H.Grinetal.,“ActivatedproteinCblocks
p-mediated apoptosis in ischemic human brain endothelium
and is neuroprotective,Nature Medicine,vol.,no.,pp.
, .
[] S.C.Christiaans,B.M.Wagener,C.T.Esmon,andJ.F.Pittet,
“Protein C and acute inammation: a clinical and biological
perspective,e American Journal of Physiology—Lung Cellular
and Molecular Physiology,vol.,no.,pp.LL,.
[] J.A.Bastarache,L.Wang,T.Geiseretal.,“ealveolarepithe-
lium can initiate the extrinsic coagulation cascade through
expression of tissue factor,orax,vol.,no.,pp.,
.
[] A.J.Ghio,J.H.Richards,K.M.Crissman,andJ.D.Carter,“Iron
disequilibrium in the rat lung aer instilled blood,Chest,vol.
,no.,pp.,.
[] J. A. Bastarache, J. L. Wynn, and L. B. Ware, “Fanning
the re: can methemoglobin enhance neutrophil activation?”
EBioMedicine,vol.,no.,pp.,.
[] E.L.Burnham,W.J.Janssen,D.W.H.Riches,M.Moss,and
G. P. Downey, “e broproliferative response in acute respira-
tory distress syndrome: mechanisms and clinical signicance,
European Respiratory Journal,vol.,no.,pp.,.
[] T. Geiser, K. Atabai, P.-H. Jarreau, L. B. Ware, J. Pugin, and M.
A. Matthay, “Pulmonary edema uid from patients with acute
lung injury augments in vitro alveolar epithelial repair by an IL-
𝛽-dependent mechanism,Amer ican Journal of Respiratory and
Critical Care Medicine, vol. , no. , pp. –, .
[] K.Atabai,M.Ishigaki,T.Geiser,I.Ueki,M.A.Matthay,andL.B.
Ware, “Keratinocyte growth factor can enhance alveolar epithe-
lial repair by nonmitogenic mechanisms,American Journal of
Physiology—Lung Cellular and Molecular Physiology,vol.,
no. , pp. L–L, .
[] M. Mura, C. C. dos Santos, D. Stewart, and M. Liu, “Vascular
endothelial growth factor and related molecules in acute lung
injury,Journal of Applied Physiology,vol.,no.,pp.
, .
[]C.Quesnel,S.Marchand-Adam,A.Fabreetal.,“Regulation
of hepatocyte growth factor secretion by broblasts in patients
with acute lung injury,American Journal of Physiology—Lung
Cellular and Molecular Physiology,vol.,no.,pp.L
L, .
[]S.Fan,Y.X.Ma,J.-A.Wangetal.,“ecytokinehepatocyte
growth factor/scatter factor inhibits apoptosis and enhances
DNA repair by a common mechanism involving signaling
through phosphatidyl inositol 󸀠kinase,Oncogene,vol.,no.
, pp. –, .
[] J.-B. Stern, L. Fierobe, C. Paugam et al., “Keratinocyte growth
factor and hepatocyte growth factor in bronchoalveolar lavage
uid in acute respiratory distress syndrome patients,Critical
Care Medicine,vol.,no.,pp.,.
[] G. M. Verghese, K. McCormick-Shannon, R. J. Mason, and M.
A. Matthay, “Hepatocyte growth factor and keratinocyte growth
factor in the pulmonary edema uid of patients with acute lung
injury: biologic and clinical signicance,American Journal of
Respiratory and Critical Care Medicine,vol.,no.,pp.
, .
[] M. A. Rahman, K. Sundaram, S. Mitra, M. A. Gavrilin, and
M. D. Wewers, “Receptor interacting protein- plays a critical
role in human lung epithelial cells survival in response to Fas-
induced cell-death,PLoS ONE,vol.,no.,ArticleIDe,
.
[]A.D.Lopez,S.Avasarala,S.Grewal,A.K.Murali,andL.
London, “Dierential role of the Fas/Fas ligand apoptotic
pathway in inammation and lung brosis associated with
reovirus /L-induced bronchiolitis obliterans organizing pneu-
monia and acute respiratory distress syndrome,e Journal of
Immunology,vol.,no.,pp.,.
