You are on page 1of 11

Am J Physiol Lung Cell Mol Physiol 307: L681–L691, 2014.

First published September 26, 2014; doi:10.1152/ajplung.00014.2014. Review

CALL FOR PAPERS Biomarkers in Lung Diseases: from Pathogenesis to Prediction


to New Therapies

Molecular biomarkers in idiopathic pulmonary fibrosis


Brett Ley,1 Kevin K. Brown,2 and Harold R. Collard1
1
Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco,
San Francisco, California; and 2Department of Medicine, National Jewish Health and the University of Colorado,
Denver, Colorado
Submitted 17 January 2014; accepted in final form 12 August 2014

Ley B, Brown KK, Collard HR. Molecular biomarkers in idiopathic pulmonary


fibrosis. Am J Physiol Lung Cell Mol Physiol 307: L681–L691, 2014. First
published September 26, 2014; doi:10.1152/ajplung.00014.2014.—Molecular bio-
markers are highly desired in idiopathic pulmonary fibrosis (IPF), where they hold
the potential to elucidate underlying disease mechanisms, accelerated drug devel-
opment, and advance clinical management. Currently, there are no molecular
biomarkers in widespread clinical use for IPF, and the search for potential markers
remains in its infancy. Proposed core mechanisms in the pathogenesis of IPF for
which candidate markers have been offered include alveolar epithelial cell dys-
function, immune dysregulation, and fibrogenesis. Useful markers reflect important
pathological pathways, are practically and accurately measured, have undergone
extensive validation, and are an improvement upon the current approach for their
intended use. The successful development of useful molecular biomarkers is a
central challenge for the future of translational research in IPF and will require
collaborative efforts among those parties invested in advancing the care of patients
with IPF.
biomarker; diagnosis; prediction; pulmonary fibrosis

IDIOPATHIC PULMONARY FIBROSIS (IPF) is a chronic fibrotic lung biological tissue or fluids from patients with IPF. At their core,
disease of unknown cause that generally affects adults over the molecular biomarkers should reflect and inform the underlying
age of 50 years (75). On surgical lung biopsy, it is characterized biological mechanisms involved in a disease. This article aims to
by the underlying histopathological pattern of usual interstitial provide a framework for thinking about molecular biomarker
pneumonia (UIP) in the absence of secondary causes or associa- development for IPF, using three core mechanistic themes rele-
tions (i.e., idiopathic UIP). IPF has a poor prognosis with an vant to IPF to review the steps necessary to the development of
average survival of 3–5 years, but there is appreciable heteroge- molecular biomarkers and to describe how the current crop of
neity among individual patients in disease course. At present, molecular biomarkers perform.
there are no FDA-approved medications for IPF, leaving limited
therapeutic options for patients with progressive disease. DEVELOPMENT OF MOLECULAR BIOMARKERS FOR IPF
In categorizing patients with IPF, clinicians and researchers
In theory, molecular biomarkers may be used in IPF in several
currently rely on clinical data — history and physical exam,
ways. These include identifying patients at risk for developing IPF
radiology, pulmonary function tests, and histopathological pat- (predisposition biomarkers); making the diagnosis of IPF (diag-
tern — that do not directly reflect the underlying pathobiologi- nostic or screening biomarkers); determining a patient’s baseline
cal mechanisms of the disease. We are unable, therefore, to prognosis, staging disease severity, and monitoring for progres-
adequately subphenotype patients with IPF who may have sion (prognostic biomarkers); and identifying target engagement
quantitatively or qualitatively different biological mechanisms with a specific mechanistic response or predicting response or
responsible for their underlying disease. This is a major limi- toxicity to therapy, either by identifying a specific subgroup most
tation to both clinical care and research in the field. likely to respond to a therapy (therapeutic, predictive, or compan-
In this article, we explore the field of molecular biomarkers, ion biomarker) or by substituting for a clinically meaningful
which we use to refer to any objectively quantifiable substance at outcome/end point (surrogate biomarker/end point) (10, 23). Cur-
the cellular level or smaller (e.g., protein) that can be measured in rently, there are no molecular biomarkers in widespread clinical
use for IPF for any of these uses.
Address for reprint requests and other correspondence: H. R. Collard, 505
There are two fundamentally different approaches to bio-
Parnassus Ave., Box 0111, San Francisco, CA 94143 (e-mail: hal.collard marker discovery, which both have value. In the hypothesis-
@ucsf.edu). based approach, a candidate marker is selected a priori on the
http://www.ajplung.org 1040-0605/14 Copyright © 2014 the American Physiological Society L681
Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
L682 MOLECULAR BIOMARKERS IN IPF

basis of preexisting rationale/evidence. This is the approach olds, inter- and intrasubject variability, and predictive perfor-
that has been taken in the majority of biomarker studies in IPF mance for the biomarker’s intended use are measured with the
to date. This approach has the advantage of a strong biological appropriate statistical tests. Although techniques to reduce
rationale and preliminary data, but it suffers from a lack of optimism (e.g., cross-validation, bootstrap resampling) can be
efficiency in the discovery process. The unbiased or hypothe- used for “internal validation,” true validation (external valida-
sis-free approach utilizes systems biology methods (e.g., tion) involves replicating results in one or more separate
genomic, transcriptomic, proteomic, and integratomic) to cohorts (ideally prospective and multicentered).
screen a large number of candidate markers for their associa- If considered for use in clinical practice, molecular biomark-
tion with the disease or outcome of interest, greatly increasing ers should improve upon current clinical practice compared
the efficiency and scope of the discovery process but increasing with non-biomarker-driven strategies and their use should
the risk of false discovery. ultimately lead to improved patient outcomes. For example, a
Most fundamentally, molecular biomarkers should reflect novel prognostic biomarker should add statistical performance
the presence and activity of relevant pathobiological processes to well-established prognostic variables (e.g., pulmonary func-
or mechanisms. In addition, biomarker measurement should be tion tests), and its measurement should lead to alterations in
simple, technically accurate, and broadly reproducible and clinical management (e.g., change of therapy, referral for lung
have acceptable risk. Therefore, measurement from easily transplantation). If molecular biomarkers do not have all of
obtainable body fluids or tissues is preferred [i.e., blood/serum, these clinical characteristics, they still may add research value
urine, exhaled breath condensates ⬎ bronchoalveolar lavage by providing insights into disease biology. Figure 1 outlines
fluid (BALF) ⬎ transbronchial biopsy ⬎ surgical lung biopsy]. the ideal qualities of molecular biomarkers in IPF.
Molecular biomarkers require broad validation across sexes,
ages, ethnicities, and disease severity to assure generalizability CANDIDATE MOLECULAR BIOMARKERS IN IPF
(13, 53). Practically, validation of the biomarker begins with a
derivation (or discovery) cohort of appropriate size and rele- Over the past 15 years, the pathogenic paradigm for IPF
vance to its proposed role, in which reference ranges, thresh- has shifted from one of uncontrolled inflammation toward

Fig. 1. Ideal qualities of molecular biomarkers for idiopathic pulmonary fibrosis. BAL, bronchoalveolar lavage; IPF, idiopathic pulmonary fibrosis; ILD,
interstitial lung disease.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
MOLECULAR BIOMARKERS IN IPF L683
one of alveolar epithelial cell dysfunction, immune dysregu- Core Pathway 1: Alveolar Epithelial Cell Dysfunction
lation, and fibroproliferation/fibrogenesis/matrix remodel- Surfactant proteins. Surfactant proteins are synthesized
ing (39, 85). In this paradigm, alveolar epithelial stress and and secreted by type II alveolar epithelial cells (AECs).
dysfunction results in activation of profibrotic signaling Their functions include facilitating surfactant function and
pathways, fibroblast proliferation, and exuberant extracellu- transport and innate host defense. Quantitative or qualitative
lar matrix deposition (39, 85). The resulting tissue destruc- abnormalities in surfactant proteins are hypothesized to
tion and architectural distortion eventually lead to organ increase alveolar epithelial endoplasmic reticulum (ER)
dysfunction (e.g., restricted ventilation, hypoxemia), dis- stress and trigger the unfolded protein response (UPR) (94).
ability (shortness of breath and exercise limitation), and Both mutations in genes encoding surfactant proteins and
death from respiratory failure. In the remainder of this the level of surfactant proteins in blood and the extracellular
article, we use these proposed core mechanistic pathways to lining fluid of the lung in patients have been proposed as
discuss and contextualize candidate biomarkers (Fig. 2). potential molecular biomarkers of IPF.
Table 1 demonstrates the level of evidence supporting a MUTATIONS IN GENES ENCODING SURFACTANT PROTEINS. Muta-
clinical role for each candidate biomarker. tions in surfactant protein genes, especially A2 and C, have

Infection (viral), microaspiration, inhalational (tobacco)

SPC ER stress
SPA2 MUC5B
Alveolar
TERT/TERC Epithelial cell TOLLIP
UPR macrophage
Short telomeres dysfunction ELMOD2
TLR3
Apoptosis

Th2 Lymphocytes
ECM
SPA, SPD, Epithelial to Collagen
CXCL13
KL6/MUC1, mesenchymal
TGFβ CCL18
α-defensins, transition
cCK18, MMP7, Fibrogenesis OPN
Immune dysregulation
MMP1, OPN YKL40
Fibroblasts and
myofibroblasts TLR3?
OPN
Periostin
CD28
Anti-
CD4 CD25
OPN CD4 CD25 HSP70
Periostin IgG
CD45
CD34
CCL18 CD4+
OPN FOXP3 T cell
Coll1 Sema7a FOXP3 Treg
Leukocytes Fibrocyte YKL40
Blood proteins Fibronectin Sema7a
SPA, SPD,
KL6/MUC1,
α-defensins,
cCK18, YKL40, Genomic Blood cells
Anti-HSP70 IgG, SPC TERT/TERC CD28+ CD4+ T cells
CXCL13, MMP7, TOLLIP SPA2 Tregs
MMP1, OPN, TLR3 Telomere length Semaphorin 7a+ T regs
Periostin MUC5B ELMOD2 Fibrocytes

