ArticlePDF Available

Update on biomarkers in systemic sclerosis: tools for diagnosis and treatment

Authors:

Abstract

Systemic sclerosis (SSc) is a complex autoimmune disease in which immune activation, vasculopathy, and extensive fibrosis of the skin and internal organs are among the principal features. SSc is a heterogeneous disease with varying manifestations and clinical outcomes. Currently, patients' clinical evaluation often relies on subjective measures, non-quantitative methods, or requires invasive procedures as markers able to predict disease trajectory or response to therapy are lacking. Therefore, current research is focusing on the discovery of useful biomarkers reflecting ongoing inflammatory or fibrotic activity in the skin and internal organs, as well as being predictive of future disease course. Recently, remarkable progress has been made towards a better understanding of numerous mechanisms involved in the pathogenesis of SSc. This has opened new possibilities for the development of novel biomarkers and therapy. However, current proposed biomarkers that could reliably describe various aspects of SSc still require further investigation. This review will summarize studies describing the commonly used and validated biomarkers, the newly emerging and promising SSc biomarkers identified to date, and consideration of future directions in this field.
REVIEW
Update on biomarkers in systemic sclerosis: tools
for diagnosis and treatment
Alsya J. Affandi
1,2
& Timothy R. D. J. Radstake
1,2
& Wioleta Marut
1,2
Received: 24 April 2015 /Accepted: 16 June 2015 /Published online: 14 July 2015
#
The Author(s) 2015. This article is published with open access at Springerlink.com
Abstract Systemic sclerosis (SSc) is a complex autoimmune
disease in which immune activation, vasculopathy, and exten-
sive fibrosis of the skin and internal organs are among the
principal features. SSc is a heterogeneous disease with varying
manifestations and clinical outcomes. Currently, patients clin-
ical evaluation often relies on subjective meas ures, non-
quantitative methods, or requires invasive procedures as
markers able to predict disease trajectory or response to thera-
py are lacking. Therefore, current research is focusing on the
discovery of useful biomarkers reflecting ongoing inflamma-
tory or fibrotic activity in the skin and internal organs, as well
as being predictive of future disease course. Recently, remark-
able progress has been made towards a better understanding of
numerous mechanisms involved in the pathogenesis of SSc.
This has opened new possibilities for the development of novel
biomarkers and therapy. However, current proposed bio-
markers that could reliably describe various aspects of SSc still
require further investigation. This review will summarize stud-
ies describing the commonly used and validated biomarkers,
the newly emerging and promising SSc biomarkers identified
to date, and consideration of future directions in this field.
Keywords Autoantibodies
.
Biomarker
.
miRNAs
.
Pulmonary fibrosis
.
Skin fibrosis
.
Systemic sclerosis
Systemic sclerosis (SSc) is an autoimmune disease characterized
by fibrosis of the skin and internal organs, preceded by vascular
and immune dysfunction [1]. Depending on the extent of cuta-
neous fibrosis, SSc is classified into two major subtypes: limited
cutaneous (lcSSc) and diffuse cutaneous SSc (dcSSc). In lcSSc,
skin thickening is restricted to the areas distal to the elbows and/
or knees, such as hands and fingers. In dcSSc, the presence of
skin lesions is more extensive and internal organs involvement
is relatively more severe. This classification is supported by the
association with specific autoantibodies that specifically define
the two types of clinical phenotypes. Both SSc phenotypes can
be complicated by severe internal organ dysfunction. Pulmonary
fibrosis and pulmonary arterial hypertensio n (PAH) are the two
most feared complications, representing the major causes of
mortality in SSc patients [2]. Owning to its complex nature
and heterogeneity, SSc remains one of the greatest challenges
to both investigators and physicians. Despite intense investiga-
tion, so far , only a few biomarkers for SSc have been fully
validated and widely accepted. Herein, we present a review of
the literature on promising prognostic biomarkers, biomarkers
of disease activity , skin fibrosis, and lung involvement, with the
aim to provide a comprehensive update on usability of bio-
markers for research and clinical guidance.
Diagnostic and prognostic biomarkers
SSc-specific autoantibodies as predictive markers
The presence of autoantibodies is a central defining aspect of
autoimmune disease s. Autoantibodies are seen at the first
This article is a contribution to the Special Issue on Immunopathology of
systemic sclerosis Guest Editors: Jacob M. van Laar and John Varga
* Timothy R. D. J. Radstake
T.R.D.J.Radstake@umcutrecht.nl
1
Department of Rheumatology and Clinical Immunology,
University Medical Center Utrecht, Heidelberglaan 100,
3584 CX Utrecht, The Netherlands
2
Laboratory of Translational Immunology, University Medical Center
Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
Semin Immunopathol (2015) 37:475487
DOI 10.1007/s00281-015-0506-4
diagnosis in more than 95 % of SSc patients and have been
associated with distinct disease subtypes and with differences
in disease severity. Antitopoisomerase I (ATAs) and
anticentromere antibodies (ACAs) are the most widely used
diagnostic biomarkers for SSc [35].
Anti-Scl-70 antibodies originally identified by Douvas
et al. [6] are directed against DNA topoisomerase I [7]and
therefore should be more accurately termed antitopoisomerase
I antibodies (ATAs). These autoantibodies are seen predomi-
nantly in dcSSc patients; however, their presence is not entire-
ly restricted to this clinical subset since a subgroup of lcSSc
patients was also found to be ATA-positive [8, 9]. ATA has
been associated with poorer prognosis, increased mortality,
pulmonary fibrosis, and cardiac involvement [912]. Another
recent study of clinical outcomes in patients with digital ulcers
showed that patients positive for ATAs developed Raynauds
phenomenon earlier and had double rate of lung fibrosis as
compared with ACA-positive patients [13]. Some reports in-
dicate that changes in ATA titers over time can be useful in
monitoring disease activity and progression and therefore use-
ful for prognostic purposes [14].
ACAs recognize centromeric protein from CENP-A to
CENP-F, of which CENP-B is reported t o be a major
autoantigen reacting with virtual ly all anti-CENP-positive
SSc sera [15, 16]. ACAs are found in 20 to 30 % of SSc
patients, and in up to 90 % of lcSSc patients [4, 17]. In patients
with Raynauds phenomenon, ACAs have been reported to
predict the onset of lcSSc [3, 18]. While severe interstitial
fibrosis and renal crisis occur rarely, pulmonary arterial hyper-
tension occurs in about 20 % of anti-CENP patients [9, 10,
19]. Anti-CENPs are often associated with other antibodies,
such as anti-Sjogrens-syndrome-related antigen A (anti-Ro)
[20] or antimitochondrial antibodies [21]. Moreover, it has
been reported that ACA positivity correlated with a more fa-
vorable prognosis and lower mortality compared with the pos-
itivity of other SSc-related autoantibodies [22].
Antibodies against RNA polymerase I and III (anti-RNP I
and III) are detected with high specificity in SSc patients (98
100 %) [23, 24]. Their prevalence varies from 10 to 25 % in
different SSc cohorts. Anti-RNP I and III are associated with
dcSSc involvement and renal crisis [25]. More recently, it has
been shown that the presence of anti-RNP is associated with
rapid onset of the disease and skin thickening. Therefore, they
are still among the best predictive markers available for rapid
skin progression [26]. Autoantibodies to RNP II are uncom-
mon and are not specific for SSc since they can be also be
found in the sera of systemic lupus erythematosus (SLE) and
overlap syndrome [27
].
Ad
ditional SSc-specific autoantibodies with diagnostic or
prognostic utility include the anti-Th/To and anti-U3 RNP
(antifibrillarin) antibodies. Th/To autoantibodies are directed
against subunit of RNase P and RNase MRP [28]. They are
found in 25 % of SSc patients and are clinically associated
with lcSSc (8.4 % of lcSSc patients, 0.6 % of dcSSc) [10, 29].
Among lcSSc patients, anti-Th/To are a marker of the worst
survival rate perhaps related to severe pulmonary embolism
(PE) preceding PAH and renal crisis [19]. It has been reported
that the presence of anti-Th/To may assist in identifying sine
SSc subset in patients with pulmonary fibrosis [30]. Anti-U3
RNP antibodies target fibrillarin, a small protein belonging to
the U3 small nuclear ribonucleoprotein (RNP) complex. Al-
though they are considered to be a specific marker for SSc,
they are found in less than 7 % of SSc sera and their confir-
mation using advanced techniques continues to be a challenge
[31]. They are most frequent in males and African Americans
with SSc and are associated with muscle involvement and
increased risk of PAH [32].
Recently, autoantibodies against angiotensin II type 1 re-
ceptor (AT
1
R) and endothelin-1 type A receptor (ET
A
R) have
been shown to be elevated in the sera of most SSc patients,
and associated with vascular and fibrotic complications [33,
34]. They are more frequent in SSc-PAH/connective tissue
disease-PAH compared to other forms of pulmonary hyper-
tension. Therefore, they could serve as new predictive and
prognostic biomarkers of PAH in SSc. These autoantibodies
not only predict development of PAH but also are associated
with higher mortality in SSc patients [33].
More recently , antiestrogen receptor antibodies (anti-ERα)
were detected in sera of 42 % SSc patients; whereas, no anti-
ERα antibodies were found in healthy controls. Anti-ERα anti-
bodies were significantly associated with disease activity and
were mainly found among patients with the diffuse form of the
disease, the ANAs positivity, and the late capillaroscopy pattern.
However , it is important to note that anti-ERα antibodies are not
specific for SSc since they were also detected in patients with
SLE but not in other patients with other autoimmune diseases
such as rheumatoid arthritis (RA) or Behcets disease [35].
Other autoantibodies of relevance to SSc but less common-
ly present in SSc patients include the anti-U11/U12 RNP an-
tibodies. They are highly associated with severe lung fibrosis,
and anti-PM-Scl antibodies found in patients with SSc overlap
and with myositis [36]. Remarkably, many of the autoanti-
bodies in SSc show correlation with other type of biomarkers
such as those for skin fibrosis and pulmonary complications,
as we will discuss below (Table 1).
Circulating miRNA
miRNAs are a class of endogenous and evolutionary con-
served short, noncoding RNAs that bind to the 3 untranslated
region of target genes. Once bound, miRNAs repress target
gene translation or promote mRNA destabilization and degra-
dation. They are expressed in a tissue-specific and cell type-
specific manner but can also circulate in the bloodstream, and
such circulating miRNAs are remarkably stable [37]. This has
raised the possibility that miRNAs may be probed in the
476 Semin Immunopathol (2015) 37:475487
circulation and can serve as novel diagnostic markers. It has
been shown that the elevated expression of pro-fibrotic
miRNAs and reduced expression of antifibrotic miRNAs are
important factors in the developments of fibrosis in SSc. Fur-
thermore, several studies have already demonstrated that the
levels of selected miRNAs were altered in the serum of SSc
patients [38, 39].
The levels of miR-150 were found to be downregulated in
the serum of SSc patients versus healthy controls and were
correlated with more severe clinical manifestations. For in-
stance, h igher incid ence of antit opoisomerase I antibodies
and a higher prevalence of pitting scars were seen in patients
with lower miR-150 levels than in those without. Although
the difference did not reach statistical significance, patients
with lower miR-150 levels had a higher ratio of dcSSc to
lcSSc and a higher modified Rodnan skin score (mRSS) when
compared with patients with normal levels of miR-150 [40].