[] G. Matute-Bello, W. C. Liles, K. P. Steinberg et al., “Soluble Fas
ligand induces epithelial cell apoptosis in humans with acute
lung injury (ARDS),Journal of Immunology,vol.,no.,pp.
–, .
[] M. Tanaka, T. Suda, T. Takahashi, and S. Nagata, “Expression
of the functional soluble form of human Fas ligand in activated
 Disease Markers
lymphocytes,e EMBO Journal,vol.,no.,pp.,
.
[] K.H.Albertine,M.F.Soulier,Z.Wangetal.,“Fasandfasligand
are up-regulated in pulmonary edema uid and lung tissue of
patients with acute lung injury and the acute respiratory distress
syndrome,e American Journal of Pathology,vol.,no.,pp.
–, .
[] J. Farjanel, D. J. Hartmann, B. Guidet, L. Luquel, and G.
Oenstadt, “Four markers of collagen metabolism as possible
indicators of disease in the adult respiratory distress syndrome,
American Review of Respiratory Disease,vol.,no.,pp.
, .
[] A.N.Chesnutt,M.A.Matthay,F.A.Tibayan,andJ.G.Clark,
“Early detection of type iii procollagen peptide in acute lung
injury: pathogenetic and prognostic signicance,American
JournalofRespiratoryandCriticalCareMedicine,vol.,no.
, pp. –, .
[]J.G.Clark,J.A.Milberg,K.P.Steinberg,andL.D.Hudson,
“Type III procollagen peptide in the adult respiratory distress
syndrome: association of increased peptide levels in bron-
choalveolar lavage uid with increased risk for death,Annals
of Internal Medicine,vol.,no.,pp.,.
[] J.-M. Forel, C. Guervilly, S. Hraiech et al., “Type III procollagen
is a reliable marker of ARDS-associated lung broproliferation,
Intensive Care Medicine, vol. , no. , pp. –, .
[] C.M.Hendrickson,B.Crestani,andM.A.Matthay,“Biology
and pathology of broproliferation following the acute respira-
tory distress syndrome,Intensive Care Medicine,vol.,no.,
pp.,.
[] L. B. Ware, T. Koyama, D. D. Billheimer et al., “Prognostic
and pathogenetic value of combining clinical and biochemical
indices in patients with acute lung injury,Chest,vol.,no.,
pp. –, .
[] J.-M. Constantin and E. Futier, “Lung imaging in patients with
acute respiratory distress syndrome: from an understanding of
pathophysiology to bedside monitoring,” Minerva Anestesiolog-
ica,vol.,no.,pp.,.
[] L.Puybasset,P.Gusman,J.-C.Muller,P.Cluzel,P.Coriat,and
J.-J. Rouby, “Regional distribution of gas and tissue in acute
respiratory distress syndrome. III. Consequences for the eects
of positive end-expiratory pressure,” IntensiveCareMedicine,
vol. , no. , pp. –, .
[] J.-M. Constantin, S. Jaber, E. Futier et al., “Respiratory eects
of dierent recruitment maneuvers in acute respiratory distress
syndrome,Critical Care,vol.,articleR,.
[] J.-M. Constantin, S. Cayot-Constantin, L. Roszyk et al.,
“Response to recruitment maneuver inuences net alveolar
uid clearance in acute respiratory distress syndrome,Anesthe-
siology,vol.,no.,pp.,.
[] R. Mayeux, “Biomarkers: potential uses and limitations,Neu-
roRx,vol.,no.,pp.,.
[] V.G.DeGruttola,P.Clax,D.L.DeMetsetal.,“Considerations
in the evaluation of surrogate endpoints in clinical trials: sum-
mary of a National Institutes of Health Workshop,Controlled
Clinical Trials, vol. , no. , pp. –, .
[]P.Ray,Y.L.Manach,B.Riou,andT.T.Houle,“Statistical
evaluation of a biomarker,Anesthesiology,vol.,no.,pp.
–, .
[]H.GerlachandS.Toussaint,“Sensitive,specic,predictive...
statisticalbasics:howtousebiomarkers,Critical Care Clinics,
vol. , no. , pp. –, .
[] M. Bhargava and C. H. Wendt, “Biomarkers in acute lung
injury,Tra nsla tio n al Res e arc h,vol.,no.,pp.,.