Fig. 2. Core mechanisms and candidate molecular biomarkers for idiopathic pulmonary fibrosis (see text and Table 1). Pictured is the alveolar space (top, light blue),
epithelial cell layer (tan), interstitial space (white), and capillary (bottom, red). Injury to the alveolar epithelium from a variety of inciting factors (dark blue box) leads
to epithelial cell dysfunction, fibrogenesis/extracellular matrix (ECM) deposition, and immune dysregulation. Proposed molecular biomarkers of these processes include
blood proteins, genomic markers, and blood cells (see light green boxes). cCK18, cleaved cytokeratin 18; CXCL, C-X-C chemokine ligand; ELMOD2, ELMO
containing domain 2 gene mutations; EMT, epithelial-to-mesenchymal transition; ER, endoplasmic reticulum; HSP, heat shock protein; MMP, matrix metalloproteinase;
KL6/MUC1, Krebs von den Lungen 6/mucin 1; MUC5B, mucin 5B promoter polymorphism; OPN, osteopontin; SPA, surfactant protein A; SPA2, surfactant protein
A2 gene mutations; SPC, surfactant protein C gene mutations; SPD, surfactant protein D; TERT/TERC, telomerase gene mutations; TGF, transforming growth factor;
TLR3, Toll-like receptor 3 gene mutation; TOLLIP, Toll-interacting protein gene mutations; Treg, regulatory T cell; UPR, unfolded protein response.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
L684 MOLECULAR BIOMARKERS IN IPF

Table 1. Candidate molecular biomarkers for idiopathic and D (SPA and SPD), have been evaluated as diagnostic and
pulmonary fibrosis and the strength of evidence supporting prognostic molecular biomarkers in IPF. Because surfactant
their clinical role proteins are synthesized and secreted by type II AECs, their
levels in extracellular fluids may reflect alterations in produc-
Biomarker Predisposition Diagnosis Prognosis Therapeutic
tion and secretion by abnormal type II AECs, increased per-
Genomic meability of the epithelial-endothelial barrier, and/or type II
SPC ⫾ AEC injury leading to nonsecretory release. Gene expression
SPA2 ⫾
MUC5B ⫾ ⫹
profiling in IPF lungs demonstrates overexpression of the
TERT/TERC ⫾ SPA1 gene in some patients with progressive disease compared
Telomere length ⫾ ⫾ with those with relatively stable disease, lending support to
ELMOD2 ⫾ altered surfactant production in patients with progressive IPF
TOLLIP ⫾ ⫾ (11). Both SPA and SPD levels are decreased in BALF of
TLR3 ⫺ ⫾
Blood proteins patients with IPF compared with healthy controls but are
SPA ⫾ ⫾ ⫺ similar to other interstitial lung diseases (ILDs) including
SPD ⫾ ⫾ ⫺ nonspecific interstitial pneumonia (NSIP) (8, 34). Conversely,
KL6/MUC1 ⫾ ⫾ ⫺ serum levels of both SPA and SPD are elevated in IPF
␣-Defensins ⫾
cCK18 ⫾
compared with healthy controls (28, 34, 62). In small studies,
CCL18 ⫾ serum levels of SPA and/or SPD were significantly higher in
YKL40 ⫾ subjects with IPF compared with other ILDs, but with insuf-
Anti-HSP70 IgG ⫾ ficient evidence to establish them as diagnostic markers (28,
CXCL13 ⫾ 34, 62). Serum SPD levels have been reported to be signifi-
MMP7 ⫾ ⫹
MMP1 ⫾ ⫾ cantly elevated in acute exacerbations of IPF (AE-IPF) com-
OPN ⫾ pared with patients with stable IPF, suggesting that type II
Periostin ⫾ AECs may play a direct or indirect role in the pathobiology of
Blood cells AE-IPF (18). Prognostically, higher serum levels of both SPA
CD28% CD4⫹
T cells ⫾
and SPD have been associated with reduced survival in IPF
Tregs ⫾ independent of clinical variables (8, 28, 38, 92). However, in a
Semaphorin 7a⫹ recent prediction model study including 118 IPF patients,
Tregs ⫾ neither SPA nor SPD significantly improved discrimination of
Fibrocytes ⫾ survival times when added to a clinical model [age, forced vital
Key: No molecular or cellular biomarkers have a proven clinical role in IPF, capacity (FVC), lung CO-diffusing capacity (DLCO), and
but the strength of evidence supporting a potential role is denoted as follows: ⫹, change in FVC] (89). In two prospective treatment trials of
Consistent or strong evidence supporting potential role; ⫾, conflicting or pirfenidone, longitudinal changes in SPA and SPD levels were
inadequate evidence to support potential role; ⫺, consistent or strong evidence
against a potential role; blank, no evidence. SPC, surfactant protein C gene not significantly different between treatment and placebo
mutations; SPA2, surfactant protein A2 gene mutations; MUC5B, mucin 5B groups (7, 93). In sum, serum levels of SPA and SPD may be
promoter polymorphism; TERT/TERC, telomerase gene mutations; ELMOD2, markers of abnormal function, injury, apoptosis, or prolifera-
ELMO containing domain 2 gene mutations; TOLLIP, Toll-interacting protein tion of type II AECs, but their measurement has yet to be
gene mutations; TLR3, toll-like receptor 3 gene mutation; SPA, surfactant
protein A; SPD, surfactant protein D; KL6/MUC1, Krebs von den Lungen
proven clinically useful in making the diagnosis or predicting
6/mucin 1; cCK18, cleaved cytokeratin 18; HSP, heat shock protein; CXCL, prognosis in patients with IPF. Whether serial measurements
C-X-C motif chemokine; MMP, matrix metalloproteinase; OPN, osteopontin; can reflect disease activity over time is unknown.
Treg, regulatory T cell. KL6/MUC1. Krebs von den Lungen-6 (KL6)/mucin 1
(MUC1) is a large membrane-bound glycoprotein in the mucin
family expressed on the extracellular surface of type II AECs
provided important insights into the pathogenesis of IPF (4, 14, and bronchiolar epithelial cells in the lung, as well as glandular
15, 17, 45– 47, 50, 52, 57, 59, 66, 86, 94, 95, 97, 99, 101, 103). epithelial cells in other tissues including pancreas and breast
For example, some mutations have been shown to be associ- (35). Originally investigated as a potential tumor marker for
ated with the development of pulmonary fibrosis and with the adenocarcinomas, it has since been studied extensively as a
induction of chronic ER stress and the UPR in AECs (14, 15, diagnostic and prognostic biomarker in ILDs. Expression of
45, 50, 57). In animal models, a second profibrotic stimulus KL6 is increased in affected lung and regenerating type II
(e.g., viral infection, bleomycin) may be necessary for the AECs in a variety of ILDs including IPF (64). In vitro, KL6 has
development of fibrosis, suggesting that IPF may involve a also demonstrated chemotactic, proproliferative, and antiapo-
“two-hit” pathobiology, similar to that proposed for some ptotic effects on lung fibroblasts, suggesting a potential patho-
malignancies (15, 45). Because IPF is an uncommon condition genic role in pulmonary fibrosis (30, 63).
in the general population, and surfactant protein mutations are Serum levels of KL6 are significantly elevated in IPF and
rarely associated with sporadic IPF, the most likely role for the other ILDs compared with controls without ILD (34, 62).
use of surfactant protein mutations as molecular biomarkers in Elevated serum levels, especially greater than 1,000 U/ml, are
clinical practice would be identifying at-risk individuals within significantly associated with reduced survival in many ILDs,
families with pulmonary fibrosis known to have specific sur- including IPF (82, 107, 108). However, in the largest study of
factant protein mutations. IPF patients (n ⫽ 118), KL6 level at baseline demonstrated
SURFACTANT PROTEIN LEVELS. Surfactant protein levels mea- poor discrimination for survival time (C-statistic 0.58) and did
surable in BALF and blood, specifically surfactant proteins A not improve the prediction of survival beyond clinical predic-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
MOLECULAR BIOMARKERS IN IPF L685
tors (age, FVC, DLCO, and change in FVC) (89). In patients leading to AEC senescence and apoptosis, provide an attractive
with AE-IPF, serum KL6 was elevated compared with patients pathogenic link between aging and the development of IPF (19,
with stable IPF, supporting a role for type II AEC injury in 76). In 2007, heterozygous mutations in telomerase genes
AE-IPF (18). However, when evaluated longitudinally in pro- (TERT and TERC) were reported in 8 –15% of families with
spective treatment trials, changes in serum KL6 levels did not IPF and rarely in sporadic cases of IPF (6, 98). Although
track with treatment response (7, 93). In sum, KL6 may be a telomerase mutations are rare in sporadic cases of IPF, shorter
marker of type II AEC injury and may indicate worse survival telomeres are common in sporadic IPF compared with age-
among patients with ILDs, but it may not be specific to the matched controls (2). In fact, nearly one-quarter of patients
pathogenesis of IPF or useful for diagnosing or prognosticating with IPF have peripheral blood leukocyte telomere lengths in
in IPF beyond readily available clinical information. Whether the ⬍10th percentile (21). Short telomeres have also been
serial changes in KL6 prove useful for monitoring disease associated with risk of developing chronic obstructive pulmo-
progression and/or diagnosing acute exacerbation in IPF de- nary disease (COPD), another aging-related lung disease and
serves further study. thus may not be specific to the pathogenesis of IPF (80).
MUC5B. Genome-wide linkage scanning involving 82 fam- Recently, peripheral blood leukocyte telomere length has been
ilies with familial pulmonary fibrosis (FPF) followed by fine shown to be predictive of transplant-free survival in three
mapping of a region of interest on chromosome 11 in 575 separate cohorts of patients with IPF (total n ⫽ 342), indepen-
subjects with pulmonary fibrosis (83 FPF, 492 sporadic IPF) dent of other prognostic factors (age, sex, FVC, and DLCO)
and 322 controls, identified a common single nucleotide poly- (90). Pending further validation and delineation of normal and
morphism (SNP) in the putative promoter region of the mucin threshold values, peripheral blood telomere length holds prom-
5B (MUC5B) gene (rs35705950) that was highly associated ise as a mechanistic biomarker that may help predict predis-
with disease (83). The association has been confirmed in position to disease and disease prognosis.
independent cohorts from the US and Europe (12, 109). In cCK18. In exploring the putative role of ER stress-UPR-
total, the minor allele occurred in 34% of FPF, 34 – 42% of induced AEC apoptosis in the pathogenesis of IPF, a marker of
sporadic IPF, and 9 –11% of control subjects. In the European the activity of this process would be highly desirable. Cyto-
study, no association was found with systemic sclerosis-asso- keratin 18 is a cytoskeletal protein found in AECs (and other
ciated ILD, suggesting potential specificity for IPF (12). In a epithelial cells). During epithelial cell apoptosis, cytokeratin 18
population-based study, there was an allele-dose-dependent is cleaved by caspases yielding a fragment, cleaved cytokeratin
association with an increased prevalence of interstitial lung 18 (cCK18), which is detectable by a specific antibody. In a
abnormalities on chest imaging by computerized tomography recent study, cCK18 was detected by immunohistochemistry in
in individuals without clinically recognized ILD (i.e., subclin- AECs of IPF lungs but not in normal lung tissue (16). In vitro,
ical ILD), lending further epidemiological support for these cCK18 and mediators of the UPR increased following induc-
genetic polymorphisms in MUC5B as a risk factor for the tion of ER stress in type II AECs. cCK18 was significantly
development of pulmonary fibrosis (32). elevated in the serum of IPF patients compared with normal
In addition to its association with an increased risk of controls and patients with hypersensitivity pneumonitis (HP)
developing IPF, the MUC5B promoter polymorphism appears and NSIP, distinguishing IPF from HP/NSIP with moderate
to have prognostic value. In two independent IPF cohorts, one accuracy (AUROC 0.76). However, cCK18 level at baseline
drawn from clinical practice (n ⫽ 148) and one drawn from a was not associated with disease severity or outcomes in IPF.
large clinical trial (n ⫽ 438), the presence of the MUC5B Therefore, cCK18 may be mechanistically informative for the
mutation (measured by two different assays) was associated ER stress-UPR-AEC apoptosis in IPF, with elevated levels
with decreased mortality compared with the wild-type SNP, suggesting higher degrees of AEC apoptosis in IPF compared
independent of clinical factors (sex, FVC, and DLCO), and with other ILDs. Although serum cCK18 levels at baseline do
when included in a clinical model significantly improved the not appear to be useful for determining prognosis, serial levels
prediction of mortality (C-statistic increased from 0.68 – 0.69 have not been evaluated for measuring ongoing disease activity
to 0.71– 0.73) (70). It is unclear how to reconcile MUC5B’s and progression. Further study will be necessary to determine
association with increased risk of developing IPF and yet a whether cCK18 serum levels could serve as an efficacy marker
better prognosis among those with established IPF. (70). The for drugs intended to target ER stress-UPR-AEC apoptotic
mechanism by which the MUC5B promoter polymorphism pathways.
could cause or contribute to IPF is unknown. MUC5B is
overexpressed in the lung of subjects with IPF regardless of Core Pathway 2: Immune Dysregulation
allele status, as well as in unaffected carriers compared with
unaffected noncarriers, potentially implicating a direct role of Innate and adaptive immunity contribute to tissue injury and
the MUC5B protein in the pathogenesis of IPF (83). Elucidat- repair including fibrogenesis in many tissues (24). The contri-
ing a mechanistic role for MUC5B in IPF could lead to bution of the immune system to tissue injury and fibrosis in IPF
important advancements in our understanding of disease risk is controversial. Clinically, IPF patients do not respond to
and prognosis in a large proportion of familial and sporadic traditional immunosuppressive therapies. In fact, treatment of
cases. IPF with prednisone and azathioprine was associated with
Telomeres. Telomeres, the noncoding repetitive nucleotide increased mortality and hospitalizations in a recent random-
sequence at the ends of DNA that protect against degradation, ized-controlled trial (33). In addition, on histopathology, cel-
shorten with age (5). After telomeres reach a critical length, lular infiltration by inflammatory cells is not conspicuous in
activation of a p53-dependent checkpoint leads to apoptosis or IPF, consisting of minimal patchy lymphocytic and plasma cell
cellular senescence. Therefore, short telomeres, potentially infiltrates with occasional lymphoid aggregates in areas of