Similar correlation was observed for another miRNA
miR196a, where patients with lower serum miR-196a levels
had significantly higher ratio of dcSSc to lcSSc and higher
mRSS; in addition, they showed higher prevalence of pitting
scars than those without [41]. Other studies evidenced that the
serum level of miR-30b [42] and let-7a [43]werehighlyde-
creased in SSc patients versus healthy controls. Both miRNAs
were again downregulated more strongly in diffuse subset of
SSc than in limited SSc. Interestingly, miR-30b and let-7a
were inversely correlated with mRSS [42, 43].
On the contrary, serum levels of miR-92a [44] and miR-142-
3p [45] were markedly higher in SSc when compared to healthy
controls or SLE, dermatomyositis, and scleroderma spectrum
disorder (SSD) patients. Therefore, these miRNAs may provide
as useful diagno stic markers for the differentiation of SSc from
other scleroderma spectrum disorders (Table 2).
Biomarkers for disease activity
One of the main challenges of SSc studies is to develop a
sufficient tool for a global measurement for disease activity
that represents an ongoing disease activity and/or response to
treatment. Unlike other autoimmune diseases such as SLE or
RA, for many SSc patients, ongoing inflammation is difficult
to assess and vascular and tissue fibroses are not easy to quan-
titate especially in the early stage of the disease. Currently, the
Valentini disease activity index, developed by The European
Scleroderma Study Group (EScSG), is the most widely used
activity score in SSc studies [46]. This activity index includes
mRSS, DLCO, and E SR, w ithout a specific biochemical
marker. The Medsger disease severity scale is also frequently
used as a measure of disease activity [47]. However, this as-
sessment may more reflect damage or severity rather than the
ongoing disease activity.
The enhanced liver fibrosis (ELF) test was developed as a
clinical grade serum test for chronic liver diseases, including
procollagen-III aminoterminal-propeptide (PIIINP), tissue in-
hibitor of matrix metalloproteinase-1 (TIMP-1), and
hyaluronic acid (HA) in its algorithm. Each of these three
serum markers is increased in SSc patients as compared to
healthy controls and associated with more severe complica-
tions or increased mortality [4850]. Recently, ELF test was
tested in SSc patients and showed significant correlations with
both disease activity and severity [51]. ELF score also corre-
lated with mRSS, Health Assessment Questionnaire-Disability
Index (HAQ-DI), and inversely correlated with DLCO, but it
did not correlate with vasculopathy features such as PAH [51].
Other candidate biochemical markers for disease activity
and severity in SSc have been derived from their association
with an organ-specific involvement. The markers for pulmo-
nary involvement in SSc, serum vWF and KL6, were signif-
icantly associated with disease severity and activity, respec-
tively [52, 53]. The serum level of cartilage oligomeric matrix
protein (COMP), a molecule that has been associated with
skin and lun g fibrosis as we describe below, also showed
correlation with disease severity [54]. The angiopoietin/Tie2
axis has gained some interests due to their roles in angiogen-
esis. Serum levels of angiopoietin-2 (Ang-2), but not
angiopoietin-1, have been shown to correlate with disease
activity [55]. Another study found a similar but not significant
Table 1 Diagnostic and prognostic biomarkersautoantibodies
Biomarker Source Association Reference
Antitopoisomerase I (ATAs) Serum dcSSc, poor prognosis, increased mortality, lung fibrosis,
cardiac involvement
[814]
Anticentromere (ACAs) Serum lcSSc, PAH, more favorable prognosis, lower mortality [4, 9, 10, 1517, 19]
Anti-RNA polymerase I and III (anti-RNAP I, III) Serum dcSSc, skin progression, renal crisis [2326]
Anti-Th/To Serum lcSSc, a marker of worst survival rate, muscle involvement, PAH [10, 19, 29, 30]
Anti-U3 RNP Serum Activity [31, 32]
Anti-AT
1
R, anti- ET
A
R Serum Activity, PAH, vascular and fibrotic complications, higher mortality [33]
Anti-ERα Serum Activity, dcSSc [35]
Anti-U11/U12 RNP Serum Severe lung fibrosis [36]
Semin Immunopathol (2015) 37:475487 477
trend in plasma, although the authors found a stronger corre-
lation using the ratio of Ang-2 and its soluble receptor Tie2
with disease activity [56].
The classic inflammatory cytokine IL-6 is increased in the
sera of SSc patients and has been associated with multiple
organ involvement including skin [57], the occurrence of pul-
monary fibrosis [58], FVC decline, and increased mortality
[59]. Plasma IL-6 level was found to be higher in ATA-
positive and anti-RNAP III-positive patients but not in
ACA-positive SSc patients [60]. In one study, serum IL-6
was shown to correlate with disease activity [61], although
this was not found by others [62]. In a genetic association
study, IL-6 polymorphism in SSc patients was shown to be
associated with disease activity and HAQ-DI, unfortunately
circulating IL-6 was not measured [63].
Growth differentiation factor 15 (GDF-15) is a distant
member of the TGFβ superfamily and found to be elevated
in the serum of SSc patients compared to healthy controls [64,
65]. In SSc patients, serum GDF-15 levels showed strong
correlation with mRSS, disease activity, and disease severity
[65], in particular those with pulmonary involvement, as we
will discuss below.
It is important to note that many of these studies assessing
disease activity are cross-sectional and limited to small co-
horts at single centers. Future multicenter validation and lon-
gitudinal study are necessary to assess their sensitivity for
changes over time in a larger population. Multibiomarker ap-
proach such as the ELF score should also be considered
(Table 3).
Biomarkers correlating with skin fibrosis
Skin fibrosis, the hallmark of SSc, is defined as an excess
deposition and accumulation of extra cellular matrix in the
dermis. Despite our growing understanding of this process
and many available targets, our therapeutic success in amelio-
rating skin fibrosis in SSc is still minimal. Even today, the
gold standard for measuring SSc skin fibr osis is mRSS, a
relatively simple determination of skin thickness, which has
significant inter-observer variability and is rather subjective.
Moreover, the mRSS may not be sensitive enough to find
smaller but important and early changes in skin thickening
[66]. Therefore, there is a need for other specific and more
precise markers for assessing skin fibrosis.
Peripheral blood biomarkers
There is a large number of potential circulating biomarkers for
skin fibrosis which include COMP, MMP-9, MMP-12, LOX,
IL-6, IL-10, and CXCL4 (Table 4). Here, we will discuss those
biomarkers that are most robustly shown to be of potential
relevance.
Cartilage oligomeric protein 1 (COMP) is a non-
collagenous glycoprotein, mostly synthesized by
chondrocytes, oste oblasts, tenocytes, synovial fibroblas ts,
and dermal fibroblasts. This protein, highly regulated by
TGF-β, is not detectable in the healthy skin but is highly
overexpressed in skin biopsies and fibroblasts of SSc patients
[67, 68]. Moreover, COMP was found to be increased in SSc
sera and correlated with the extent of skin involvement, as
assessed by mRSS and ultrasound [69]. More recent study
confirmed high levels of COMP in the serum of SSc patients,
and its level was higher in dcSSc subset than in lcSSc [54].
Matrix metalloproteinases (MMPs), responsible for the
degradation of collagens and other extra cellular matrix
(ECM) proteins, are also involved in the release and activation
of many cytokines and growth factors [70]. Several inhibitors
are known to control their activity. Both, MMPs and their
inhibitors, were extensively studied in the pathogenesis of
SSc. MMP-9 and MMP-12 w ere found to be a potential
markers for skin fibrosis.
Table 2 Prognostic biomarkerscirculating miRNA
Biomarker Expression in SSc Source Association Reference
miR-150 Downregulated Serum dcSSc, skin fibrosis [40]
miR196a Downregulated Serum dcSSc, skin fibrosis, higher prevalence of pitting scars [41]
miR-30b Downregulated Serum dcSSc, inversely correlated with skin fibrosis [42]
let-7a Downregulated Serum dcSSc, inversely correlated with skin fibrosis [43]
miR-92a Upregulated Serum SSc [44]
miR-142-3p Upregulated Serum SSc [45]
Table 3 Biomarker in SSc disease activity
Biomarker Source Association Reference
ELF test Serum Activity, severity [51]
vWF Serum Severity [52]
KL-6 Serum Activity [53]
Ang-2 Serum Activity [55]
COMP Serum Activity [54]
IL-6 Serum Activity [61, 62]
GDF-15 Serum Activity, severity [65]
478 Semin Immunopathol (2015) 37:475487
MMP-9, whose substrates include type IV collagen in
basement membrane, has been associated with chronic in-
flammatory autoimmune diseases, including rheumatoid ar-
thritis [71]andSLE[72]. Moreover, its overexpression has
been reported in various pathologic conditions characterized
by excessive fibrosis, including idiopathic pulmonary fibrosis
[73] and chronic pancreatitis [74]. In SSc, fibroblasts isolated
from SSc patients expressed more MMP-9 than healthy con-
trols. Furthermore, serum level of MMP-9 was elevated in
SSc, with higher concentration in dcSSc compared to lcSSc,
and correlated well with mRSS [75].
MMP-12, also known as macrophage metalloelastase
(MME), has a broad substrate specificity for matrix mac-
romolecules, recognizing elastin, type IV collagen, fibro-
nectin, or v itronectin. MMP-12 has been implicated in
different pathological conditions including atherosclerosis,
cancers, and skin diseases [76, 77]. In SSc patients, der-
mal fibro blasts expresse d an d re leased MMP-12 [78].
More recent studies reported that serum levels of MMP-
12 were significantly increased in SSc patients, also cor-
relating well with skin fibrosis, with dcSSc having higher
levels of MMP-12 [79].
Lysyl oxidase (LOX) is an extracellular copper enzyme
that cross-links collagen and elastin, thus stabilizing collagen
fibrils. Consistent with its expression in the skin and fibro-
blasts in the context of SSc, the levels of LOX were elevated
in the serum of SSc patients versus healthy controls. Further
analysis revealed a correlation of LOX concentration with the
mRSS in patients without lung fibrosis, indicating its specific
correlation with skin fibrosis. Moreover, LOX levels were
higher in SSc patients with dcSSc than in those with lcSSc,
which may reflect a more advance fibrosis in diffuse subset of
SSc [80].
In SSc patients, there is a strong relationship between in-
flammation and fibrosis supported by the upregulation of
both, pro-inflammatory and pro-fibrotic markers in the serum
as well as in skin. The role of different cytokines and
chemokines has been analyzed in skin fibrosis of SSc in sev-
eral studies. For instance, IL-6 and IL-10 serum levels were
found to be elevated in SSc patients and significantly corre-
lated with skin fibrosis assessed by mRSS [57]. However,
recently, Codullo et al. confirmed that SSc patients expressed
high level of IL-6 but did not find clear associations with
mRSS or other clinical parameters [62].
CXCL4, largely viewed as a pro-inflammatory chemo-
kine, in addition to its chemoattractant activity, regulates
an array of immune cells, including T cells, monocytes,
dendritic cells, as well as non-immune cells like endothe-
lial cells. Recently, van Bon et al. used a proteomic ap-
proach and identified CXCL4 as a potential biomarker
associated with multiple organ involvement in SSc. Circu-
lating CXCL4 levels strongly correlated with the extent of
skin fibrosis more with dcSSc subsets than lcSSc. In a
prospective cohort study, elevated CXCL4 in the serum
of SSc predicted a faster progression of skin fibrosis [81].