... At the tissue level, ARDS pathophysiology is characterized by diffuse alveolar epithelial cell apoptosis or necrosis as a result of exposure to pathogens or noninfectious irritants (9,10), with the clinical status of affected patients thus being profoundly shaped by multiple proinflammatory mechanisms. ARDS and associated progression to the development of multi-organ failure (MOF) have been proposed as forms of secondary bystander tissue damage that lie along a spectrum and stem from the effects of dysregulated or excessive inflammatory responses to a primary infectious or non-infectious insult (11). ...
... Patients with severe disease were more likely than nonsevere patients to have hypertension ( Table 1). The median duration of hospitalization for the overall ARDS patient cohort was 15 [8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23] days, with a significantly longer duration of hospitalization among individuals with severe disease as compared to those with nonsevere disease (19 vs. 12 [8][9][10][11][12][13][14][15][16][17][18] days, P = 0.017). ...
... Patients with severe disease were more likely than nonsevere patients to have hypertension ( Table 1). The median duration of hospitalization for the overall ARDS patient cohort was 15 [8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23] days, with a significantly longer duration of hospitalization among individuals with severe disease as compared to those with nonsevere disease (19 vs. 12 [8][9][10][11][12][13][14][15][16][17][18] days, P = 0.017). ...
Article
Full-text available
Introduction Acute respiratory distress syndrome (ARDS) is a major cause of death among critically ill patients in intensive care settings, underscoring the need to identify biomarkers capable of predicting ARDS patient clinical status and prognosis at an early time point. This study specifically sought to explore the utility and clinical relevance of TM9SF1 as a biomarker for the early prediction of disease severity and prognostic outcomes in patients with ARDS. Methods This study enrolled 123 patients with severe ARDS and 116 patients with non-severe ARDS for whom follow-up information was available. The mRNA levels of TM9SF1 and cytokines in peripheral blood mononuclear cells from these patients were evaluated by qPCR. The predictive performance of TM9SF1 and other clinical indicators was evaluated using received operating characteristic (ROC) curves. A predictive nomogram was developed based on TM9SF1 expression and evaluated for its ability in the early prediction of severe disease and mortality in patients with ARDS. Results TM9SF1 mRNA expression was found to be significantly increased in patients with severe ARDS relative to those with non-severe disease or healthy controls. ARDS severity increased in correspondence with the level of TM9SF1 expression (odds ratio [OR] = 2.43, 95% confidence interval [CI] = 2.15–3.72, P = 0.005), and high TM9SF1 levels were associated with a greater risk of mortality (hazard ratio [HR] = 2.27, 95% CI = 2.20–4.39, P = 0.001). ROC curves demonstrated that relative to other clinical indicators, TM9SF1 offered superior performance in the prediction of ARDS severity and mortality. A novel nomogram incorporating TM9SF1 expression together with age, D-dimer levels, and C-reactive protein (CRP) levels was developed and was used to predict ARDS severity (AUC = 0.887, 95% CI = 0.715–0.943). A separate model incorporating TM9SF1 expression, age, neutrophil-lymphocyte ratio (NLR), and D-dimer levels (C-index = 0.890, 95% CI = 0.627–0.957) was also developed for predicting mortality. Conclusion Increases in ARDS severity and patient mortality were observed with rising levels of TM9SF1 expression. TM9SF1 may thus offer utility as a novel biomarker for the early prediction of ARDS patient disease status and clinical outcomes.
... AREG is highly expressed in the lung and has a key involvement in many inflammatory processes but also in ARDS [32]. An impaired interferon response to COVID-19 predicts severe disease [25], and it has been proposed that at least IFN-gamma can suppress ADAM-17 activity [33]. ...