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
L686 MOLECULAR BIOMARKERS IN IPF

dense fibrosis (51, 75). A more nuanced look at immunity, Alveolar macrophage activation. CCL18. CC chemokine li-
however, suggests that there are important aspects of the IPF gand 18 (CCL18), also known as PARC (for pulmonary and
disease pathobiology that may be influenced by components of activation-regulated chemokine), is a chemokine protein pro-
the immune system. duced by alveolar macrophages that stimulates collagen pro-
Innate immunity. TLR3. Toll-like receptors (TLRs) are com- duction in pulmonary fibroblasts (40, 71, 72). In fibrotic ILDs
ponents of the innate immune system that are critical for including IPF, there are increased numbers of CCL18-positive
response to infection and tissue injury. The pathogenic role of macrophages (71). CCL18 gene expression is increased in
a functional SNP in the TLR3 gene (L412F) was recently alveolar macrophages derived from BALF, and this expression
investigated in IPF (61). Through a series of in vitro studies, correlates with impairment in pulmonary function (71, 72). In
activation of TLR3 in IPF lung fibroblasts with the L412F vitro, upregulation of CCL18 in alveolar macrophages occurs
mutation demonstrated reduced cytokine responses and dys- with Th2 cytokine stimulation (e.g., IL-13) and coculture with
regulated fibroproliferation compared with wild-type (61). fibroblasts via a ␤2-integrin/scavenger receptor pathway (71).
Also, TLR3null mice had increased collagen and profibrotic In turn, CCL18 derived from alveolar macrophages induces
cytokine production compared with wild type. In two indepen- fibroblast production of collagen, which appears independent
dent cohorts of IPF totaling 308 subjects, this mutation was of TGF-␤ signaling pathways (49, 71). These studies suggest a
associated with increased mortality independent of clinical positive feedback loop for collagen production in which Th2-
parameters (age, FVC, and DLCO), and in a clinical trial cohort activated alveolar macrophages [so called alternatively acti-
it was associated with accelerated physiological progression vated (M2) macrophages] produce CCL18, CCL18 stimulates
(61). The TLR3 SNP was common (32– 43% were heterozy- collagen production by pulmonary fibroblasts, and fibroblast-
gous, 6.5–11% were homozygous) and may be a factor leading derived collagen stimulates further CCL18 production by al-
to increased progression in established IPF. veolar macrophages (71). In 72 patients with IPF, serum
ELMOD2. Genome-wide linkage scanning of six multiplex
CCL18 concentration at baseline predicted subsequent physi-
FPF families from southeastern Finland, an area with cluster- ological progression and survival independent of other clinical
ing of FPF due to common ancestry, identified mutations in parameters (73). Therefore, CCL18 may be a mediator and
ELMOD2 (ELMO domain containing 2) that was associated marker of a fibrogenic pathway common to IPF and other
with decreased ELMOD2 mRNA expression in the lung (31). fibrotic ILDs, and its serum level may prove to be a useful
In the lung, ELMOD2 is expressed in macrophages and type II prognostic biomarker.
YKL40. YKL40 is a chitinase-like protein expressed in many
AECs. In vitro, AEC cell lines overexpressing ELMOD2
cell types that is involved in tissue response to injury and is
activated gene expression pathways involved in response to
dysregulated in many acute and chronic inflammatory condi-
viral infections, and ELMOD2 inhibition blunted antiviral
tions including COPD and asthma (48). Mechanisms by which
cytokine expression after TLR3 activation, thus supporting an
it may be involved in tissue fibrosis include mediating Th2
antiviral response role for ELMOD2 (74).
cell-IL-13 signaling pathways, inflammatory cell and fibroblast
TOLLIP. In a large genome-wide association study of IPF and
survival and proliferation, and alternative alveolar macrophage
control subjects, SNPs in the TOLLIP gene (Toll-interacting activation (48). YKL40 expression localizes to bronchiolar
protein) were associated with development of IPF and de- epithelial cells and alveolar macrophages adjacent to fibrotic
creased expression of TOLLIP protein (60). Counterintuitively, areas in IPF, and protein levels are elevated in the BALF and
one of the minor alleles was protective against the development serum of patients with IPF compared with healthy controls (25,
of IPF but associated with increased mortality among estab- 42). Elevated serum concentrations correlate with worse gas
lished IPF subjects. TOLLIP is an adapter protein with ubiq- exchange (25), and in a small single-center study both BALF
uitin-binding domains that modulates TLR, interleukin-1, and and serum levels were associated with survival in IPF (42).
TGF-␤ signaling through receptor trafficking and degradation Adaptive immunity. ANTI-HSP70 ANTIBODIES. In a study inves-
(110). tigating the role of antigen-specific autoimmunity in IPF, IgG
␣-DEFENSINS. The ␣-defensins are small antimicrobial pro- autoantibodies to heat shock protein (HSP)70 were identified in
teins secreted by neutrophils and epithelial cells that are in- the serum of 25% of patients with IPF compared with 3% of
volved in multiple functions of innate immunity (41). In a healthy controls (37). HSP70 protein, antibody-antigen com-
small study, plasma protein levels were elevated in IPF and plexes, and complement were common in IPF lung tissue.
correlated with measures of disease severity (56). More re- HSP70 protein has been shown to induce CD4 T cells from IPF
cently, global gene expression profiling demonstrated ␣-de- patients to proliferate and produce profibrotic cytokines (IL-4),
fensins 3 and 4 to be significantly upregulated in the blood and and anti-HSP70 antibodies isolated from IPF patients have
lung tissue of AE-IPF compared with stable IPF (41). In lung been shown to activate monocytes and induce IL-8 production.
tissue from AE-IPF, ␣-defensin proteins were localized to type In a small, single-center study, anti-HSP70 IgG positivity in
II AECs in the setting of widespread evidence of AEC apo- IPF patients was associated with physiological progression and
ptosis and proliferation. Peripheral blood gene expression pro- worse 1-year survival.
filing of 130 patients with IPF demonstrated that ␣-defensins 3 CXCL13. The importance of B cells in the pathogenesis of IPF
and 4 were among 13 genes whose expression levels distin- is unknown, but a potential role is suggested by the identifi-
guished severe from mild IPF as defined by impairment in cation of lymphoid aggregates in IPF lungs along with evi-
DLCO (106). Interestingly, ␣-defensins are activated by MMP7, dence for antibody dysregulation in some IPF patients (37, 51,
which is also highly expressed in type II AECs in IPF lungs 111). C-X-C motif chemokine 13 (CXCL13) is a chemokine
and may be important in disease pathogenesis (see section on important for homing CXCR5-expressing B lymphocytes into
MMPs below). lymphoid aggregates (100). In a recent study of 95 subjects