Gene expression profiling
Gene expression profiling from skin biopsies is another
interesting approach to identify biomarkers for skin fi-
brosis. Skin biopsies, although more difficult to obtain,
allow for a more direct insight into the ongoing fibrotic
reaction. Moreover, they can lead to the discovery of
genes specific f or different subsets of SSc and to predict
if patients will develop more severe subset of the dis-
ease. For in stance, in 2008, Milano et al. reported a
177-gene signature that was associated with severity of
skin disease in diffuse subsets of SSc [82]. This identi-
fication not only allows for a better understanding of
the disease pathogenesis but also provides important in-
formation for novel therapeutic targets.
TGF-β is one of the most potent pro-fibrotic cytokines
in SSc and also one of the strongest stimulators for the
differentiation of fibroblasts into activated myofibroblasts.
Therefore, a group of researchers examined the expression
of genes highly upregulated by TGF-β and found that
some of these genes were highly correlated with the skin
score. When they expande d their studies to interferon-
regulated genes, they found that several of these genes
also strongly correlated with the mRSS. Therefore, the
combination of both TGF-β and IFN-regulated genes,
namely COMP a nd thrombospondin-1 (TSP-1) (TGF-β
Table 4 Emerging biomarker in
SSc skin fibrosis
Biomarker Source Association Reference
COMP Skin, serum dcSSc, skin fibrosis [54, 6769]
MMP-9, MMP-12 Skin, serum dcSSc, skin fibrosis [75, 78, 79]
LOX Skin, serum dcSSc, skin fibrosis [80]
IL-6, IL-10 Serum Skin fibrosis [57, 62]
CXCL4 Serum dcSSc, skin fibrosis [81]
TSP-1, IFI44, Siglec-1 Skin Skin fibrosis [83]
LH2 Skin Skin fibrosis activity [84]
Semin Immunopathol (2015) 37:475487 479
regulated genes), and IFN-inducible 44 (IFI44) and
sialoadhesin (Siglec-1) resulted in a particularly strong cor-
relation with skin score [83].
Lysyl hydroxylase-2 (LH2), an enzyme involved in colla-
gen biosynthesis, was found to be elevated in the skin biopsies
and isolated fibroblasts of SSc patients and could represent a
marker for the skin fibrotic activity in these patients. LH-2
overexpression was found to be accompanied by an associated
increase in the Pyr cross-links present in the accumulated col-
lagen in the SSc patients. These Pyr cross-links are critical for
the mechanical stability and tensile strength of collagen [84]
(Table 4).
Biomarkers involved in SSc lung involvement
Pulmonary complications are common in SSc patients. They
are most often manifested by the fibrotic interstitial lung dis-
ease (ILD), or pulmonary vascular disease leading to pulmo-
nary arterial hypertension (PAH), or co-occurrence of both.
Together, SSc-associated ILD and PAH are the major cause
of disease-related mortality in SSc [85, 86].
Interstitial lung disease
The majority of SSc patients have evidence of pulmonary
fibrosis, based on autopsy and radiographic findings [87]. In
the European League Against Rheumatism Scleroderma Tri-
als and Research (EUSTAR) registry, pulmonary fibrosis ap-
peared more common in diffuse than in limited SSc (53.4 vs
34.7 %) [9]. To detect ILD in SSc, a chest imaging using high-
resolution computed tomography (HRCT) and pulmonary
function tests (PFT), including the measurement of forced
vital capacity (FVC) and diffusing capacity of lung for carbon
monoxide (DLCO), are being used. A new quantitative HRCT
has improved visual radiographic assessment of ILD but not
yet widely available [88]. Moreover, repeated exposure to
radiation can be detrimental. Although FVC and DLCO show
correlation with HRCTand adequately measure lung function,
they are not specific for ILD or the ongoing fibrotic process.
Therefore, additional biomarkers that are more accessible, re-
peatable, and complement both HRCT and PFT are needed
[89].
Several different studies have examined the use of the lung-
epith elial-derived protein surfactant protein-D (SP-D) and
glycoprotein Krebs von den Lungen-6 (KL-6). As reviewed
by others previously, most studies showed increased serum
SP-D and KL-6 and their correlation with decline in FVC
and DLCO in SSc patients, however with varying degrees of
correlation [90, 91]. A recent study showed that serum KL-6
correlated strongly with HRCT-fibrosis score, serum ATA ti-
ters, and correlated inversely with FVC and DLCO [53]. They
found a moderate correlation of SP-D with HRCT-fibrosis
score, but no significant association with other clinical param-
eters tested. Other studies also found correlation of serum KL-
6, as well as COMP, with lung fibrosis [92]. Additionally, SSc
patients with elevated SP-D and KL-6 had far more frequent
ATA positivity and less frequent ACA compared to those with
normal level [93]. Serum KL-6 also showed strong correlation
with mRSS and disease activity index, indicating it to be a
multipurpose biomarker candidate in SSc [53].
CCL18 is a chemokine produced by antigen presenting
cells, particularly by alveolar macrophages in different inter-
stitial lung diseases [94]. In SSc, the level of CCL18 was
elevated in the bronchoalveolar lavage (BAL) fluid, lung, se-
rum, and associated with lung involvement [95, 96]. One
study showed moderate but significant negative correlation
of serum CCL18 with DLCO and FVC in SSc [97]. In their
retrospective cohort analysis, serum CCL-18 level was de-
creased in SSc patients having an improvement of pulmonary
fibrosis (as measured by HRCT, PFT, and BAL analysis) and
comparable to the decrease of KL-6 and SP-D [97]. Another
study found a similar observation where serum CCL18 in SSc
correlated with DLCO decline and total lung capacity (TLC)
decline, and changes of TLC over a period of at least 6 months
[95]. A longitudinal study of a 4-year period showed that a
cut-off serum CCL18 value at 187 ng/ml is able to predict
worsening ILD [98]. This was later reproduced in an indepen-
dent cohort with a similar cut-off value and hazard ratios [99],
but another study challenged this finding suggesting that cor-
relation between CCL18 and changes in FVC could only be
seen at a short term (1 year) but not at a longer period [100].
Interestingly, a recent microarray analysis of SSc-ILD lung
showed that lung CCL18 RNA expression correlated with
changes of HRCT-score FibMax and negatively correlated,
although not strongly, with % predicted FVC [96].
van Bon et al. showed that chemokine CXCL4 was asso-
ciated with lung disease manifestations in SSc [
81].
Patients
who had high circulating CXCL4 (>10 ng/ml) developed lung
fibrosis earlier compared to those who had low CXCL4, as
measured by >30 % decrease of FVC and HRCT. In the pro-
spective cohort, patients with a high CXCL4 baseline showed
a significantly faster decline in DLCO and a higher prevalence
of HRCT-confirmed lung fibrosis. Earlier study has showed a
significant increase of CXCL4 in BAL fluid from SSc patients
exclusively those with ILD [101].
CXCL8 (IL-8) functions as the main chemotactic factor for
neutrophils and other granulocytes. CXCL8 gene polymor-
phisms were associated with an increased susceptibility to
SSc [102, 103]. Circulating CXCL8 level has been reported
to be elevated in SSc patients [62, 104], but this was found not
in all studies [60]. Two studies showed an increase of CXCL8
level in the BAL fluid from SSc patients that correlated with a
more extensive lung fibrosis based on HRCT [92] and in-
versely correlated with DLCO, FVC, and TLC [105], but nei-
ther study measured circulating CXCL8. Other investigations
480 Semin Immunopathol (2015) 37:475487
using CXCL8 level in the serum of SSc patients showed
no significant association with PFT or any future pulmo-
nary involvement [62, 106]; whereas, one report showed
serum CXCL8 association w ith DLCO decrease [104].
These findings suggest that CXCL8 level may more
strongly reflect disease progression locally rather than
systemically.
S100A8 (MRP-8, calgranulin A) and S100A9 (MRP-14,
calgranulin B) are members of the S100 calcium-binding pro-
teins. Together they form a complex, S100A8/9 (calprotectin,
calgranulinA/B), that is able to modulate inflammatory pro-
cesses mainly by binding to toll-like receptor 4 (TLR4) [107].
Earlier studies have shown increased of S100A8/9 or their
homodimeric form ats in the sera [108, 109], f eces [110,
111], saliva [112], BAL fluids [92], and skin [109, 113]of
SSc patients compared to healthy individuals. High level of
S100A8/9 in the BAL fluid [92] and serum [109]inpatients
has been associated with an extensive lung fibrosis and ATA
positivity, although no direct correlation to PFT was found. A
recent proteomic analysis of serum samples from SSc patients
also showed S100A8/9 to be increased particularly in lcSSc
having lung fibrosis and ATA-positive patients [114]. As re-
cent evidences point to the role of TLR4 signaling in fibrosis
[115117], S100A8/9 is a potentially interesting molecule to
investigate further.
The serum level of GDF-15 has been shown to be strongly
associated with multiple organ involvement in SSc especially
in the lung. Serum GDF-15 level was higher in patients with
ILD compared to those without, and its level in the serum
negatively correlated with DLCO and FVC [64, 65]. Patients
with high level of serum GDF-15 had a more frequent ATA
and a less frequent ACA positivity [64]. Importantly, SSc
patients with higher baseline GDF-15 leve l showed lower
DLCO and worsened l ung diseases severity score over a
follow-up period of up to 30 months, suggesting its value as
a predictive prognostic marker of lung function and fibrosis in
SSc [65](Table5).
Pulmonary arterial hypertension
Although PAH appears to be more frequent in the lcSSc than
dcSSc (9.2 vs 5.9 % according to EULAR registry [9]), it can
occur in all forms of SSc. Patients with SSc-PAH has a poor
prognosis; therefore, an early detection of SSc-PAH and initi-
ation of therapy are essential in disease management. Trans-
thoracic echocardiography (TTE) of pulmonary artery systolic
pressure is the most widely used screening tool for PAH; how-
ever, the method has considerable measurement variability
and may not be sufficiently sensitive for the detection of early
disease [120]. The more invasive right heart catheterization
(RHC) is still the golden standard to confirm PAH in SSc
patients.
N-terminal pro-brain natriuretic peptide (NT-proBNP), a
biomarker of myocardial stress, has been intensively studied
as a diagnostic marker for SSc- PAH . Serum NT-pro-BNP
levels have been showed to strongly correlate with mean pul-
monary arterial pressure (PAP) and pulmonary vascular resis-
tance (PVR) [121]. They also showed association of NT-
proBNP with severity of PAH and risk of mortality [121].
Later studies also showed similar correlation of NT-proBNP
with increased PAP based on TTE and RHC, as well as neg-
ative correlation with DLCO and the presence of ATA
[122
12
6]. More recently, NT-proBNP has been in used as a
diagnostic marker in combination with other modalities in-
cluding TTE and PFT. This approac h, as r eported b y the
DETECT study and others, gave an improved sensitivity and
reduced missed diagnosis for early SSc-PAH as compared to
the ESC/ERS guidelines [125, 127, 128]. However, it is im-
portant to note that this marker is not specific to PAH, as it
may results from pulmonary venous hypertension or other
cardiac dysfunction.