Article
Full-text available
There is a lack of studies aiming to assess cellular a disintegrin and metalloproteinase-17 (ADAM-17) activity in COVID-19 patients and the eventual associations with the shedding of membrane-bound angiotensin-converting enzyme 2 (mACE2). In addition, studies that investigate the relationship between ACE2 and ADAM-17 gene expressions in organs infected by SARS-CoV-2 are lacking. We used data from the Massachusetts general hospital COVID-19 study (306 COVID-19 patients and 78 symptomatic controls) to investigate the association between plasma levels of 33 different ADAM-17 substrates and COVID-19 severity and mortality. As a surrogate of cellular ADAM-17 activity, an ADAM-17 substrate score was calculated. The associations between soluble ACE2 (sACE2) and the ADAM-17 substrate score, renin, key inflammatory markers, and lung injury markers were investigated. Furthermore, we used data from the Genotype-Tissue Expression (GTEx) database to evaluate ADAM-17 and ACE2 gene expressions by age and sex in ages between 20–80 years. We found that increased ADAM-17 activity, as estimated by the ADAM-17 substrates score, was associated with COVID-19 severity (p = 0.001). ADAM-17 activity was also associated with increased mortality but did not reach statistical significance (p = 0.06). Soluble ACE2 showed the strongest positive correlation with the ADAM-17 substrate score, follow by renin, interleukin-6, and lung injury biomarkers. The ratio of ADAM-17 to ACE2 gene expression was highest in the lung. This study indicates that increased ADAM-17 activity is associated with severe COVID-19. Our findings also indicate that there may a bidirectional relationship between membrane-bound ACE2 shedding via increased ADAM-17 activity, dysregulated renin–angiotensin system (RAS) and immune signaling. Additionally, differences in ACE2 and ADAM-17 gene expressions between different tissues may be of importance in explaining why the lung is the organ most severely affected by COVID-19, but this requires further evaluation in prospective studies.
... Timely intervention and risk stratification are paramount to improving respiratory management [110]. Key potential biomarkers include urinary desmosine levels, indicative of ECM degradation and lung damage; surfactant proteins (SPs) reflecting alveolar-capillary barrier integrity; inflammatory cytokines such as IL-6 and IL-8, highlighting inflammation; and plasma biomarkers such as angiopoietin-2 and receptor for advanced glycation end-products (RAGE), associated with endothelial and epithelial cell damage, respectively [111]. These biomarkers could revolutionize the management of patients at risk for ARDS by enabling personalized, lung-protective strategies and dynamic care adjustments [112]. ...
Article
Full-text available
Background Mechanical ventilation, a lifesaving intervention in critical care, can lead to damage in the extracellular matrix (ECM), triggering inflammation and ventilator-induced lung injury (VILI), particularly in conditions such as acute respiratory distress syndrome (ARDS). This review discusses the detailed structure of the ECM in healthy and ARDS-affected lungs under mechanical ventilation, aiming to bridge the gap between experimental insights and clinical practice by offering a thorough understanding of lung ECM organization and the dynamics of its alteration during mechanical ventilation. Main text Focusing on the clinical implications, we explore the potential of precise interventions targeting the ECM and cellular signaling pathways to mitigate lung damage, reduce inflammation, and ultimately improve outcomes for critically ill patients. By analyzing a range of experimental studies and clinical papers, particular attention is paid to the roles of matrix metalloproteinases (MMPs), integrins, and other molecules in ECM damage and VILI. This synthesis not only sheds light on the structural changes induced by mechanical stress but also underscores the importance of cellular responses such as inflammation, fibrosis, and excessive activation of MMPs. Conclusions This review emphasizes the significance of mechanical cues transduced by integrins and their impact on cellular behavior during ventilation, offering insights into the complex interactions between mechanical ventilation, ECM damage, and cellular signaling. By understanding these mechanisms, healthcare professionals in critical care can anticipate the consequences of mechanical ventilation and use targeted strategies to prevent or minimize ECM damage, ultimately leading to better patient management and outcomes in critical care settings.
... Our findings indicated that AZC exhibited a protective role by mitigating pulmonary edema, leading to reductions in total protein levels in the BALF, overall infiltration of inflammatory cells, lung wet-to-dry (W/D) weight ratio, and hyperpermeability caused by PM2.5 exposure (Fig. 1-3). Assessments of polymorphonuclear leukocyte activation and oxidative stress were carried out using MPO levels and NO generation (Blondonnet et al., 2016). PM-induced pulmonary damage is characterized by the activation of the TLR4-MyD88 pathway, which triggers the release of inflammatory mediators like IL-1β and TNF-α, initiating an inflammatory cascade that leads to neutrophil migration to alveoli and lung injury (Gu et al., 2017). ...