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
MOLECULAR BIOMARKERS IN IPF L687
with IPF, CXCL13 was significantly elevated in blood and MMPs are some of the most highly expressed genes in IPF
lung tissue compared with controls (healthy subjects and sub- lungs (87, 111). In IPF lungs, MMPs are primarily produced by
jects with COPD), and CXCL13 expression localized to lym- activated type II AECs, but a few are produced by fibroblasts
phoid aggregates in the lung (100). Both baseline levels of within fibroblast foci (39). MMP1 degrades type 1 and 3
CXCL13 and increases in CXCL13 over time were associated collagen and is expressed by type II AECs. MMP1 is increased
with reduced survival, independent of baseline demographic in lung tissue and BALF from patients with IPF, and serum
and physiological parameters. These findings require external levels of MMP1 are higher in IPF compared with HP, sarcoid-
validation but suggest a potential role for CXCL13 as prog- osis, and COPD (81). MMP7 is also produced by activated type
nostic biomarker in IPF, and the study authors theorize that II AECs and upregulated in IPF (67, 87, 111). It has broad
CXCL13 measurement could identify IPF patients who may be substrate activity including multiple basement membrane and
responsive to therapies that target B cells. ECM components, signaling molecules, and receptors, sug-
T CELL SUBSETS. T cells are the predominant mononuclear cell gesting a potential role in mediating an array of biological
type in IPF lungs, and their density in IPF lung tissue has been processes (68). MMP7 colocalizes in IPF AECs with osteo-
associated with disease severity and poor survival (69). CD4⫹ pontin (OPN), a proinflammatory and profibrotic cytokine (see
T cells express CD28, a costimulatory protein whose expres- next section on OPN below), and MMP7 expression/activation
sion may be lost after repetitive cycles of antigen-driven is induced by OPN, which in turn is cleaved and activated by
stimulation and T cell proliferation. CD4⫹ CD28null cells are MMP7 (1, 67). Similar to MMP1, serum protein levels of
therefore considered a marker of chronic adaptive immune MMP7 are higher in IPF compared with HP, sarcoidosis, and
activation (27). In a study involving 89 patients with IPF, the COPD (81). In one study, the combination of serum MMP1
proportion of CD28⫹ CD4 T cells in the peripheral blood and MMP7 levels was able to distinguish IPF from HP with
(CD28%) was low compared with healthy controls (27). Also, 96% sensitivity and 87% specificity (81). Serum MMP7 levels
low CD28% was correlated with low DLCO and reduced sur- also correlated with impairment in pulmonary function (FVC
vival. In a subgroup of patients with serial measurements, and DLCO) (81). In a candidate serum protein screening ap-
longitudinal changes in CD28% correlated with longitudinal proach in IPF patients, MMP7 concentration was identified as
changes in FVC (27). Clustering of peripheral blood mononu- significantly associated with survival and progression-free and
clear cell gene expression profiles in IPF patients identified a transplant-free survival in the discovery cohort (n ⫽ 140) and
subgroup with downregulated genes involved in the “costimu- transplant-free survival in the validation cohort (n ⫽ 101) (79).
latory signal during T cell activation” pathway including MMP7 was further included in a prognostic model along with
CD28, ICOS, LCK, and ITK (29). Quantitative expression clinical parameters (sex, FVC, and DLCO) derived from the
levels of these four genes correlated with the percentage of discovery cohort, demonstrating a good to excellent discrimi-
peripheral blood CD4⫹CD28⫹ T cells, and in two small native performance for survival in the validation cohort (C-
cohorts, improved prediction of transplant-free survival when statistic 0.73– 0.84) (89). However, model calibration was not
added to clinical predictors (29). These findings were con- presented. In a large clinical trial cohort of IPF subjects (n ⫽
firmed in a validation cohort and were not explained by 438), MMP7 was an independent predictor of survival in a
immunosuppression use. model including clinical parameters (sex, FVC, DLCO) and
Regulatory T cells (Tregs) play an important role in modu- MUC5B genotype (P ⫽ 0.04) (70). Change in serum MMP7
lation of the adaptive immune response. In a small study, Tregs level is currently being used as a primary efficacy outcome in
(CD4⫹CD25⫹FOXP3⫹) were reduced in the BALF and a phase 2 trial of inhaled carbon monoxide therapy for IPF
blood of patients with IPF compared with healthy controls and (clinicaltrials.gov NCT01214187).
other ILDs, and Treg suppressor function, not number, was Several other MMPs are of interest in the pathobiology
correlated with impairment in pulmonary function (43). Sema- of IPF. MMP3 expression is increased in IPF lungs, and
phorin 7a (Sema 7a) is a membrane protein that modulates MMP3null mice are protected from bleomycin-induced lung
lymphocyte function. Its expression on Tregs is increased in injury, suggesting a potential pathogenic role (104). In vitro,
the lung and peripheral blood of patients with IPF, and periph- MMP3 also cleaves and activates OPN (1), and exposure of
eral blood Sema 7a⫹ Tregs are elevated in patients with lung epithelial cells to MMP3 results in ␤-catenin signaling
rapidly progressive disease (78). Sema 7a⫹ Tregs have re- activation via cleavage of E-cadherin and subsequent activa-
duced expression of regulatory cytokines (e.g., IL-10) and tion of epithelial-to-mesenchymal transition (104). MMPs may
were able to induce fibrosis in a TGF-␤-overexpression mouse play a role in fibrocyte migration into lung tissue. Human
model. fibrocytes strongly express matrix metalloproteinases (MMPs)
2, 7, 8, and 9, where MMP8 specifically mediates migration
Core Pathway 3: Fibroproliferation, Fibrogenesis, through collagen 1 and MMP2 and 9 may facilitate migration
and Extracellular Matrix Remodeling through basement membrane components (26). MMP9 is ex-
pressed in phenotypically altered (Thy-1 negative) fibroblasts
MMPs. MMPs are a large family of zinc-containing metal- within fibroblast foci in response to TGF-␤1 stimulation (77),
lopeptidases that degrade multiple components of ECM, acti- and its activated form is increased in the BALF of patients
vate and degrade biological mediators (e.g., growth factors, with rapidly progressive compared with slowly progressive
chemokines, and cell surface receptors), and facilitate cell disease (84).
migration (68). There are 23 known MMPs expressed from 24 OPN. OPN is a secreted phosphorylated glycoprotein (also
gene loci, and most are secreted (68). MMPs are critically called secreted phosphoprotein 1 or SPP1) that functions as a
important in the homeostasis of the ECM, expressed at low mediator of inflammation and wound healing in many organs
levels in healthy tissue and upregulated in wound healing (68). and may play a role in multiple TGF-␤-mediated processes

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
L688 MOLECULAR BIOMARKERS IN IPF