In search for surrogate marker for SSc-PAH, many inves-
tigators have looked into the molecules produced by or acting
on the endothelium and have attempted to correlate them to
hemodynamic and pulmonary function parameters. Markers
of vascular injury such as vascular endothelial growth factor
(VEGF), endothelin-1 (ET-1), and von Willebrand factor
(vWF), as well as the soluble adhesion molecules ICAM-1
andVCAM-1havebeenshowedtobeelevatedinSScsera
and associated with PAH [129, 130]. Both circulating VEGF
and ET-1 levels were found higher in SSc patients with PAH
compared to those without and their level correlated positively
with pulmonary arterial pressure [129, 131]. However, ET-1
associat ion with pulmona ry pressure or function was not
Table 5 Biomarker in SSc-ILD
Biomarker Source Association Reference
SP-D and KL-6 Serum HRCT, FVC, DLCO [53, 92, 93, 118, 119]
CCL18 Serum, BAL, lung HRCT, FVC, TLC, DLCO [95, 96, 98100]
CXCL4 Plasma, BAL HRCT, FVC, DLCO [81, 101]
CXCL8 Serum, BAL HRCT, FVC, TLC, DLCO [62, 92, 104106]
S100A8/9 Serum, plasma, BAL HRCT [92, 109, 114]
COMP Serum HRCT [92]
GDF-15 Serum DLCO, FVC [64, 65]
Semin Immunopathol (2015) 37:475487 481
found in another study [132]. Serum VEGF levels also corre-
lated with decline of DLCO in cohorts of SSc patients without
ILD [131] and in limited subtype [56]. The level of vWF in
plasma of SSc patients was found to correlate with PAP, based
on Doppler cardiography [133]. In a substudy from QUINs
randomized placebo-controlled trial, baseline serum vWF an-
tigen concentrations significantly related to disease activity,
inversely correlated to %FVC and %DLCO at baseline, and
were able to predict elevated PAP of >40 mmHg after 3 years,
based on TTE [52]. However, another study did not find cor-
relation between serum vWF and PFT measurements, perhaps
due to a smaller cohort and a difference in statistical analysis
[134]. In an 18-month prospective cohort, plasma vWF did
not correlate with future changes in DLCO or skin score [81].
Furthermore, the use of vWF as a biomarker can be challeng-
ing since there is already a large variation in healthy individ-
uals: ABO blood group, genetic polymorphism, and age are
among the determinants [135, 136].
In addition to its association with skin and lung fibrosis,
circulating CXCL4 levels were elevated in SSc patients who
had evidence of PAH compared to those without, as deter-
mined on RHC [81]. In this study, high CXCL4 levels were
also associated with an earlier development of PAH. In a tran-
scriptome analysis of lung biopsies from idiopathic PAH
patients, CXCL4 was one of the most highly and differentially
expressed genes as compared to healthy controls [137]. As
CXCL4 exerts angiostatic properties on pulmonary arterial endo-
thelial cells [138], this suggests that CXCL4 might be involved in
the pathophysiology of PAH in SSc.
As mentioned above, serum GDF-15 was associated with
pulmonary fibrosis and impaired lung function. In SSc pa-
tients with PAH, the plasma level of GDF-15 was significantly
higher as compared to those without [139]. Plasma concentra-
tions of GDF-15 showed strong correlation with right ventricular
systolic pressure (RVSP) on echocardiography, NT-proBNP
plasma levels, and negative correlation with DLCO, but no
Table 6 Biomarker in SSc-PAH
Biomarker Source Association Reference
NT-proBNP Serum PAP, PVR, DLCO [121126]
VEGF Serum PAP, DLCO [56, 131]
ET-1 Plasma PAP [130, 132]
vWF Serum, plasma PAP, FVC, DLCO [52, 81, 133, 134]
Anti-AT1R and anti-ETAR Serum PAH development [33, 34]
CXCL4 Plasma PAH development [81]
GDF-15 Plasma RVSP, DLCO [64, 65, 139]
Macrophage
Monocyte
IFI44
Siglec1
IL-10
IL-6
CXCL4
S100A8/9
Type-I IFN
IL-6
CXCL8
pDC
Endothelial
cell
ET-1
vWF
ICAM-1
Ang-2
VEGF
Epithelial
cell
KL-6
SP-D
CXCL8
B cell
Plasma
cell
Muscle cell
mDC
Fibroblast
ATA
ACA
Anti-Th/To
Anti-U3RNP
Anti-AT
1
R
Anti ET
A
R
Others Abs
NT-proBNP
VEGF
GDF-15
Undefined
sources
miR-30b
miR-92
miR-142-3p
miR-150
miR-196a
COMP
TSP-1
TIMP-1
Inflammatory
mediators
Vascular injury
Pro-fibrotic
factors
Myofibroblast
Fibrosis
ECM
Vasculopathy
Tissue remodelling
Other
cells
let-7a
HA
Loss of organ function
MMPs
LH2
LOX
VEGF
GDF-15
CCL-18
Fig. 1 A schematic depiction of biomarkers in systemic sclerosis and
their production by different cell types. Immune cells produce a large
number of biomarkers that have been investigated in SSc, and their
interaction with endothelial cells, fibroblasts, and other cell types may
eventually lead to extracellular matrix (ECM) deposition and the
progression of disease
482 Semin Immunopathol (2015) 37:475487
correlation with any RHC-based hemodynamics [139]. Im-
portantly, a ROC curve analysis showed that a plasma GDF-15
cut-off level at 125 pg/ml was able to identify SSc-PAH better
than NT-proBNP at 473 pmol/L (93 % sensitivity and 88 %
specificity vs 86 % sensitivity and 30 % specificity) and was able
to predict mortality in SSc patients [139]. Later studies measuring
serum GDF-15 in SSc patients had very few patients with PAH
in their cohorts that make it difficult to interpret [64, 65]. In a
cohort of patients with idiopathic PAH, elevated serum GDF-15
level was associated with right atrial pressure, wedge pressure,
and serum NT-proBNP level [140]. They also showed potential
prognostic value of serum GDF-15 as it was related to changes of
serum NT-proBN P and venous oxygen saturation in their
follow-up cohort [140](Table6).
Novel approaches to identify biomarkers in SSc
The fast advancement of current molecular biology and bio-
chemical techniques has moved research from the reductionist
approach of studying one individual component at a time,
towards a more holistic approach where multiple high-
throughput omics layersso called systems medicinecan
be determined in clinically well-defined patient groups.
Genomic-wide association studies, whole transcriptome, and
proteome analysis have been performed in recent studies and
yielded novel candidate biomarkers for SSc [141143]. The
use of more recent state-of-the-art technologies such as mass
cytometry, that would enable us to phenotype immune com-
partments or other cells of interest in a great detail, are cur-
rently underway. The challenge of systemic multilayered
large-data gathering approach is the complexity of big-data
management, analysis, and interpretation. Computational
models and computer learning algorithms are essential to an-
swer specific research questions that hopefully lead investiga-
tors to the discovery of new biomarkers and understanding
pathways.
Conclusions
Discoveries of new biomarkers and composite scores in SSc
have supported the more conventional approaches in patients
evaluation including mRSS, PFTs, RHC, and HRCT. For ex-
ample, incorporating NT-proBNP to TTE and PFT measures
has improved diagnosis of SSc-PAH significantly. Several
biomarkers with clinically important multipurpose utility can
give an added value, for example, both KL-6 and CXCL4
showed correlation with skin and lung involvement and pre-
dictive of future disease course. Many of these new promising
biomarkers (see Fig. 1), however, still require validation and
assessment in longitudinal cohorts or in clinical trials. More
investigations in prognostic markers that can predict patients
disease trajectory or differences in response to therapy are of
urgent need. In the near future, systems medicine approaches
including the true integration of multilayered data may pro-
vide more complete assessment of patients, novel biomarkers,
understanding of disease, or even drug discovery and person-
alized therapy.
Acknowledgments This study was supported by the Dutch Arthritis
Association (T.R.D.J. Radstake), the Netherlands Organization for Scien-
tific Research (VIDI grant to T.R.D.J. Radstake and Mosaic grant to A.J.
Affandi), ERC starting grant (to T.R.D.J. Radstake), and Marie Curie
Intra-European Fellowship (to W. Marut).
Conflict of interest The authors declare that they have no competing
interests.
Open Access This article is distributed under the terms of the Creative
Commons Attribution 4.0 International License (http://
creativecommons.org/licenses/by/4.0/), which permits unrestricted use, dis-
tribution, and reproduction in any medium, provided you give appropriate
credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made.
References
1. Gabrielli A, Avvedimento EV, Krieg T (2009) Scleroderma. N
Engl J Med 360:19892003. doi:10.1056/NEJMra0806188
2. Masi AT (1988) Classification of systemic sclerosis (scleroderma):
relationship of cutaneous subgroups in early disease to outcome
and serologic reactivity. J Rheumatol 15:894898
3. Steen VD (2005) Autoantibodies in systemic sclerosis. Semin
Arthritis Rheum 35:3542. doi:10.1016/j.semarthrit.2005.03.005
4. Walker JG, Fritzler MJ (2007) Update on autoantibodies in sys-
temic sclerosis. Curr Opin Rheumatol 19:580591. doi:10.1097/
BOR.0b013e3282e7d8f9
5. Graf SW, Hakendorf P, Lester S et al (2012) South Australian
Scleroderma Register: autoantibodies as predictive biomarkers of
phenotype and outcome. Int J Rheum Dis 15:102109. doi:10.
1111/j.1756-185X.2011.01688.x
6. Douvas AS, Achten M, Tan EM (1979) Identification of a nuclear
protein (Scl-70) as a unique target of human antinuclear antibodies
in scleroderma. J Biol Chem 254:1051410522
7. Shero JH, Bordwell B, Rothfield NF, Earnshaw WC (1986) High
titers of autoantibodies to topoisomerase I (Scl-70) in sera from
scleroderma patients. Science 231:737740. doi:10.1126/science.
3003910
8. Basu D, Reveille JD (2005) Anti-scl-70. Autoimmunity 38:6572.
doi:10.1080/08916930400022947
9. Walker UA, Tyndall A, Czirják L et al (2007) Clinical risk assess-
ment of organ manifestations in systemic sclerosis: a report from
the EULAR Scleroderma Trials and Research group database.
Ann Rheum Dis 66:754763. doi:10.1136/ard.2006.062901
10. Reveille JD, Solomon DH (2003) Evidence-based guidelines for
the use of immunologic tests: anticentromere, Scl-70, and nucle-
olar antibodies. Arthritis Rheum 49:399412. doi:10.1002/art .
11113
11. Steen VD, Powell DL, Medsger TA (1988) Clinical correlations
and prognosis based on serum autoantibodies in patients with sys-
temic sclerosis. Arthritis Rheum 31:196203. doi:10.1016/0923-
1811(93)91335-R
Semin Immunopathol (2015) 37:475487 483
12. Hesselstrand R, Scheja A, Shen GQ et al (2003) The association of
antinuclear antibodies with organ involvement and survival in
systemic sclerosis. Rheumatology 42:534540. doi:10.1093/
rheumatology/keg170
13. Denton CP, Krieg T, Guillevin L et al (2012) Demographic, clin-
ical and antibody characteristics of patients with digital ulcers in
systemic sclerosis: data from the DUO Registry. Ann Rheum Dis
71:718721. doi:10.1136/annrheumdis-2011-200631
14. Hanke K, Dähnrich C, Brückner CS et al (2009) Diagnostic value
of anti-topoisomerase I antibodies in a large monocentric cohort.