Article
Particulate matter (PM) constitutes a hazardous blend of organic and inorganic particles that poses health risks. Inhalation of fine airborne PM with a diameter of ≤ 2.5 μm (PM2.5) can lead to significant lung impairments. (+)-afzelechin (AZC), a natural compound sourced from Bergenia ligulata, boasts a range of attributes, including antioxidant, antimicrobial, anticancer, and cardiovascular effects. However, knowledge about the therapeutic potential of AZC for patients with PM2.5-induced lung injuries remains limited. Thus, in this study, we investigated the protective attributes of AZC against lung damage caused by PM2.5 exposure. AZC was administered to the mice 30 min after intratracheal instillation of PM2.5. Various parameters, such as changes in lung tissue wet/dry (W/D) weight ratio, total protein/total cell ratio, lymphocyte counts, levels of inflammatory cytokines in bronchoalveolar lavage fluid (BALF), vascular permeability, and histology, were evaluated in mice exposed to PM2.5. Data demonstrated that AZC mitigated lung damage, reduced W/D weight ratio, and curbed hyperpermeability induced by PM2.5 exposure. Furthermore, AZC effectively lowered plasma levels of inflammatory cytokines produced by PM2.5 exposure. It reduced the total protein concentration in BALF and successfully alleviated PM2.5-induced lymphocytosis. Additionally, AZC substantially diminished the expression levels of Toll-like receptors 4 (TLR4), MyD88, and autophagy-related proteins LC3 II and Beclin 1. In contrast, it elevated the protein phosphorylation of the mammalian target of rapamycin (mTOR). Consequently, the anti-inflammatory attribute of AZC positions it as a promising therapeutic agent for mitigating PM2.5-induced lung injuries by modulating the TLR4-MyD88 and mTOR-autophagy pathways.
... Increased expression of VEGF by alveolar macrophages invigorates their phagocytic activity towards apoptotic cells [86]. Also, VEGF-A has been shown to enhance the air-blood barrier permeability and thereby increase the risks of pulmonary edema [87]. The increase in transcription levels of Vegfa in response to the treatment with M2-polarized macrophages, revealed by us in this study, may indicate the efferocytosis activation; on the other hand, it may be correlated with increased permeability of the blood-brain barrier and the appearance of perivascular lymphoid sleeves revealed histologically. ...
Article
Full-text available
Innate immunity reactions are core to any immunological process, including systemic inflammation and such extremes as acute respiratory distress syndrome (ARDS) and cytokine storm. Macrophages, the key cells of innate immunity, show high phenotypic plasticity: depending on microenvironmental cues, they can polarize into M1 (classically activated, pro-inflammatory) or M2 (alternatively activated, anti-inflammatory). The anti-inflammatory M2 macrophage polarization-based cell therapies constitute a novel prospective modality. Systemic administration of ‘educated’ macrophages is intended at their homing in lungs in order to mitigate the pro-inflammatory cytokine production and reduce the risks of ‘cytokine storm’ and related severe complications. Acute respiratory distress syndrome (ARDS) is the main mortality factor in pneumonia including SARS-CoV-associated cases. This study aimed to evaluate the influence of infusions of RAW 264.7 murine macrophage cell line polarized towards M2 phenotype on the development of LPS-induced ARDS in mouse model. The results indicate that the M2-polarized RAW 264.7 macrophage infusions in the studied model of ARDS promote relocation of lymphocytes from their depots in immune organs to the lungs. In addition, the treatment facilitates expression of M2-polarization markers Arg1, Vegfa and Tgfb and decreases of M1-polarization marker Cd38 in lung tissues, which can indicate the anti-inflammatory response activation. However, treatment of ARDS with M2-polarized macrophages didn't change the neutrophil numbers in the lungs. Moreover, the level of the Arg1 protein in lungs decreased throughtout the treatment with M2 macrophages, which is probably because of the pro-inflammatory microenvironment influence on the polarization of macrophages towards M1. Thus, the chemical polarization of macrophages is unstable and depends on the microenvironment. This adverse effect can be reduced through the use of primary autologous macrophages or some alternative methods of M2 polarization, notably siRNA-mediated.
... The study on biomarkers for ARDS may help us to understand the cellular and molecular mechanism underlying different ARDS treatment options, as well as the possible proinflammatory effects. [30]. Toll-like receptors (TLRs) are pattern recognition receptors (PRRs) that play a role in both adaptive and innate immunity. ...