(22, 102). Its expression is increased in IPF lungs and localizes data generated by systems biology research, which provides an
to AECs and macrophages (67, 91, 105, 111). In vitro, OPN unbiased approach to identifying and validating candidate
promotes migration and proliferation of fibroblasts, and its biomarkers and mechanistic pathways.
expression is increased by bleomycin-induced lung injury in IPF, like many other clinically defined diseases, is a biolog-
mice (67, 91). OPN induces upregulation of MMP7 in AECs ically heterogenous disease, involving multiple complex and
and colocalizes with MMP7 in AECs of IPF lungs (67). OPN interactive mechanisms, the relative importance of which may
is elevated in the BALF and plasma of IPF patients compared vary among individual patients. Molecular biomarkers could
with healthy controls, but plasma levels are not significantly someday soon lead to diagnostic reclassification and/or sub-
different from other ILDs (36, 67). In a mix of ILD including phenotyping of IPF patients on the basis of an individual
IPF, plasma OPN levels correlated with oxygenation but not patient’s relevant biology, directly informing treatment op-
pulmonary function parameters (36). OPN gene expression is tions. We believe that the development of clinically useful
increased in the lung tissue of IPF patients with progressive molecular biomarkers for IPF is the central challenge to trans-
disease compared with those with stable disease (11). lational research in the field over the next decade and that
Periostin. Periostin is an ECM and intracellular protein meeting this challenge will require collaborative efforts among
(so-called matricellular) that promotes ECM deposition, mes- those parties invested in advancing the care of patients with
enchymal cell proliferation, and wound closure and contributes IPF.
to fibrosis in multiple organs (44, 58). In mice, periostin is
ACKNOWLEDGMENTS
upregulated after bleomycin-induced lung injury, and blocking
periostin improves survival and limits collagen deposition. The authors wish to thank Dean Sheppard for thoughtful review of the
manuscript.
Periostinnull mice are protected from bleomycin-induced fibro-
sis. Periostin is increased in lung tissue of IPF patients and DISCLOSURES
localizes to fibroblasts, especially to fibroblasts in fibroblast H. R. Collard is a consultant to companies interested in the development of
foci (58, 65). Serum periostin levels are elevated in IPF and biomarkers in IPF and is partially funded by an NIH grant focused on
correlate with physiological progression (58, 65). In asthma, biomarker development in IPF.
periostin is secreted by bronchial epithelial cells in response to
AUTHOR CONTRIBUTIONS
IL-13 and can serve as a marker of IL-13 expression and
treatment response to IL-13 antagonists (20, 88). B.L., K.K.B., and H.R.C. conception and design of research; B.L., K.K.B.,
Circulating fibrocytes. Circulating fibrocytes are a subpop- and H.R.C. prepared figures; B.L., K.K.B., and H.R.C. drafted manuscript;
B.L., K.K.B., and H.R.C. edited and revised manuscript; B.L., K.K.B., and
ulation of leukocytes that express hematopoietic (CD45, H.R.C. approved final version of manuscript.
CD34) and mesenchymal (Coll 1, fibronectin) markers (9).
Fibrocytes are mesenchymal progenitor cells believed to be REFERENCES
capable of producing ECM and differentiating into fibroblasts 1. Agnihotri R, Crawford HC, Haro H, Matrisian LM, Havrda MC,
and myofibroblasts (9). In murine pulmonary fibrosis models, Liaw L. Osteopontin, a novel substrate for matrix metalloproteinase-3
circulating fibrocytes contributed to the lung fibroblast popu- (stromelysin-1) and matrix metalloproteinase-7 (matrilysin). J Biol Chem
lation (96) and impaired recruitment of fibrocytes protected 276: 28261–28267, 2001.
2. Alder JK, Chen JJ, Lancaster L, Danoff S, Su SC, Cogan JD, Vulto
against fibrosis (55). Fibrocytes are detected in IPF lungs but I, Xie M, Qi X, Tuder RM, Phillips JA 3rd, Lansdorp PM, Loyd JE,
not healthy control lungs and are recruited by CXCL12 ex- Armanios MY. Short telomeres are a risk factor for idiopathic pulmo-
pression by AECs (3). In one study, circulating fibrocytes were nary fibrosis. Proc Natl Acad Sci USA 105: 13051–13056, 2008.
higher in IPF patients compared with normal controls and were 3. Andersson-Sjoland A, de Alba CG, Nihlberg K, Becerril C, Ramirez
R, Pardo A, Westergren-Thorsson G, Selman M. Fibrocytes are a
elevated in AE-IPF compared with stable IPF (54). In three potential source of lung fibroblasts in idiopathic pulmonary fibrosis. Int
patients who recovered from AE-IPF, circulating fibrocyte per- J Biochem Cell Biol 40: 2129 –2140, 2008.
centage returned to preexacerbation levels in recovery. Circulat- 4. Ando M, Miyazaki E, Ito T, Hiroshige S, Nureki SI, Ueno T,
ing fibrocyte percent did not correlate with pulmonary func- Takenaka R, Fukami T, Kumamoto T. Significance of serum vascular
tion, exercise capacity, or extent of fibrosis on high-resolution endothelial growth factor level in patients with idiopathic pulmonary
fibrosis. Lung 188: 247–252, 2010.
computed tomography; however, ⬎5% circulating fibrocytes 5. Armanios M. Syndromes of telomere shortening. Annu Rev Genomics
was associated with worse survival (54). Hum Genet 10: 45–61, 2009.
6. Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, Markin
C, Lawson WE, Xie M, Vulto I, Phillips JA 3rd, Lansdorp PM,
CONCLUSIONS AND FUTURE DIRECTIONS
Greider CW, Loyd JE. Telomerase mutations in families with idio-
pathic pulmonary fibrosis. N Engl J Med 356: 1317–1326, 2007.
Major advances in the understanding the pathobiology of 7. Azuma A, Nukiwa T, Tsuboi E, Suga M, Abe S, Nakata K, Taguchi
IPF have occurred over the past decade, leading to the recog- Y, Nagai S, Itoh H, Ohi M, Sato A, Kudoh S. Double-blind, placebo-
nition of numerous potential molecular biomarkers, but the controlled trial of pirfenidone in patients with idiopathic pulmonary
field of molecular biomarkers for IPF remains in its infancy. fibrosis. Am J Respir Crit Care Med 171: 1040 –1047, 2005.
Only two prognostic biomarkers (MUC5B and MMP7) appear 8. Barlo NP, van Moorsel CH, Ruven HJ, Zanen P, van den Bosch JM,
Grutters JC. Surfactant protein-D predicts survival in patients with
promising enough at this time to consider for translation into idiopathic pulmonary fibrosis. Sarcoidosis Vasc Diffuse Lung Dis 26:
clinical practice. Currently, there is insufficient evidence to 155–161, 2009.
support any biomarkers for other clinical roles. Mechanisti- 9. Bellini A, Mattoli S. The role of the fibrocyte, a bone marrow-derived
cally informative molecular biomarkers that are practical, ac- mesenchymal progenitor, in reactive and reparative fibroses. Lab Invest
87: 858 –870, 2007.
curate, validated, and clinically useful are needed to move the 10. Biomarkers Definitions Working Group. Biomarkers and surrogate
field forward. Characterization of potential molecular biomark- endpoints: preferred definitions and conceptual framework. Clin Phar-
ers for IPF should continue to take advantage of the wealth of macol Ther 69: 89 –95, 2001.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
MOLECULAR BIOMARKERS IN IPF L689
11. Boon K, Bailey NW, Yang J, Steel MP, Groshong S, Kervitsky D, Kaminski N. Peripheral blood mononuclear cell gene expression profiles
Brown KK, Schwarz MI, Schwartz DA. Molecular phenotypes distin- predict poor outcome in idiopathic pulmonary fibrosis. Sci Transl Med 5:
guish patients with relatively stable from progressive idiopathic pulmo- 205ra136, 2013.
nary fibrosis (IPF). PLoS One 4: e5134, 2009. 30. Hirasawa Y, Kohno N, Yokoyama A, Inoue Y, Abe M, Hiwada K.
12. Borie R, Crestani B, Dieude P, Nunes H, Allanore Y, Kannengiesser KL-6, a human MUC1 mucin, is chemotactic for human fibroblasts. Am
C, Airo P, Matucci-Cerinic M, Wallaert B, Israel-Biet D, Cadranel J, J Respir Cell Mol Biol 17: 501–507, 1997.
Cottin V, Gazal S, Peljto AL, Varga J, Schwartz DA, Valeyre D, 31. Hodgson U, Pulkkinen V, Dixon M, Peyrard-Janvid M, Rehn M,