Arthritis Res Ther 11:R28. doi:10.1186/ar2622
15. Earnshaw W, Bordwell B, Marino C, Rothfield N (1986) Three
human chromosomal autoantigens are recognized by sera from
patients with anti-centromere antibodies. J Clin Invest 77:426
430. doi:10.1172/JCI112320
16. Rothfield N, Whitaker D, Bordwell B et al (1987) Detection of
anticentromere antibodies using cloned autoantigen CENP-B.
Arthritis Rheum 30:14161419
17. Carwile LeRoy E, Black C, Fleischmajer R et al (1988)
Scleroderma (systemic sclerosis): classification, subsets and path-
ogenesis. J Rheumatol 15:202205
18. Koenig M, Dieudé M, Senécal JL (2008) Predictive value of an-
tinuclear autoantibodies: the lessons of the systemic sclerosis au-
toantibodies. Autoimmun Rev 7:588593. doi:10.1016/j.autrev.
2008.06.010
19. Mitri GM, Lucas M, Fertig N et al (2003) A comparison between
anti-TH/To- and anticentromere antibody-positive systemic scle-
rosis patients with limited cutaneous involvement. Arthritis
Rheum 48:203209. doi:10.1002/art.10760
20. Miyawaki S, Asanuma H, Nishiya ma S, Yoshinaga Y (2005)
Clinical and serological heterogeneity in patients with
anticentromere antibodies. J Rheumatol 32:14881494
21. Akiyama Y, Tanaka M, Takeishi M et al (2000) Clinical, serolog-
ical and genetic study in patients with CREST syndrome. Intern
Med 39:451456. doi:10.2169/internalmedicine.39.451
22. Ferri C, Valentini G, Cozzi F et al (2002) Systemic sclerosis:
demographic, clinical, and serologic features and survival in 1,
012 Italian patients. Medicine (Baltimore) 81:139153. doi:10.
1097/00005792-200203000-00004
23. Chang M, Wang RJ, Yangco DT et al (1998) Analysis of autoan-
tibodies against RNA polymerases using immunoaffinity-purifed
RNA polymerase I, II, and III antigen in an enzyme-linked immu-
nosorbent assay. Clin Immunol Immunopathol 89:7178. doi:10.
1006/clin.1998.4591
24. Kuwana M, Okano Y, Pandey JP et al (2005) Enzyme-linked
immunosorbent assay for detection of anti-RNA polymerase III
antibody: analytical accuracy and clinical associations in systemic
sclerosis. Arthritis Rheum 52:24252432. doi:10.1002/art.21232
25. Kuwana M, Kaburaki J, Mimori T et al (1993) Autoantibody
reactive with three classes of RNA polymerases in sera from pa-
tients with systemic sclerosis. J Clin Invest 91:13991404. doi:10.
1172/JCI116343
26. Cavazzana I, Angela C, Paolo A et al (2009) Anti-RNA polymer-
ase III antibodies: a marker of systemic sclerosis with rapid onset
and skin thickening progression. Autoimmun Rev 8:580584. doi:
10.1016/j.autrev.2009.02.002
27. Satoh M, Ajmani AK, Ogasawara T et al (1994) Autoantibodies to
RNA polymerase II are common in systemic lupus erythematosus
and overlap syndrome: specific recognition of the phosphorylated
(IIO) form by a subset of human sera. J Clin Invest 94:19811989.
doi:10.1172/JCI117550
28. Van Eenennaam H, Vogelzangs JHP, Lugtenberg D et al (2002)
Identity of the RN ase MRP- and RNase P-associated Th/To
autoantigen. Arthritis Rheum 46:32663272. doi:10.1002/art.
10673
29. Okano Y, Medsger TA (1990) Autoantibody to Th ribonucleopro-
tein (nucleolar 7
2
RNA protein particle) in patients with systemic
sclerosis. Arthritis Rheum 33:18221828. doi:10.1002/art.
1780331210
30. Fischer A, Pfalzgraf FJ, Feghali-Bostwick CA et al (2006) Anti-th/
to-positivity in a cohort of patients with idiopathic pulmonary
fibrosis. J Rheumatol 33:16001605
31. Kipnis RJ, Craft J, Hardin JA (1990) The analysis of antinuclear
and antinucleolar autoantibodies of scleroderma by
radioimmunoprecipitation assays. Arthritis Rheum 33:14311437
32. Aggarwal R, Lucas M, Fertig N et al (2009) Anti-U3 RNP auto-
antibodies in systemic sclerosis. Arthritis Rheum 60:11121118.
doi:10.1002/art.24409
33. Becker MO, Kill A, Kutsche M et al (2014) Vascular receptor
autoantibodies in pulmonary arterial hypertension associated with
systemic sclerosis. Am J Respir Crit Care Med 190:808817. doi:
10.1164/rccm.201403-0442OC
34. Riemekasten G, Philippe A, Näther M et al (2011) Involvement of
functional autoantibodies against vascular receptors in systemic
sclerosis. Ann Rheum Dis 70:530536. doi:10.1136/ard.2010.
135772
35. Giovannetti A, Maselli A, Colasanti T et al (2013) Autoantibodies
to estrogen receptor α in systemic sclerosis (SSc) as pathogenetic
determinants and markers of progression. PLoS One. doi:10.1371/
journal.pone.0074332
36. Fertig N, Domsic RT, Rodriguez-Reyna T et al (2009) Anti-U11/
U12 RNP antibodies in systemic sclerosis: a new serologic marker
associated with pulmonary fibrosis. Arthritis Care Res 61:958
965. doi:10.1002/art.24586
37. Friedman RC, Farh KKH, Burge CB, Bartel DP (2009) Most
mammalian mRNAs are conserved targets of microRNAs.
Genome Res 19:92105. doi:10.1101/gr.082701.108
38. Li H, Yang R, Fan X et al (2012) MicroRNA array analysis of
microRNAs related to systemic scleroderma. Rheumatol Int 32:
307313. doi:10.1007/s00296-010-1615-y
39. Zhu H, Li Y, Qu S et al (2012) MicroRNA expression abnormal-
ities in limited cutaneous scleroderma and diffuse cutaneous
scleroderma. J Clin Immunol 32:514522. doi:10.1007/s10875-
011-9647-y
40. Honda N, Jinnin M, Kira-Etoh T et al (2013) MiR-150 down-
regulation contributes to the constitutive type i collagen overex-
pression in scleroderma dermal fibroblasts via the induction of
integrin β3. Am J Pathol 182:206216. doi:10.1016/j.ajpath.
2012.09.023
41. Honda N, Jinnin M, Kajihara I et al (2012) TGF-β-mediated
downregulation of microRNA-196a contributes to the constitutive
upregulated type I collagen expression in scleroderma dermal fi-
broblasts. J Immunol 188:3 3233331. doi:10.4049/jimmunol.
1100876
42. Tanaka S, Suto A, Ikeda K et al (2013) Alteration of circulating
miRNAs in SSc: miR-30b regulates the expression of PDGF re-
ceptor β. Rheumatology (Oxford) 52:19631972. doi:10.1093/
rheumatology/ket254
43. Makino K, Jinnin M, Hirano A et al (2013) The downregulation of
microRNA let-7a contributes to the excessive expression of type I
collagen in systemic and localized scleroderma. J Immunol 190:
39053915. doi:10.4049/jimmunol.1200822
44. Sing T, Jinnin M, Yamane K et al (2012) MicroRNA-92a expres-
sion in the sera and dermal fibroblasts increases in patients with
scleroderma. Rheumatology (Oxford) 51:1550155
6. doi:10.
1093/rheumatology/kes120
45. Makino K, Jinnin M, Kajihara I et al (2012) Circulating miR-142-
3p levels in patients with systemic sclerosis. Clin Exp Dermatol
37:3439. doi:10.1111/j.1365-2230.2011.04158.x
46. Valentini G, Bencivelli W, Bombardieri S et al (2003) European
Scleroderma Study Group to define disease activity criteria for
484 Semin Immunopathol (2015) 37:475487
systemic sclerosis. III. Assessment of the construct validity of the
preliminary activity criteria. Ann Rheum Dis 62:901903. doi:10.
1136/ard.62.9.901
47. Valentini G, Silman AJ, Veale D (2003) Assessment of disease
activity. Clin Exp Rheumatol 21:S39S41
48. Young-Min SA, Beeton C, Laughton R et al (2001) Serum TIMP-
1, TIMP-2, and MMP-1 in patients with systemic sclerosis, pri-
mary Raynauds phenomenon, and in normal controls. Ann
Rheum Dis 60:846851
49. Scheja A, Akesson A, Hørslev-Petersen K (1992) Serum levels of
aminoterminal type III procollagen peptide and hyaluronan predict
mortality in systemic sclerosis. Scand J Rheumatol 21:59. doi:10.
3109/03009749209095054
50. Nagy Z, Czirják L (2005) Increased levels of amino terminal
propeptide of type III procollagen are an unfavourable predictor
of survival in systemic sclerosis. Clin Exp Rheumatol 23:165172
51. Abignano G, Cuomo G, Buch MH et al (2014) The enhanced liver
fibrosis test: a clinical grade, validated serum test, biomarker of
overall fibrosis in systemic sclerosis. Ann Rheum Dis 73:420
427. doi:10.1136/annrheumdis-2012-202843
52. Barnes T, Gliddon A, Do CJ et al (2012) Baseline vWF factor
predicts the development of elevated pulmonary artery pressure in
systemic sclerosis. Rheumatology (Oxford) 51:16061609. doi:
10.1093/rheumatology/kes068
53. Bonella F, Volpe A, Caramaschi P et al (2011) Surfactant protein
D and KL-6 serum levels in systemic sclerosis: correlation with
lung and systemic involvement. Sarcoidosis Vasc Diffuse Lung
Dis 28:2733
54. Gheita TA, Hussein H (2012) Cartilage oligomeric matrix protein
(COMP) in systemic sclerosis (SSc): role in disease severity and
subclinical rheumatoid arthritis overlap. Joint Bone Spine 79:51
56. doi:10.1016/j.jbspin.2011.02.022
55. Michalska-Jakubus M, Kowal-Bielecka O, Chodorowska G et al
(2011) Angiopoietins-1 and -2 are differentially expressed in the
sera of patients with systemic sclerosis: high angiopoietin-2 levels
are associated with greater severity and higher activity of the dis-
ease. Rheumatology 5 0:7467 55. doi:10.1093/rheumatology/
keq392
56. Dunne JV, Keen KJ, Van Eeden SF (2013 ) Circulating
angiopoietin and Tie-2 levels in systemic sclerosis. Rheumatol
Int 33:475484. doi:10.1007/s00296-012-2378-4
57. Sato S, Hasegawa M, Takehara K (2001) Serum levels of
interleukin-6 and interleukin-10 correlate with total skin thickness
score in patients with systemic sclerosis. J Dermatol Sci 27:140
146. doi:10.1016/S0923-1811(01)00128-1
58. Scala E, Pallotta S, Frezzolini A et al (2004) Cytokine and che-
mokine levels in systemic sclerosis: relationship with cutaneous
and internal organ involvement. Clin Exp Immunol 138:540546.
doi:10.111 1/j.1365-2249.2004.02642.x
59. De Lauretis A, Sestini P, Pantelidis P et al (2013) Serum interleu-
kin 6 is predictive of early functional decline and mortality in
interstitial l ung disease as sociated with sy stemic sclerosis. J
Rheumatol 40:435446. doi:10.3899/jrheum.120725
60. Gourh P, Arnett FC, Assassi S et al (2009) Plasma cytokine pro-
files in systemic sclerosis: associations with autoantibody subsets
and clinical manifestations. Arthritis Res Ther 11:R147. doi:10.