Article
Full-text available
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury that is characterized by the critical onset of pulmonary edema and severe hypoxemia because of failure in pulmonary gas exchange. Severe hypoxemia due to pulmonary gas exchange failure was first recognized in the 1960s. ARDS arises utmost often in patients suffering from sepsis, aspiration of gastric contents, pneumonia or severe trauma and occupies in approximately 10% of intensive care units patients worldwide. High-throughput gene profiling has been reported as a powerful tool in revealing several chief pathways linked with the pathogenesis of ARDS such as: MyD88-dependent pathway, TNF signaling pathway and MAPK signaling pathway, etc. There are certain proteins that majorly get overexpressed during ARDS. Among them, few of the primarily reported genes are: ICAM-1, TLR-4, TNF-α, and IL-8 have been found to be closely associated with the pathogenesis of ARDS. Distinct expression of various proteins has been observed from the MyD88-dependent pathway, TNF signaling pathway and MAPK signaling pathway which explicitly facilitate the pathogenesis of ARDS in human subjects. Analysis of these overexpressed proteins in different combinations can be used as an indication of the progression of the syndrome. This review article mainly focuses on analyzing the overexpression of these biomarker proteins in the lungs which lead to ARDS mediated inflammation.
Preprint
Full-text available
Purpose Acute respiratory distress syndrome (ARDS) is a heterogeneous syndrome with substantial morbidity and mortality globally. Body of evidence revealed that the epidemiologic estimates are currently disproportional due to differences in patient populations, risk factors, resources, and practice protocols around the world, and the rate of mortality and its predictors are uncertain in Ethiopia. Method A multi-centre longitudinal study was conducted in Ethiopia from January 2018 to June 2023. After receiving ethical clearance from the Institutional Review Board (IRB) of Dilla University College of Health Science and Medicine, 356 ARDS patients’ records were retrieved with a systematic random sampling technique. A multilevel multivariate analysis was used to control the effect of clustering. A P < 0.05 was taken as statistically significant. Results This study demonstrated that the cumulative mortality rate of patients with ARDS was 59% (95% CI: 53.5 to 63.9). The multilevel multivariable model analysis showed that GCS < 8 (AOR = 7.4; 95% CI: 2.79, 19.75), severe form of ARDS (AOR 4.7 95% CI 1.64, 13.36), invasive ventilation (AOR 3.2, 95% CI 1.56, 6.42), and respiratory comorbidity (AOR 4.9, 95% CI 1.71, 14.32) were independent predictors of in-hospital mortality among patients with ARDS. Conclusion The study revealed that the hospital mortality rate was substantially higher than that of developed nations. The study also highlighted various risk factors that independently predicted in-hospital mortality.The findings of this study call for mitigating strategies to improve ICU care for ARDS patients.
Article
Sepsis-induced acute lung injury (ALI) poses a common and formidable challenge in clinical practice, currently lacking efficacious therapeutic approaches. This study delves into the evaluation of (+)-afzelechin (AZC), a natural compound derived from Bergenia ligulata with a diverse array of properties, encompassing antioxidant, anticancer, antimicrobial, and cardiovascular effects to ascertain its effectiveness and underlying mechanisms in mitigating sepsis-induced ALI through animal experimentation. An ALI mouse model induced by sepsis was established through lipopolysaccharide (LPS) administration, and various analytical techniques, including quantitative real-time polymerase chain reaction, Western blotting, and enzyme-linked immunosorbent assay were employed to gauge inflammatory cytokine levels, lung injury, and associated signaling pathways. The animal experiments revealed that AZC offered safeguards against lung injury induced by LPS while reducing inflammatory cytokine levels in both blood serum and lung tissue. Western blotting experiments revealed AZC's downregulation of the toll-like receptor (TLR)4/NF-κB pathway and the upregulation of PI3K/Akt, coupled with inhibition of the Hippo and Rho signaling pathways. These findings underscore AZC's efficacy in ameliorating sepsis-induced ALI by modulating cytokine storms and curtailing inflammation via the regulation of TLR4/NF-κB, PI3K/Akt, Hippo, and Rho signaling pathways. This work serves as a foundation for additional exploration into AZC's mechanisms and its potential as a therapy for sepsis-induced ALI. Animals in accordance with Kyungpook National University (IRB No. KNU 2022-174).