Grandchamp B. The MUC5B variant is associated with idiopathic Lahermo P, Ollikainen V, Salmenkivi K, Kinnula V, Kere J, Tuki-
pulmonary fibrosis but not with systemic sclerosis interstitial lung disease ainen P, Laitinen T. ELMOD2 is a candidate gene for familial idio-
in the European Caucasian population. PLoS One 8: e70621, 2013. pathic pulmonary fibrosis. Am J Hum Genet 79: 149 –154, 2006.
13. Bossuyt PM, Reitsma JB, Bruns DE, Gatsonis CA, Glasziou PP, 32. Hunninghake GM, Hatabu H, Okajima Y, Gao W, Dupuis J, La-
Irwig LM, Lijmer JG, Moher D, Rennie D, de Vet HC; STARD tourelle JC, Nishino M, Araki T, Zazueta OE, Kurugol S, Ross JC,
Group. Towards complete and accurate reporting of studies of diagnostic San Jose Estepar R, Murphy E, Steele MP, Loyd JE, Schwarz MI,
accuracy: the STARD Initiative. Ann Intern Med 138: 40 –44, 2003. Fingerlin TE, Rosas IO, Washko GR, O’Connor GT, Schwartz DA.
14. Bridges JP, Wert SE, Nogee LM, Weaver TE. Expression of a human MUC5B promoter polymorphism and interstitial lung abnormalities. N
surfactant protein C mutation associated with interstitial lung disease Engl J Med 368: 2192–2200, 2013.
disrupts lung development in transgenic mice. J Biol Chem 278: 52739 – 33. Idiopathic Pulmonary Fibrosis Clinical Research Network, Raghu
52746, 2003. G, Anstrom KJ, King TE, Jr., Lasky JA, Martinez FJ. Prednisone,
15. Bridges JP, Xu Y, Na CL, Wong HR, Weaver TE. Adaptation and azathioprine, and N-acetylcysteine for pulmonary fibrosis. N Engl J Med
increased susceptibility to infection associated with constitutive expres- 366: 1968 –1977, 2012.
sion of misfolded SP-C. J Cell Biol 172: 395–407, 2006. 34. Ishii H, Mukae H, Kadota J, Kaida H, Nagata T, Abe K, Matsukura
16. Cha SI, Ryerson CJ, Lee JS, Kukreja J, Barry SS, Jones KD, Elicker S, Kohno S. High serum concentrations of surfactant protein A in usual
BM, Kim DS, Papa FR, Collard HR, Wolters PJ. Cleaved cytokera- interstitial pneumonia compared with non-specific interstitial pneumonia.
tin-18 is a mechanistically informative biomarker in idiopathic pulmo- Thorax 58: 52–57, 2003.
nary fibrosis. Respir Res 13: 105, 2012. 35. Ishikawa N, Hattori N, Yokoyama A, Kohno N. Utility of KL-6/
17. Chibbar R, Shih F, Baga M, Torlakovic E, Ramlall K, Skomro R, MUC1 in the clinical management of interstitial lung diseases. Respir
Cockcroft DW, Lemire EG. Nonspecific interstitial pneumonia and Investig 50: 3–13, 2012.
usual interstitial pneumonia with mutation in surfactant protein C in 36. Kadota J, Mizunoe S, Mito K, Mukae H, Yoshioka S, Kawakami K,
familial pulmonary fibrosis. Mod Pathol 17: 973–980, 2004. Koguchi Y, Fukushima K, Kon S, Kohno S, Saito A, Uede T, Nasu M.
18. Collard HR, Calfee CS, Wolters PJ, Song JW, Hong SB, Brady S, High plasma concentrations of osteopontin in patients with interstitial
Ishizaka A, Jones KD, King TE, Jr., Matthay MA, Kim DS. Plasma pneumonia. Respir Med 99: 111–117, 2005.
biomarker profiles in acute exacerbation of idiopathic pulmonary fibrosis. 37. Kahloon RA, Xue J, Bhargava A, Csizmadia E, Otterbein L, Kass
Am J Physiol Lung Cell Mol Physiol 299: L3–L7, 2010. DJ, Bon J, Soejima M, Levesque MC, Lindell KO, Gibson KF,
19. Copley SJ, Wells AU, Hawtin KE, Gibson DJ, Hodson JM, Jacques Kaminski N, Banga G, Oddis CV, Pilewski JM, Sciurba FC, Dona-
AE, Hansell DM. Lung morphology in the elderly: comparative CT hoe M, Zhang Y, Duncan SR. Patients with idiopathic pulmonary
study of subjects over 75 years old versus those under 55 years old. fibrosis with antibodies to heat shock protein 70 have poor prognoses. Am
Radiology 251: 566 –573, 2009. J Respir Crit Care Med 187: 768 –775, 2013.
20. Corren J, Lemanske RF, Hanania NA, Korenblat PE, Parsey MV, 38. Kinder BW, Brown KK, McCormack FX, Ix JH, Kervitsky A,
Arron JR, Harris JM, Scheerens H, Wu LC, Su Z, Mosesova S, Schwarz MI, King TE, Jr. Serum surfactant protein-A is a strong
Eisner MD, Bohen SP, Matthews JG. Lebrikizumab treatment in adults predictor of early mortality in idiopathic pulmonary fibrosis. Chest 135:
with asthma. N Engl J Med 365: 1088 –1098, 2011. 1557–1563, 2009.
21. Cronkhite JT, Xing C, Raghu G, Chin KM, Torres F, Rosenblatt RL, 39. King TE, Jr., Pardo A, Selman M. Idiopathic pulmonary fibrosis.
Garcia CK. Telomere shortening in familial and sporadic pulmonary Lancet 378: 1949 –1961, 2011.
fibrosis. Am J Respir Crit Care Med 178: 729 –737, 2008. 40. Kodera M, Hasegawa M, Komura K, Yanaba K, Takehara K, Sato
22. Denhardt DT, Noda M, O’Regan AW, Pavlin D, Berman JS. Osteo- S. Serum pulmonary and activation-regulated chemokine/CCL18 levels
pontin as a means to cope with environmental insults: regulation of in patients with systemic sclerosis: a sensitive indicator of active pulmo-
inflammation, tissue remodeling, and cell survival. J Clin Invest 107: nary fibrosis. Arthritis Rheum 52: 2889 –2896, 2005.
1055–1061, 2001. 41. Konishi K, Gibson KF, Lindell KO, Richards TJ, Zhang Y, Dhir R,
23. Fleming TR, Powers JH. Biomarkers and surrogate endpoints in clinical Bisceglia M, Gilbert S, Yousem SA, Song JW, Kim DS, Kaminski N.
trials. Stat Med 31: 2973–2984, 2012. Gene expression profiles of acute exacerbations of idiopathic pulmonary
24. Friedman SL, Sheppard D, Duffield JS, Violette S. Therapy for fibrosis. Am J Respir Crit Care Med 180: 167–175, 2009.
fibrotic diseases: nearing the starting line. Sci Transl Med 5: 167sr161, 42. Korthagen NM, van Moorsel CH, Barlo NP, Ruven HJ, Kruit A,
2013. Heron M, van den Bosch JM, Grutters JC. Serum and BALF YKL-40
25. Furuhashi K, Suda T, Nakamura Y, Inui N, Hashimoto D, Miwa S, levels are predictors of survival in idiopathic pulmonary fibrosis. Respir
Hayakawa H, Kusagaya H, Nakano Y, Nakamura H, Chida K. Med 105: 106 –113, 2011.
Increased expression of YKL-40, a chitinase-like protein, in serum and 43. Kotsianidis I, Nakou E, Bouchliou I, Tzouvelekis A, Spanoudakis E,
lung of patients with idiopathic pulmonary fibrosis. Respir Med 104: Steiropoulos P, Sotiriou I, Aidinis V, Margaritis D, Tsatalas C,
1204 –1210, 2010. Bouros D. Global impairment of CD4⫹CD25⫹FOXP3⫹ regulatory T
26. Garcia-de-Alba C, Becerril C, Ruiz V, Gonzalez Y, Reyes S, Garcia- cells in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 179:
Alvarez J, Selman M, Pardo A. Expression of matrix metalloproteases 1121–1130, 2009.
by fibrocytes: possible role in migration and homing. Am J Respir Crit 44. Kudo A. Periostin in fibrillogenesis for tissue regeneration: periostin
Care Med 182: 1144 –1152, 2010. actions inside and outside the cell. Cell Mol Life Sci 68: 3201–3207,
27. Gilani SR, Vuga LJ, Lindell KO, Gibson KF, Xue J, Kaminski N, 2011.
Valentine VG, Lindsay EK, George MP, Steele C, Duncan SR. CD28 45. Lawson WE, Cheng DS, Degryse AL, Tanjore H, Polosukhin VV, Xu
down-regulation on circulating CD4 T-cells is associated with poor XC, Newcomb DC, Jones BR, Roldan J, Lane KB, Morrisey EE,
prognoses of patients with idiopathic pulmonary fibrosis. PLoS One 5: Beers MF, Yull FE, Blackwell TS. Endoplasmic reticulum stress
e8959, 2010. enhances fibrotic remodeling in the lungs. Proc Natl Acad Sci USA 108:
28. Greene KE, King TE, Jr., Kuroki Y, Bucher-Bartelson B, Hunning- 10562–10567, 2011.
hake GW, Newman LS, Nagae H, Mason RJ. Serum surfactant 46. Lawson WE, Crossno PF, Polosukhin VV, Roldan J, Cheng DS, Lane
proteins-A and -D as biomarkers in idiopathic pulmonary fibrosis. Eur KB, Blackwell TR, Xu C, Markin C, Ware LB, Miller GG, Loyd JE,
Respir J 19: 439 –446, 2002. Blackwell TS. Endoplasmic reticulum stress in alveolar epithelial cells is
29. Herazo-Maya JD, Noth I, Duncan SR, Kim S, Ma SF, Tseng GC, prominent in IPF: association with altered surfactant protein processing
Feingold E, Juan-Guardela BM, Richards TJ, Lussier Y, Huang Y, and herpesvirus infection. Am J Physiol Lung Cell Mol Physiol 294:
Vij R, Lindell KO, Xue J, Gibson KF, Shapiro SD, Garcia JG, L1119 –L1126, 2008.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
L690 MOLECULAR BIOMARKERS IN IPF