1186/ar2821
61. Jurisic Z, Martinovic-Kaliterna D, Marasovic-Krstulovic D et al
(2013) Relationship between interleukin-6 and cardiac involve-
ment in systemic sclerosis. Rheumatology (Oxford) 52:1298
1302. doi:10.1093/rheumatology/ket131
62. Codullo V, Baldwin HM, Singh MD et al (2011) An investigation
of the inflammatory cytokine and chemokine network in systemic
sclerosis. Ann Rheum Dis 70:11151121. doi:10.1136/ard.2010.
137349
63. Sfrent-Cornateanu R, Mihai C, Balan S et al (2006) The IL-6
promoter polymorphism is associated with disease activity and
disability in systemic sclerosis. J Cell Mol Med 10:95595
9
64. Yanaba K, Asano Y, Tada Y et al (2012) Clinical significance of
serum growth differentiation factor-15 levels in systemic sclerosis:
association with disease severity. Mod Rheumatol 22:668675.
doi:10.1007/s10165-01 1-0568-7
65. Lambrecht S, Smith V, De Wilde K et al (2014) Growth differen-
tiation factor 15, a marker of lung involvement in systemic scle-
rosis, is involved in fibrosis development but is not indispensable
for fibrosis development. Arthritis Rheum 66:418427. doi:10.
1002/art.38241
66. Czirják L, Foeldvari I, Müller-Ladner U (2008) Skin involvement
in systemic sclerosis. Rheumatology (Oxford) 47(Suppl 5):v44
v45. doi:10.1093/rheumatology/ken309
67. Farina G, Lemaire R, Korn JH, Widom RL (2006) Cartilage olig-
omeric matrix protein is overexpressed by scleroderma dermal
fibroblasts. Matrix Biol 25:213222. doi:10.1016/j.matbio.2006.
01.007
68. Farina G, Lemaire R, Pancari P et al (2009) Cartilage oligomeric
matrix protein expression in systemic sclerosis reveals heteroge-
neity of dermal fibroblast responses to transforming growth factor
beta. Ann Rheum Dis 68:435441. doi:10.1136/ard.2007.086850
69. Hesselstrand R, Kassner A, Heinegård D, Saxne T (2008) COMP:
a candidate molecule in the pathogenesis of systemic sclerosis
with a potential as a disease marker. Ann Rheum Dis 67:1242
1248. doi:10.1136/ard.2007.082099
70. Wynn TA (2007) Common and unique mechanisms regulate fi-
brosis in various fibroproliferative diseases. J Clin Invest 117:
524529. doi:10.1172/JCI31487
71. Ahrens D, Koch AE, Pope RM et al (1996) Expression of matrix
metalloproteinase 9 (96-kd gelatinase B) in human rheumatoid
arthritis. Arthritis Rheum 39:15761587
72. Liu Y, Zheng M, Yin W, Zhang B (2004) Relationship of serum
levels of HGF and MMP-9 with disease activity of patients with
systemic lupus erythematosus. Zhejiang Da Xue Xue Bao Yi Xue
Ban 33(340343):348
73. Pardo A, Selman M (2012) Role of matrix metalloproteases in
idiopathic pulmonary fibrosis. Fibrogenesis Tissue Repair
5(Suppl 1):S9. doi:10.1186/1755-1536-5-S1-S9
74. Venkateshwari A, Sri Manjari K, Krishnaveni D et al (2011) Role
of plasma MMP 9 levels in the pathogenesis of chronic pancrea-
titis. Indian J Clin Biochem 26:136139. doi:10.1007/s12291-
010-0103-1
75. Kim W-U, Min S-Y, Cho M-L et al (2005) Elevated matrix
metalloproteinase-9 in patients with systemic sclerosis. Arthritis
Res Ther 7:R71R79. doi:10.1186/ar1454
76. Said AH, Raufman J-P, Xie G (2014) The role of matrix metallo-
proteinases in colorectal cancer. Cancers (Basel) 6:366375. doi:
10.3390/cancers6010366
77. Pardo A, Selman M (2006) Matrix metalloproteases in aberrant
fibrotic tissue remodeling. Proc Am Thorac Soc 3:383388. doi:
10.1513/pats.200601-012TK
78. Serratì S, Cinelli M, Margheri F et al (2006) Systemic sclerosis
fibroblast inhibit in vitro angiogenesis by MMP-12-dependent
cleavage of the endothelial cell urokinase receptor. J Pathol 210:
240248. doi:10.1002/path.2048
79. Manetti M, Guiducci S, Romano E et al (2012) Increased serum
levels and tissue expression of matrix metalloproteinase-12 in pa-
tients with systemic sclerosis: correlation with severity of skin and
pulmonary fibrosis and vascular damage. Ann Rheum Dis 71:
10641072. doi:10.1136/annrheumdis-2011-200837
80. Rimar D, Rosner I, Nov Yet al (2014) Brief report: lysyl oxidase is
a potential biomarker of fibrosis in systemic sclerosis. Arthritis
Rheumatol 66:726730. doi:10.1002/art.38277
Semin Immunopathol (2015) 37:475487 485
81. Van Bon L, Affandi AJ, Broen J et al (2014) Proteome-wide anal-
ysis and CXCL4 as a biomarker in systemic sclerosis. N Engl J
Med 370:433443. doi:10.1056/NEJMoa1114576
82. Milano A, Pendergrass SA, Sargent JL et al (2008) Molecular
subsets in the gene expression signatures of scleroderma skin.
PLoS One 3, e2696. doi:10.1371/journal.pone.0002696
83. Farina G, Lafyatis D, Lemaire R et al (2010) A four-gene biomark-
er predicts skin disease in patients with diffuse cutaneous systemic
sclerosis. Arthritis Rheum 62:580588. doi:10.1002/art.27220
84. Brinckmann J, Kim S, Wu J et al (2005) Interleukin 4 and
prolonged hypoxia induce a higher gene expression of lysyl hy-
droxylase 2 and an altered cross-link pattern: important pathoge-
netic steps in early and late stage of systemic scleroderma? Matrix
Biol 24:459468. doi:10.1016/j.matbio.2005.07.002
85. Steen VD, Medsger TA (2007) Changes in causes of death in
systemic sclerosis, 19722002. Ann Rheum Dis 66:940944.
doi:10.1136/ard.2006.066068
86. Wells AU, Steen V, Valentini G (2009) Pulmonary complications:
one of the most challenging complications of systemic sclerosis.
Rheumatology (Oxford) 48(Suppl 3):iii 40iii44. doi:10.1093/
rheumatology/kep109
87. Solomon JJ, Olson AL, Fischer A et al (2013) Scleroderma lung
disease. Eur Respir Rev 22:619. doi:10.1183/09059180.
00005512
88. Tashkin DP, Volkmann ER, Tseng C-HC-H, et al. (2014)
Relationship between quantitative radiographic assessments of
interstitial lung disease and physiological and clinical features of
systemic sclerosis. Ann Rheum Dis 0:18. doi:10.1136/
annrheumdis-2014-206076
89. Abignano G, Buch M, Emery P, Del Galdo F (2011) Biomarkers
in the management of scleroderma: an update. Curr Rheumatol
Rep 13:412. doi:10.1007/s11926-010-0140-z
90. Lafyatis R (2012) Application of biomarkers to clinical trials in
systemic sclerosis. Curr Rheumatol Rep 14:4755. doi:10.1007/
s11926-011-0216-4
91. Lota HK, Renzoni EA (2012) Circulating biomarkers of interstitial
lung disease in systemic sclerosis. Int J Rheum. doi:10.1155/2012/
121439
92. Hesselstrand R, Wildt M, Bozovic G et al (2013) Biomarkers from
bronchoalveolar lavage fluid in systemic sclerosis patients with
interstitial lung disease relate to severity of lung fibrosis. Respir
Med 107:10791086. doi:10.1016/j.rmed.2013.03.015
93. Yanaba K, Hasegawa M, Takehara K, Sato S (2004) Comparative
study of serum surfactant protein-D and KL-6 concentrations in
patients with systemic sclerosis as markers for monitoring the
activity of pulmonary fibrosis. J Rheumatol 31:11121120
94. Cai M, Bonella F, He X et al (2013) CCL18 in serum, BAL fluid
and alveolar macrophage culture supernatant in interstitial lung
diseases. Respir Med 107:14441452. doi:10.1016/j.rmed.2013.
06.004
95. Prasse A, Pechkovsky DV, Toews GB et al (2007) CCL18 as an
indicator of pulmonary fibrotic activity in idiopathic interstitial
pneumonias and systemic sclerosis. Arthritis Rheum 56:1685
1693. doi:10.1002/art.22559
96. Christmann RB, Sampaio-Barros P, Stifano G et al (2014)
Association of inte rferon- and transforming growth factor β-
regulated genes and macro phage activation with systemic
sclerosis-related progressive lung fibrosis. Arthritis Rheumatol
66:714725. doi:10.1002/art.38288
97. Kodera M, Hasegawa M, Komura K et al (2005) Serum pulmo-
nary and activation-regulated chemokine/CCL18 levels in patients
with systemic sclerosis: a sensitive indicator of active pulmonary
fibrosis. Arthritis Rheum 52:28892896. doi:10.1002/art.21257
98. Tiev KP, Hua-Huy T, Kettaneh A et al (2011) Serum CC chemo-
kine ligand-18 predicts lung diseas e worsening in sy stemic
scle
rosis. Eur Respir J 38:13551360. doi:10.1183/09031936.
00004711
99. Schupp J, Becker M, Günther J et al (2014) Serum CCL18 is
predictive for lung disease progression and mortality in systemic
sclerosis. Eur Respir J 43:15301532. doi:10.1183/09031936.
00131713
100. Elhaj M, Charles J, Pedroza C et al (2013) Can serum surfactant
protein d or cc-chemokine ligand 18 predict outcome of interstitial
lung disease in patients with early systemic sclerosis? J Rheumatol
40:11141 120. doi:10.3899/jrheum.120997
101. Kowal-Bielecka O, Kowal K, Lewszuk A et al (2005) Beta
thromboglobulin and platelet factor 4 in bronchoalveolar lavage
fluid of patients with systemic sclerosis. Ann Rheum Dis 64:484
486. doi:10.1136/ard.2004.022970
102. Eun BL, Zhao J, Jeong YK et al (2007) Evidence of potential
interaction of chemokine genes in susceptibility to systemic scle-
rosis. Arthritis Rheum 56:24432448. doi:10.1002/art.22742
103. Salim PH, Jobim M, Bredemeier M et al (2012) Combined effects
of CXCL8 and CXCR2 gene polymorphisms on susceptibility to
systemic sclerosis. Cytokine 60:473 477. doi:10.1016/j.cyto.
2012.05.026
104. Furuse S, Fujii H, Kaburagi Yet al (2003) Serum concentrations of
the CXC chemokines interleukin 8 and growth-regulated onco-
gene-alpha are elevated in patients with systemic sclerosis . J
Rheumatol 30:15241528
105. Schmidt K, Martinez-Gamboa L, Meier S et al (2009)
Bronchoalveolar lavage fluid cytokines and chemokines as
markers and predictors for the outcome of interstitial lung disease
in systemic sclerosis patients. Arthritis Res Ther 11:R111. doi:10.