Article
Full-text available
Acute respiratory distress syndrome (ARDS) is an acute diffuse inflammatory lung injury characterized by the damage of alveolar epithelial cells and pulmonary capillary endothelial cells. It is mainly manifested by non-cardiogenic pulmonary edema, resulting from intrapulmonary and extrapulmonary risk factors. ARDS is often accompanied by immune system disturbance, both locally in the lungs and systemically. As a common heterogeneous disease in critical care medicine, researchers are often faced with the failure of clinical trials. Latent class analysis had been used to compensate for poor outcomes and found that targeted treatment after subgrouping contribute to ARDS therapy. The subphenotype of ARDS caused by sepsis has garnered attention due to its refractory nature and detrimental consequences. Sepsis stands as the most predominant extrapulmonary cause of ARDS, accounting for approximately 32% of ARDS cases. Studies indicate that sepsis-induced ARDS tends to be more severe than ARDS caused by other factors, leading to poorer prognosis and higher mortality rate. This comprehensive review delves into the immunological mechanisms of sepsis-ARDS, the heterogeneity of ARDS and existing research on targeted treatments, aiming to providing mechanism understanding and exploring ideas for accurate treatment of ARDS or sepsis-ARDS.
Article
Full-text available
Background: The main soluble form of the receptor for advanced glycation end-products (sRAGE) is elevated during acute respiratory distress syndrome (ARDS). However other RAGE isoforms and multiple ligands have been poorly reported in the clinical setting, and their respective contribution to RAGE activation during ARDS remains unclear. Our goal was therefore to describe main RAGE isoforms and ligands levels during ARDS. Methods: 30 ARDS patients and 30 mechanically ventilated controls were prospectively included in this monocenter observational study. Arterial, superior vena cava and alveolar fluid levels of sRAGE, endogenous-secretory RAGE (esRAGE), high mobility group box-1 protein (HMGB1), S100A12 and advanced glycation end-products (AGEs) were measured in duplicate ELISA on day 0, day 3 and day 6. In patients with ARDS, baseline lung morphology was assessed with computed tomography. Results: ARDS patients had higher arterial, central venous and alveolar levels of sRAGE, HMGB1 and S100A12, but lower levels of esRAGE and AGEs, than controls. Baseline arterial sRAGE, HMGB1 and S100A12 were correlated with nonfocal ARDS (AUC 0.79, 0.65 and 0.63, respectively). Baseline arterial sRAGE, esRAGE, S100A12 and AGEs were associated with severity as assessed by PaO2/FiO2. Conclusions: This is the first kinetics study of levels of RAGE main isoforms and ligands during ARDS. Elevated sRAGE, HMGB1 and S100A12, with decreased esRAGE and AGEs, were found to distinguish patients with ARDS from those without. Our findings should prompt future studies aimed at elucidating RAGE/HMGB1/S100A12 axis involvement in ARDS. Trial registration: clinicaltrials.gov Identifier: NCT01270295.
Article
The soluble form of the receptor for advanced glycation end-products (sRAGE) is elevated and correlated with severity in patients with acute respiratory distress syndrome (ARDS). The impact of ventilator settings on plasma levels of sRAGE, in patients with or without pre-existing lung injury, remains under-investigated to date. Our objective was to assess the effects of a lung-protective ventilation strategy (combining low tidal volume, positive end-expiratory pressure and recruitment maneuvers), as compared with a non-protective approach (with high tidal volume and zero end-expiratory pressure), on plasma levels of sRAGE in patients without lung injury undergoing major abdominal surgery. Plasma samples were obtained from 95 patients enrolled in a large randomized controlled trial of lung-protective ventilation for major abdominal surgery. Plasma levels of sRAGE were measured in duplicate with an enzyme-linked immunoassay on day 1, immediately after surgery, and on postoperative days 1, 3 and 7. Early postoperative plasma levels of sRAGE were significantly lower in the lung-protective ventilation group (n = 47) than in the non-protective ventilation group (n = 48) (mean (standard deviation), 1782 (836) vs 2171 (1678) pg/mL, respectively, P = 0.03). Intraoperative changes in plasma sRAGE were associated with postoperative hypoxemia and ARDS. A lung-protective ventilation strategy decreased plasma sRAGE in patients without lung injury undergoing major abdominal surgery compared with the patients with non-protective ventilation. This intraoperative decrease could reflect a lesser degree of epithelial injury. © 2015 Asian Pacific Society of Respirology.