47. Lawson WE, Grant SW, Ambrosini V, Womble KE, Dawson EP, survival of lung fibroblasts. Biochem Biophys Res Commun 338: 1845–
Lane KB, Markin C, Renzoni E, Lympany P, Thomas AQ, Roldan J, 1852, 2005.
Scott TA, Blackwell TS, Phillips JA 3rd, Loyd JE, du Bois RM. 64. Ohtsuki Y, Fujita J, Hachisuka Y, Uomoto M, Okada Y, Yoshinou-
Genetic mutations in surfactant protein C are a rare cause of sporadic chi T, Lee GH, Furihata M, Kohno N. Immunohistochemical and
cases of IPF. Thorax 59: 977–980, 2004. immunoelectron microscopic studies of the localization of KL-6 and
48. Lee CG, Da Silva CA, Dela Cruz CS, Ahangari F, Ma B, Kang MJ, epithelial membrane antigen (EMA) in presumably normal pulmonary
He CH, Takyar S, Elias JA. Role of chitin and chitinase/chitinase-like tissue and in interstitial pneumonia. Med Mol Morphol 40: 198 –202,
proteins in inflammation, tissue remodeling, and injury. Annu Rev 2007.
Physiol 73: 479 –501, 2011. 65. Okamoto M, Hoshino T, Kitasato Y, Sakazaki Y, Kawayama T,
49. Luzina IG, Tsymbalyuk N, Choi J, Hasday JD, Atamas SP. CCL18- Fujimoto K, Ohshima K, Shiraishi H, Uchida M, Ono J, Ohta S,
stimulated upregulation of collagen production in lung fibroblasts re- Kato S, Izuhara K, Aizawa H. Periostin, a matrix protein, is a novel
quires Sp1 signaling and basal Smad3 activity. J Cell Physiol 206: biomarker for idiopathic interstitial pneumonias. Eur Respir J 37: 1119 –
221–228, 2006. 1127, 2011.
50. Maitra M, Wang Y, Gerard RD, Mendelson CR, Garcia CK. Sur- 66. Ono S, Tanaka T, Ishida M, Kinoshita A, Fukuoka J, Takaki M,
factant protein A2 mutations associated with pulmonary fibrosis lead to Sakamoto N, Ishimatsu Y, Kohno S, Hayashi T, Senba M, Yasunami
protein instability and endoplasmic reticulum stress. J Biol Chem 285: M, Kubo Y, Yoshida LM, Kubo H, Ariyoshi K, Yoshiura K, Mo-
22103–22113, 2010. rimoto K. Surfactant protein C G100S mutation causes familial pulmo-
51. Marchal-Somme J, Uzunhan Y, Marchand-Adam S, Valeyre D, nary fibrosis in Japanese kindred. Eur Respir J 38: 861–869, 2011.
Soumelis V, Crestani B, Soler P. Cutting edge: nonproliferating mature 67. Pardo A, Gibson K, Cisneros J, Richards TJ, Yang Y, Becerril C,
immune cells form a novel type of organized lymphoid structure in Yousem S, Herrera I, Ruiz V, Selman M, Kaminski N. Up-regulation
idiopathic pulmonary fibrosis. J Immunol 176: 5735–5739, 2006. and profibrotic role of osteopontin in human idiopathic pulmonary
52. Markart P, Ruppert C, Wygrecka M, Schmidt R, Korfei M, Harbach fibrosis. PLoS Med 2: e251, 2005.
H, Theruvath I, Pison U, Seeger W, Guenther A, Witt H. Surfactant 68. Pardo A, Selman M. Role of matrix metaloproteases in idiopathic
protein C mutations in sporadic forms of idiopathic interstitial pneumo- pulmonary fibrosis. Fibrogenesis Tissue Repair 5, Suppl 1: S9, 2012.
nias. Eur Respir J 29: 134 –137, 2007. 69. Parra ER, Kairalla RA, Ribeiro de Carvalho CR, Eher E, Capelozzi
53. McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark VL. Inflammatory cell phenotyping of the pulmonary interstitium in
GM; Statistics Subcommittee of NCI-EORTC Working Group on idiopathic interstitial pneumonia. Respiration 74: 159 –169, 2007.
Cancer Diagnostics. Reporting recommendations for tumor marker 70. Peljto AL, Zhang Y, Fingerlin TE, Ma SF, Garcia JG, Richards TJ,
prognostic studies. J Clin Oncol 23: 9067–9072, 2005. Silveira LJ, Lindell KO, Steele MP, Loyd JE, Gibson KF, Seibold
54. Moeller A, Gilpin SE, Ask K, Cox G, Cook D, Gauldie J, Margetts MA, Brown KK, Talbert JL, Markin C, Kossen K, Seiwert SD,
PJ, Farkas L, Dobranowski J, Boylan C, O’Byrne PM, Strieter RM, Murphy E, Noth I, Schwarz MI, Kaminski N, Schwartz DA. Asso-
Kolb M. Circulating fibrocytes are an indicator of poor prognosis in ciation between the MUC5B promoter polymorphism and survival in
patients with idiopathic pulmonary fibrosis. JAMA 309: 2232–2239,
idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 179: 588 –594,
2013.
2009.
71. Prasse A, Pechkovsky DV, Toews GB, Jungraithmayr W, Kollert F,
55. Moore BB, Kolodsick JE, Thannickal VJ, Cooke K, Moore TA,
Goldmann T, Vollmer E, Muller-Quernheim J, Zissel G. A vicious
Hogaboam C, Wilke CA, Toews GB. CCR2-mediated recruitment of
circle of alveolar macrophages and fibroblasts perpetuates pulmonary
fibrocytes to the alveolar space after fibrotic injury. Am J Pathol 166:
fibrosis via CCL18. Am J Respir Crit Care Med 173: 781–792, 2006.
675–684, 2005.
72. Prasse A, Pechkovsky DV, Toews GB, Schafer M, Eggeling S,
56. Mukae H, Iiboshi H, Nakazato M, Hiratsuka T, Tokojima M, Abe K,
Ludwig C, Germann M, Kollert F, Zissel G, Muller-Quernheim J.
Ashitani J, Kadota J, Matsukura S, Kohno S. Raised plasma concen-
CCL18 as an indicator of pulmonary fibrotic activity in idiopathic
trations of alpha-defensins in patients with idiopathic pulmonary fibrosis.
interstitial pneumonias and systemic sclerosis. Arthritis Rheum 56: 1685–
Thorax 57: 623–628, 2002. 1693, 2007.
57. Mulugeta S, Nguyen V, Russo SJ, Muniswamy M, Beers MF. A 73. Prasse A, Probst C, Bargagli E, Zissel G, Toews GB, Flaherty KR,
surfactant protein C precursor protein BRICHOS domain mutation Olschewski M, Rottoli P, Muller-Quernheim J. Serum CC-chemokine
causes endoplasmic reticulum stress, proteasome dysfunction, and ligand 18 concentration predicts outcome in idiopathic pulmonary fibro-
caspase 3 activation. Am J Respir Cell Mol Biol 32: 521–530, 2005. sis. Am J Respir Crit Care Med 179: 717–723, 2009.
58. Naik PK, Bozyk PD, Bentley JK, Popova AP, Birch CM, Wilke CA, 74. Pulkkinen V, Bruce S, Rintahaka J, Hodgson U, Laitinen T, Alenius
Fry CD, White ES, Sisson TH, Tayob N, Carnemolla B, Orecchia P, H, Kinnula VL, Myllarniemi M, Matikainen S, Kere J. ELMOD2, a
Flaherty KR, Hershenson MB, Murray S, Martinez FJ, Moore BB. candidate gene for idiopathic pulmonary fibrosis, regulates antiviral
Periostin promotes fibrosis and predicts progression in patients with responses. FASEB J 24: 1167–1177, 2010.
idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 303: 75. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK,
L1046 –L1056, 2012. Colby TV, Cordier JF, Flaherty KR, Lasky JA, Lynch DA, Ryu JH,
59. Nogee LM, Dunbar AE 3rd, Wert SE, Askin F, Hamvas A, Whitsett Swigris JJ, Wells AU, Ancochea J, Bouros D, Carvalho C, Costabel
JA. A mutation in the surfactant protein C gene associated with familial U, Ebina M, Hansell DM, Johkoh T, Kim DS, King TE, Jr., Kondoh
interstitial lung disease. N Engl J Med 344: 573–579, 2001. Y, Myers J, Muller NL, Nicholson AG, Richeldi L, Selman M,
60. Noth I, Zhang Y, Ma SF, Flores C, Barber M, Huang Y, Broderick Dudden RF, Griss BS, Protzko SL, Schunemann HJ; ATS/ERS/JRS/
SM, Wade MS, Hysi P, Scuirba J, Richards TJ, Juan-Guardela BM, ALAT Committee on Idiopathic Pulmonary Fibrosis. An official
Vij R, Han MK, Martinez FJ, Kossen K, Seiwert SD, Christie JD, ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evi-
Nicolae D, Kaminski N, Garcia JG. Genetic variants associated with dence-based guidelines for diagnosis and management. Am J Respir Crit
idiopathic pulmonary fibrosis susceptibility and mortality: a genome- Care Med 183: 788 –824, 2011.
wide association study. Lancet Respir Med 1: 309 –317, 2013. 76. Raghu G, Weycker D, Edelsberg J, Bradford WZ, Oster G. Incidence
61. O’Dwyer DN, Armstrong ME, Trujillo G, Cooke G, Keane MP, and prevalence of idiopathic pulmonary fibrosis. Am J Respir Crit Care
Fallon PG, Simpson AJ, Millar AB, McGrath EE, Whyte MK, Med 174: 810 –816, 2006.
Hirani N, Hogaboam CM, Donnelly SC. The Toll-like receptor 3 77. Ramirez G, Hagood JS, Sanders Y, Ramirez R, Becerril C, Segura L,
L412F polymorphism and disease progression in idiopathic pulmonary Barrera L, Selman M, Pardo A. Absence of Thy-1 results in TGF-beta
fibrosis. Am J Respir Crit Care Med 188: 1442–1450, 2013. induced MMP-9 expression and confers a profibrotic phenotype to
62. Ohnishi H, Yokoyama A, Kondo K, Hamada H, Abe M, Nishimura human lung fibroblasts. Lab Invest 91: 1206 –1218, 2011.
K, Hiwada K, Kohno N. Comparative study of KL-6, surfactant pro- 78. Reilkoff RA, Peng H, Murray LA, Peng X, Russell T, Montgomery
tein-A, surfactant protein-D, and monocyte chemoattractant protein-1 as R, Feghali-Bostwick C, Shaw A, Homer RJ, Gulati M, Mathur A,
serum markers for interstitial lung diseases. Am J Respir Crit Care Med Elias JA, Herzog EL. Semaphorin 7a⫹ regulatory T cells are associated
165: 378 –381, 2002. with progressive idiopathic pulmonary fibrosis and are implicated in
63. Ohshimo S, Yokoyama A, Hattori N, Ishikawa N, Hirasawa Y, transforming growth factor-beta1-induced pulmonary fibrosis. Am J Re-
Kohno N. KL-6, a human MUC1 mucin, promotes proliferation and spir Crit Care Med 187: 180 –188, 2013.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.
Review
MOLECULAR BIOMARKERS IN IPF L691