1186/ar2766
106. Hasegawa M, Asano Y, Endo H et al (2012) Serum chemokine
levels as prognostic markers in patients with early systemic scle-
rosis: a multicenter, prospective, observational study. Mod
Rheumatol 9:10761084. doi:10.1007/s10165-012-0795-6
107. Vogl T, Tenbrock K, Ludwig S et al (2007) Mrp8 and Mrp14 are
endogenous activators of Toll-like receptor 4, promoting lethal,
endotoxin-induced shock. Nat Med 13:10421049. doi:10.1038/
nm1638
108. Kuruto R, Nozawa R, Takeishi K et al (1990) Myeloid calcium
binding proteins: expression in the differentiated HL-60 cells and
detection in sera of patients with connective tissue diseases. J
Biochem 108:650653
109. Xu X, Wu WY, Tu WZ et al (2013) Increased expression of
S100A8 and S100A9 in patients with diffuse cutaneous systemic
sclerosis. A correlation with organ involvement and immunolog-
ical abnormalities. Clin Rheumatol 32:15011510. doi:10.1007/
s10067-013-2305-4
110. Andréasson K, Scheja A, Saxne T et al (2011) Faecal calprotectin:
a biomarker of gastrointestinal disease in systemic sclerosis. J
Intern Med 270:5057. doi:10.1111/j.1365-2796.2010.02340.x
111. Andréasson K, Saxne T, Scheja A et al (2014) Faecal levels of
calprotectin in systemic sclerosis are stable over time and are
higher compared to primary Sjogrens syndrome and rheumatoid
arthritis. Arthritis Res Ther 16:R46. doi:10.1186/ar4475
112. Giusti L, Bazzichi L, Baldini C et al (2007) Specific proteins
identified in whole saliva from patients with diffuse systemic scle-
rosis. J Rheumatol 34:20632069
113. Nikitorow icz-Buniak J, Shiwe n X, Denton CP et al (2014)
Abnormally differentiating keratinocytes in the epidermis of sys-
temic sclerosis patients show enhanced secretion of CCN2 and
S100A9. J Investig Dermatol 134:110. doi:10.1038/jid.2014.253
114. Van Bon L, Cossu M, Loof A et al (2014) Proteomic analysis of
plasma identifies the toll-like receptor agonists S100A8/A9 as a
novel possible marker for systemic sclerosis phenotype. Ann
Rheum Dis 73:1585
1589. doi:10.1
136/annrheumdis-2013-
205013
486 Semin Immunopathol (2015) 37:475487
115. Bhattacharyya S, Kelley K, Melichian D et al (2012) Toll-like
receptor 4 signaling augments transforming growth factor re-
sponses: a novel mechanism for maintaining and amplifying fibro-
sis in scleroderma. Am J Pathol. doi:10.1016/j.ajpath.2012.09.007
116. Stifano G, Affandi AJ, Mathes AL et al (2014) Chronic toll-like
receptor 4 stimulation in skin induces inflammation, macrophage
activation, transforming growth factor beta signature gene expres-
sion, and fibrosis. Arthritis Res Ther 16:R136. doi:10.1186/ar4598
117. Takahashi T, Asano Y, Ichimura Y et al (2015) Amelioration of
tissue fibrosis by toll-like receptor 4 knockout in murine models of
systemic sclerosis. Arthritis Rheumatol 67:254265. doi:10.1002/
art.38901
118. Asano Y, Ihn H, Yamane K et al (2001) Clinical significance of
surfactant protein D as a serum marker for evaluating pulmonary
fibrosis in patients with systemic sclerosis. Arthritis Rheum 44:
13631369. doi:10.1002/1529-0131(200106)44:6<1363::AID-
ART229>3.0.CO;2[-]5
119. Hant FN, Ludwicka-Bradley A, Wang H-J et al (2009) Surfactant
protein D and KL-6 as serum biomarkers of interstitial lung dis-
ease in patients with scleroderma. J Rheumatol 36:773780. doi:
10.3899/jrheum.080633
120. Mukerjee D, St George D, Knight C et al (2004)
Echocardiography and pulmonary function as screening tests
for p ulmonary arterial hypertension in systemic sclerosis.
Rheumatology (Oxford) 43:461 466. doi:10.1093/
rheumatology/keh067
121. Williams MH, Handler CE, Akram R et al (2006) Role of N-
terminal brain natriuretic peptide (N-TproBNP) in scleroderma-
associated pulmonary arterial hypertension. Eur Heart J 27:
14851494. doi:10.1093/eurheartj/ehi891
122. Choi HJ, Shin YK, Lee HJ et al (2008) The clinical significance of
serum N-terminal pro-brain natriuretic peptide in systemic sclero-
sis patients. Clin Rheumatol 27:437442. doi:10.1007/s10067-
007-0724-9
123. Cavagna L, Caporali R, Klersy C et al (2010) Comparison of brain
natriuretic peptide (BNP) and NT-proBNP in screening for pulmo-
nary arterial hypertension in patients with systemic sclerosis. J
Rheumatol 37:20642070. doi:10.3899/jrheum.090997
124. Elshamy HA, Ibrahim SE, Farouk HM et al (2011) N-terminal
pro-brain natriuretic peptide in systemic sclerosis: new insights.
Eur J Dermatol 21:686690. doi:10.1684/ejd.2011.1423
125. Thakkar V, Stevens WM, Prior D et al (2012) N-terminal pro-brain
natriuretic peptide in a novel screening algorithm for pulmonary
arterial hypertension in systemic sclerosis: a casecontrol study.
Arthritis Res Ther 14:R143. doi:10.1186/ar3876
126. Oravec RM, Bredemeier M, Laurino CC et al (2010) NT-proBNP
levels in systemic sclerosis: association with clinical and laborato-
ry abnormalities . Clin Bioche m 43:745749. doi:10.1016/j.
clinbiochem.2010.03.011
127. Coghlan JG, Denton CP, Grünig E et al (2014) Evidence-based
detection of pulmonary arterial hypertension in systemic sclerosis:
the DETECT study. Ann Rheum Dis 73:13401349. doi:10.1136/
annrheumdis-2013-203301
128. Allanore Y, Borderie D, Avouac J et al (2008) High N-terminal
pro-brain natriuretic peptide levels and low diffusing capacity for
carbon monoxide as independent predictors of the occurrence of
precapillary pulmonary arterial hypertension in patients with sys-
temic sclerosis. Arthritis Rheum 58:284291. doi:10.1002/art.
23187
129. Schmidt J, Launay D, Soudan B et al (2007) Assessment of plas-
ma endothelin level measurement in systemic sclerosis. Rev Med
Interne 28:371376. doi:10.1016/j.revmed.2006.12.019
130. Pendergrass SA, Hayes E, Farina G et al (2010) Limited systemic
sclerosis patients with pulmonary arterial hypertension show bio-
markers of inflammation and vascular injury. PLoS One. doi:10.
1371/journal.pone.0012106
131. Papaioannou AI, Zakynthinos E, Kostikas K et al (2009) Serum
VEGF levels are related to the presence of pulmonary arterial
hyp
ertension in systemic sclerosis. BMC Pulm Med 9:18. doi:
10.1186/1471-2466-9-18
132. Morelli S, Ferri C, Di Francesco L et al (1995) Plasma endothelin-
1 levels in patients with systemic sclerosis: influence of pulmonary
or systemic arterial hypertension. Ann Rheum Dis 54:730734.
doi:10.1136/ard.54.9.730
133. Scheja A, Akesson A, Geborek P et al (2001) Von Willebrand
factor propeptide as a marker of disease activity in systemic scle-
rosis (scleroderma). Arthritis Res 3:178182
134. Iannone F, Riccardi MT, Guiducci S et al (2008) Bosentan regu-
lates the expression of adhesion molecules on circulating T cells
and serum soluble adhesion molecules in systemic sclerosis-
associated pulmonary arterial hypertension. Ann Rheum Dis 67:
11211 126. doi:10.1136/ard.2007.080424
135. Jenkins PV, ODonnell JS (2006) ABO blood group determines
plasma von Willebrand factor levels: a biologic function after all?
Transfusion 46:18361844. doi:1 0.1111/j.1537- 2995 .2006.
00975.x
136. Van Loon JE, Kavousi M, Leebeek FWG et al (2012) von
Willebrand factor plasma levels, genetic variations and coronary
heart disease in an older population. J Thromb Haemost 10:1262
1269. doi:10.1111/j.1538-7836.2012.04771.x
137. Rajkumar R, Konishi K, Richards TJ et al (2010) Genomewide
RNA expression profiling in lung identifies distinct signatures in
idiopathic pulmonary arterial hypertension and secondary pulmo-
nary hypertension. Am J Physiol Heart Circ Physiol 298:H1235
H1248. doi:10.1152/ajpheart.00254.2009
138. Zabini D, Nagaraj C, Stacher E et al (2012) Angiostatic factors in
the pulmonary endarterectomy material from chronic thromboem-
bolic pulmonary hypertension patients cause endothelial dysfunc-
tion. PLoS One. doi:10.1371/journal.pone.0043793
139. Meadows CA, Risbano MG, Zhang L et al (2011) Increased ex-
pression of growth differentiation factor-15 in systemic sclerosis-
associated pulmonary arterial hypertension. Chest 139:9941002.
doi:10.1378/chest.10-0302
140. Nickel N, Kempf T, Tapken H et al (2008) Growth differentiation
factor-15 in idiopathic pulmonary arterial hypertension. Am J
Respir Crit Care Med 178:534541. doi:10.1164/rccm.200802-
235OC
141. Korman BD, Criswell LA (2015) Recent advances in the genetics
of systemic. Toward Biological and Clinical Significance. Curr
Rheumatol Rep, Sclerosis. doi:10.1007/s11926-014-0484-x
142. Bălănescu P, Lădaru A, Bălănescu E et al (2014) Systemic sclero-
sis biomarkers discovered using mass-spectrometry-based proteo-
mics: a systematic review. Biomarkers 19:345355. doi:10.3109/
1354750X.2014.920046
143. Limpers A, van Royen-Kerkhof A, van Roon JA et al (2014)
Overlapping gene expression profiles indicative of antigen pro-
cessing and the interferon pathway characterize inflammatory fi-
brotic skin diseases. Expert Rev Clin Immunol. doi:10.1586/
1744666X.2014.872561
Semin Immunopathol (2015) 37:475487 487
... The diagnosis of SSc relies on clinical evaluation and detection of antibodies that recognize the nucleus of the patient's own cells, the so-called antinuclear antibodies (ANAs) [17,18]. ANAs recognize nuclear components and are detected in as many as 95% of SSc patients [19]. ...
... ANAs specific for SSc recognize topoisomerase I, an enzyme that breaks single stranded DNA, relaxes supercoiled DNA, and facilitates chromosome condensation [18]. Antitopoisomerase-I antibodies (ATAs; anti-Scl-70) are mostly found in dcSSc patients (prevalence 20-40%), and correlate with poor prognosis, lung fibrosis, and disease progression [17,18]. Anti-RNA polymerase III autoantibodies are detected in dcSSc patients (prevalence 10-25%), and serve as markers of rapid disease development and renal crisis [17,18]. ...