79. Richards TJ, Kaminski N, Baribaud F, Flavin S, Brodmerkel C, to-mesenchymal transition in response to endoplasmic reticulum stress. J
Horowitz D, Li K, Choi J, Vuga LJ, Lindell KO, Klesen M, Zhang Y, Biol Chem 286: 30972–30980, 2011.
Gibson KF. Peripheral blood proteins predict mortality in idiopathic 96. Tanjore H, Xu XC, Polosukhin VV, Degryse AL, Li B, Han W,
pulmonary fibrosis. Am J Respir Crit Care Med 185: 67–76, 2012. Sherrill TP, Plieth D, Neilson EG, Blackwell TS, Lawson WE.
80. Rode L, Bojesen SE, Weischer M, Vestbo J, Nordestgaard BG. Short Contribution of epithelial-derived fibroblasts to bleomycin-induced lung
telomere length, lung function and chronic obstructive pulmonary disease fibrosis. Am J Respir Crit Care Med 180: 657–665, 2009.
in 46,396 individuals. Thorax 68: 429 –435, 2013. 97. Thomas AQ, Lane K, Phillips J 3rd, Prince M, Markin C, Speer M,
81. Rosas IO, Richards TJ, Konishi K, Zhang Y, Gibson K, Lokshin AE, Schwartz DA, Gaddipati R, Marney A, Johnson J, Roberts R, Haines
Lindell KO, Cisneros J, Macdonald SD, Pardo A, Sciurba F, Dauber J, Stahlman M, Loyd JE. Heterozygosity for a surfactant protein C gene
J, Selman M, Gochuico BR, Kaminski N. MMP1 and MMP7 as mutation associated with usual interstitial pneumonitis and cellular non-
potential peripheral blood biomarkers in idiopathic pulmonary fibrosis. specific interstitial pneumonitis in one kindred. Am J Respir Crit Care
PLoS Med 5: e93, 2008. Med 165: 1322–1328, 2002.
82. Satoh H, Kurishima K, Ishikawa H, Ohtsuka M. Increased levels of 98. Tsakiri KD, Cronkhite JT, Kuan PJ, Xing C, Raghu G, Weissler JC,
KL-6 and subsequent mortality in patients with interstitial lung diseases. Rosenblatt RL, Shay JW, Garcia CK. Adult-onset pulmonary fibrosis
J Intern Med 260: 429 –434, 2006. caused by mutations in telomerase. Proc Natl Acad Sci USA 104:
83. Seibold MA, Wise AL, Speer MC, Steele MP, Brown KK, Loyd JE, 7552–7557, 2007.
Fingerlin TE, Zhang W, Gudmundsson G, Groshong SD, Evans CM, 99. van Moorsel CH, van Oosterhout MF, Barlo NP, de Jong PA, van der
Garantziotis S, Adler KB, Dickey BF, du Bois RM, Yang IV, Herron Vis JJ, Ruven HJ, van Es HW, van den Bosch JM, Grutters JC.
A, Kervitsky D, Talbert JL, Markin C, Park J, Crews AL, Slifer SH, Surfactant protein C mutations are the basis of a significant portion of
Auerbach S, Roy MG, Lin J, Hennessy CE, Schwarz MI, Schwartz adult familial pulmonary fibrosis in a Dutch cohort. Am J Respir Crit
DA. A common MUC5B promoter polymorphism and pulmonary fibro- Care Med 182: 1419 –1425, 2010.
sis. N Engl J Med 364: 1503–1512, 2011. 100. Vuga LJ, Tedrow JR, Pandit KV, Tan J, Kass DJ, Xue J, Chandra
84. Selman M, Carrillo G, Estrada A, Mejia M, Becerril C, Cisneros J, D, Leader JK, Gibson KF, Kaminski N, Sciurba FC, Duncan SR.
Gaxiola M, Perez-Padilla R, Navarro C, Richards T, Dauber J, King C-X-C motif chemokine 13 (CXCL13) is a prognostic biomarker of
TE, Jr., Pardo A, Kaminski N. Accelerated variant of idiopathic idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 189: 966 –974,
pulmonary fibrosis: clinical behavior and gene expression pattern. PLoS 2014.
One 2: e482, 2007. 101. Wang Y, Kuan PJ, Xing C, Cronkhite JT, Torres F, Rosenblatt RL,
85. Selman M, King TE, Pardo A; American Thoracic Society; Euro- DiMaio JM, Kinch LN, Grishin NV, Garcia CK. Genetic defects in
pean Respiratory Society; American College of Chest Physicians. surfactant protein A2 are associated with pulmonary fibrosis and lung
Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about cancer. Am J Hum Genet 84: 52–59, 2009.
its pathogenesis and implications for therapy. Ann Intern Med 134: 102. Weber CE, Li NY, Wai PY, Kuo PC. Epithelial-mesenchymal transi-
136 –151, 2001. tion, TGF-beta, and osteopontin in wound healing and tissue remodeling
86. Selman M, Lin HM, Montano M, Jenkins AL, Estrada A, Lin Z, after injury. J Burn Care Res 33: 311–318, 2012.
103. Willander H, Askarieh G, Landreh M, Westermark P, Nordling K,
Wang G, DiAngelo SL, Guo X, Umstead TM, Lang CM, Pardo A,
Keranen H, Hermansson E, Hamvas A, Nogee LM, Bergman T,
Phelps DS, Floros J. Surfactant protein A and B genetic variants
Saenz A, Casals C, Aqvistg J, Jornvall H, Berglund H, Presto J,
predispose to idiopathic pulmonary fibrosis. Hum Genet 113: 542–550,
Knight SD, Johansson J. High-resolution structure of a BRICHOS
2003.
domain and its implications for anti-amyloid chaperone activity on lung
87. Selman M, Pardo A, Barrera L, Estrada A, Watson SR, Wilson K,
surfactant protein C. Proc Natl Acad Sci USA 109: 2325–2329, 2012.
Aziz N, Kaminski N, Zlotnik A. Gene expression profiles distinguish
104. Yamashita CM, Dolgonos L, Zemans RL, Young SK, Robertson J,
idiopathic pulmonary fibrosis from hypersensitivity pneumonitis. Am J
Briones N, Suzuki T, Campbell MN, Gauldie J, Radisky DC, Riches
Respir Crit Care Med 173: 188 –198, 2006.
DW, Yu G, Kaminski N, McCulloch CA, Downey GP. Matrix metal-
88. Sidhu SS, Yuan S, Innes AL, Kerr S, Woodruff PG, Hou L, Muller loproteinase 3 is a mediator of pulmonary fibrosis. Am J Pathol 179:
SJ, Fahy JV. Roles of epithelial cell-derived periostin in TGF-beta 1733–1745, 2011.
activation, collagen production, and collagen gel elasticity in asthma. 105. Yang IV, Coldren CD, Leach SM, Seibold MA, Murphy E, Lin J,
Proc Natl Acad Sci USA 107: 14170 –14175, 2010. Rosen R, Neidermyer AJ, McKean DF, Groshong SD, Cool C,
89. Song JW, Do KH, Jang SJ, Colby TV, Han S, Kim DS. Blood Cosgrove GP, Lynch DA, Brown KK, Schwarz MI, Fingerlin TE,
biomarkers MMP-7 and SP-A: predictors of outcome in idiopathic Schwartz DA. Expression of cilium-associated genes defines novel
pulmonary fibrosis. Chest 143: 1422–1429, 2013. molecular subtypes of idiopathic pulmonary fibrosis. Thorax 2013.
90. Stuart BD, Lee JS, Kozlitina J, Noth I, Devine MS, Glazer CS, 106. Yang IV, Luna LG, Cotter J, Talbert J, Leach SM, Kidd R, Turner
Torres F, Kaza V, Girod CE, Jones KD, Elicker BM, Ma SF, Vij R, J, Kummer N, Kervitsky D, Brown KK, Boon K, Schwarz MI,
Collard HR, Wolters PJ, Garcia CK. Effect of telomere length on Schwartz DA, Steele MP. The peripheral blood transcriptome identifies
survival in patients with idiopathic pulmonary fibrosis: an observational the presence and extent of disease in idiopathic pulmonary fibrosis. PLoS
cohort study with independent validation. Lancet Respir Med 2: 557–565, One 7: e37708, 2012.
2014. 107. Yokoyama A, Kohno N, Hamada H, Sakatani M, Ueda E, Kondo K,
91. Takahashi F, Takahashi K, Okazaki T, Maeda K, Ienaga H, Maeda Hirasawa Y, Hiwada K. Circulating KL-6 predicts the outcome of
M, Kon S, Uede T, Fukuchi Y. Role of osteopontin in the pathogenesis rapidly progressive idiopathic pulmonary fibrosis. Am J Respir Crit Care
of bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 24: Med 158: 1680 –1684, 1998.
264 –271, 2001. 108. Yokoyama A, Kondo K, Nakajima M, Matsushima T, Takahashi T,
92. Takahashi H, Fujishima T, Koba H, Murakami S, Kurokawa K, Nishimura M, Bando M, Sugiyama Y, Totani Y, Ishizaki T, Ichiyasu
Shibuya Y, Shiratori M, Kuroki Y, Abe S. Serum surfactant proteins H, Suga M, Hamada H, Kohno N. Prognostic value of circulating KL-6
A and D as prognostic factors in idiopathic pulmonary fibrosis and their in idiopathic pulmonary fibrosis. Respirology 11: 164 –168, 2006.
relationship to disease extent. Am J Respir Crit Care Med 162: 1109 – 109. Zhang Y, Noth I, Garcia JG, Kaminski N. A variant in the promoter
1114, 2000. of MUC5B and idiopathic pulmonary fibrosis. N Engl J Med 364:
93. Taniguchi H, Ebina M, Kondoh Y, Ogura T, Azuma A, Suga M, 1576 –1577, 2011.
Taguchi Y, Takahashi H, Nakata K, Sato A, Takeuchi M, Raghu G, 110. Zhu L, Wang L, Luo X, Zhang Y, Ding Q, Jiang X, Wang X, Pan Y,
Kudoh S, Nukiwa T, Pirfenidone Clinical Study Group in Japan. Chen Y. Tollip, an intracellular trafficking protein, is a novel modulator
Pirfenidone in idiopathic pulmonary fibrosis. Eur Respir J 35: 821–829, of the transforming growth factor-beta signaling pathway. J Biol Chem
2010. 287: 39653–39663, 2012.
94. Tanjore H, Blackwell TS, Lawson WE. Emerging evidence for endo- 111. Zuo F, Kaminski N, Eugui E, Allard J, Yakhini Z, Ben-Dor A, Lollini
plasmic reticulum stress in the pathogenesis of idiopathic pulmonary L, Morris D, Kim Y, DeLustro B, Sheppard D, Pardo A, Selman M,
fibrosis. Am J Physiol Lung Cell Mol Physiol 302: L721–L729, 2012. Heller RA. Gene expression analysis reveals matrilysin as a key regu-
95. Tanjore H, Cheng DS, Degryse AL, Zoz DF, Abdolrasulnia R, lator of pulmonary fibrosis in mice and humans. Proc Natl Acad Sci USA
Lawson WE, Blackwell TS. Alveolar epithelial cells undergo epithelial- 99: 6292–6297, 2002.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00014.2014 • www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (187.192.249.172) on April 19, 2021.

You might also like