... Antitopoisomerase-I antibodies (ATAs; anti-Scl-70) are mostly found in dcSSc patients (prevalence 20-40%), and correlate with poor prognosis, lung fibrosis, and disease progression [17,18]. Anti-RNA polymerase III autoantibodies are detected in dcSSc patients (prevalence 10-25%), and serve as markers of rapid disease development and renal crisis [17,18]. ...
Chapter
Full-text available
Fibrosis in systemic sclerosis (SSc or scleroderma) is characterized by an abundance of chromosome segregation defects and chromosome instability (CIN) that lead to overactivation of autoimmunity and inflammation. This chapter will emphasize the most recent findings on the involvement of centromere and telomere dysfunction in scleroderma. We will discuss how centromere and telomere dysfunction contribute to CIN, fibrosis, and cellular autoimmunity in scleroderma. We will also summarize how chromosome segregation defects in the form of aneuploidy and micronuclei formation activate the Cyclic GMP–AMP synthase (cGAS) Stimulator of interferon genes (STING) pathway of cellular immunity. Activation of this pathway induces production of inflammatory cytokines IFNβ and IL6. Finally, we will summarize the most recent therapies to block the cGAS-STING pathway and treat fibrosis.
... SSc diagnosis relies on clinical evaluation accompanied by screening for antinuclear antibodies (ANAs) 14,15 . Anti-Topoisomerase-I antibodies (ATAs) (usually called anti-Scl-70) are found predominately in dcSSc patients (prevalence 20-40%), and are associated with poor prognosis, pulmonary fibrosis, and disease progression [14][15][16] . ...
... SSc diagnosis relies on clinical evaluation accompanied by screening for antinuclear antibodies (ANAs) 14,15 . Anti-Topoisomerase-I antibodies (ATAs) (usually called anti-Scl-70) are found predominately in dcSSc patients (prevalence 20-40%), and are associated with poor prognosis, pulmonary fibrosis, and disease progression [14][15][16] . Anticentromere antibodies (ACAs) are found in up to 43% of lcSSc, previously known as the CREST variant (CREST: calcinosis, Raynaud's phenomenon, esophageal dysmotility, sclerodactyly, and telangiectasia) 16 . ...
... ACAs recognize a large proportion of the centromere proteins (CENPA, CENPB, CENPC, and so on, to CENPT) and, indeed, most of the key proteins involved in centromere formation were discovered using antibodies from SSc patients 16 . ACAs are associated with a generally more favorable prognosis as compared with positivity for ATAs [14][15][16] , but patients with the former have a high predilection for pulmonary arterial hypertension. Anti-RNA polymerase I and III antibodies are detected in 10-25% of dcSSc patients and are used as a predictive marker of rapid onset of the disease, skin thickening and renal crisis [2][3][4] . ...
Article
Full-text available
Centromere defects in Systemic Sclerosis (SSc) have remained unexplored despite the fact that many centromere proteins were discovered in patients with SSc. Here we report that lesion skin fibroblasts from SSc patients show marked alterations in centromeric DNA. SSc fibroblasts also show DNA damage, abnormal chromosome segregation, aneuploidy (only in diffuse cutaneous (dcSSc)) and micronuclei (in all types of SSc), some of which lose centromere identity while retaining centromere DNA sequences. Strikingly, we find cytoplasmic “leaking” of centromere proteins in limited cutaneous SSc (lcSSc) fibroblasts. Cytoplasmic centromere proteins co-localize with antigen presenting MHC Class II molecules, which correlate precisely with the presence of anti-centromere antibodies. CENPA expression and micronuclei formation correlate highly with activation of the cGAS-STING/IFN-β pathway as well as markers of reactive oxygen species (ROS) and fibrosis, ultimately suggesting a link between centromere alterations, chromosome instability, SSc autoimmunity, and fibrosis.
... Several works have previously tried to address this issue. [6][7][8][9][10][11][12] Most of them focused on identifying surrogates for haemodynamic diagnostic parameters, with biomarkers that could accurately discriminate SSc patients with and without PAH. [7][8][9][10] Few studies, however, have tried to find surrogate markers for severity parameters, such as PVR. ...
... [6][7][8][9][10][11][12] Most of them focused on identifying surrogates for haemodynamic diagnostic parameters, with biomarkers that could accurately discriminate SSc patients with and without PAH. [7][8][9][10] Few studies, however, have tried to find surrogate markers for severity parameters, such as PVR. 6 11 This is a matter of importance, since PVR reflects the ongoing vascular remodelling underlying the disease progression and can be used to guide treatment initiation and assess therapeutic efficacy. ...
Article
Full-text available
Objectives To mine the serum proteome of patients with systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH) and to detect biomarkers that may assist in earlier and more effective diagnosis and treatment. Methods Patients with limited cutaneous SSc, no extensive interstitial lung disease and no PAH-specific therapy were included. They were classified as cases if they had PAH confirmed by right heart catheterisation (RHC) and serum collected on the same day as RHC; and as controls if they had no clinical evidence of PAH. Results Patients were mostly middle-aged females with anticentromere-associated SSc. Among 1129 proteins assessed by a high-throughput proteomic assay (SOMAscan), only 2 were differentially expressed and correlated significantly with pulmonary vascular resistance (PVR) in SSc-PAH patients (n=15): chemerin ( ρ =0.62, p=0.01) and SET ( ρ =0.62, p=0.01). To validate these results, serum levels of chemerin were measured by ELISA in an independent cohort. Chemerin levels were confirmed to be significantly higher (p=0.01) and correlate with PVR ( ρ =0.42, p=0.04) in SSc-PAH patients (n=24). Chemerin mRNA expression was detected in fibroblasts, pulmonary artery smooth muscle cells (PA-SMCs)/pericytes and mesothelial cells in SSc-PAH lungs by single-cell RNA-sequencing. Confocal immunofluorescence revealed increased expression of a chemerin receptor, CMKLR1, on SSc-PAH PA-SMCs. SSc-PAH serum seemed to induce higher PA-SMC proliferation than serum from SSc patients without PAH. This difference appeared neutralised when adding the CMKLR1 inhibitor α-NETA. Conclusion Chemerin seems an interesting surrogate biomarker for PVR in SSc-PAH. Increased chemerin serum levels and CMKLR1 expression by PA-SMCs may contribute to SSc-PAH pathogenesis by inducing PA-SMC proliferation.
... However, the diagnosis of SSc is not always straightforward given the overlap with other autoimmune conditions, particularly in the early stages of the disease (10)(11)(12). These challenges have prompted the search for novel biomarkers of SSc and its complications, e.g., interstitial lung disease (ILD), pulmonary arterial hypertension (PAH), and digital ulcers (DU), to enhance diagnosis and management (13)(14)(15)(16)(17). ...
Article
Full-text available
Introduction The identification of new, easily measurable biomarkers might assist clinicians in diagnosing and managing systemic sclerosis (SSc). Although the full blood count is routinely assessed in the evaluation of SSc, the diagnostic utility of specific cell-derived inflammatory indices, i.e., neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and monocyte-to-lymphocyte ratio (MLR), has not been critically appraised in this patient group. Methods We conducted a systematic review and meta-analysis of studies investigating the NLR, PLR, and MLR, in SSc patients and healthy controls and in SSc patients with and without relevant complications. PubMed, Scopus, and Web of Science were searched from inception to 23 February 2024. Risk of bias and certainty of evidence were assessed using validated tools. Results In 10 eligible studies, compared to controls, patients with SSc had significantly higher NLR (standard mean difference, SMD=0.68, 95% CI 0.46 to 0.91, p<0.001; I² = 74.5%, p<0.001), and PLR values (SMD=0.52, 95% CI 0.21 to 0.83, p=0.001; I² = 77.0%, p=0.005), and a trend towards higher MLR values (SMD=0.60, 95% CI -0.04 to 1.23, p=0.066; I² = 94.1%, p<0.001). When compared to SSc patients without complications, the NLR was significantly higher in SSc with interstitial lung disease (ILD, SMD=0.31, 95% CI 0.15 to 0.46, p<0.001; I² = 43.9%, p=0.11), pulmonary arterial hypertension (PAH, SMD=1.59, 95% CI 0.04 to 3.1, p=0.045; I² = 87.6%, p<0.001), and digital ulcers (DU, SMD=0.43, 95% CI 0.13 to 0.74, p=0.006; I² = 0.0%, p=0.49). The PLR was significantly higher in SSc patients with ILD (SMD=0.42, 95% CI 0.25 to 0.59, p<0.001; I² = 24.8%, p=0.26). The MLR was significantly higher in SSc patients with PAH (SMD=0.63, 95% CI 0.17 to 1.08, p=0.007; I² = 66.0%, p=0.086), and there was a trend towards a higher MLR in SSc patients with ILD (SMD=0.60, 95% CI -0.04 to 1.23, p=0.066; I² = 94.1%, p<0.001). Discussion Pending the results of appropriately designed prospective studies, the results of this systematic review and meta-analysis suggest that blood cell-derived indices of inflammation, particularly the NLR and PLR, may be useful in the diagnosis of SSc and specific complications. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024520040.
... Aberrant expression of miRNAs is closely linked to the pathophysiology of diseases, suggesting that circulating miRNAs are potential biomarkers and therapeutic targets [12]. Studies have reported an altered miRNA profile in the serum of SSc patients, with differentially expressed miRNAs implicated in various fibrotic processes [13][14][15], indicating a potential role for miRNAs in SSc development. Based on the study of the miRNA profile that changes in disease conditions, the development of therapeutics using the form of miRNA mimics or anti-miRs is considered a promising treatment strategy. ...
Article
Full-text available
Background MicroRNA (miRNA)-21-5p participates in various biological processes, including cancer and autoimmune diseases. However, its role in the development of fibrosis in the in vivo model of systemic sclerosis (SSc) has not been reported. This study investigated the effects of miRNA-21a-5p overexpression and inhibition on SSc fibrosis using a bleomycin-induced SSc mouse model. Methods A murine SSc model was induced by subcutaneously injecting 100 μg bleomycin dissolved in 0.9% NaCl into C57BL/6 mice daily for 5 weeks. On days 14, 21, and 28 from the start of bleomycin injection, 100 μg pre-miRNA-21a-5p or anti-miRNA-21a-5p in 1 mL saline was hydrodynamically injected into the mice. Fibrosis analysis was conducted in lung and skin tissues of SSc mice using hematoxylin and eosin as well as Masson’s trichrome staining. Immunohistochemistry was used to examine the expression of inflammatory cytokines, phosphorylated signal transducer and activator of transcription-3 (STAT3) at Y705 or S727, and phosphatase and tensin homologue deleted on chromosome-10 (PTEN) in skin tissues of SSc mice. Results MiRNA-21a-5p overexpression promoted lung fibrosis in bleomycin-induced SSc mice, inducing infiltration of cells expressing TNF-α, IL-1β, IL-6, or IL-17, along with STAT3 phosphorylated cells in the lesional skin. Conversely, anti-miRNA-21a-5p injection improved fibrosis in the lung and skin tissues of SSc mice, reducing the infiltration of cells secreting inflammatory cytokines in the skin tissue. In particular, it decreased STAT3-phosphorylated cell infiltration at Y705 and increased the infiltration of PTEN-expressing cells in the skin tissue of SSc mice. Conclusion MiRNA-21a-5p promotes fibrosis in an in vivo murine SSc model, suggesting that its inhibition may be a therapeutic strategy for improving fibrosis in SSc.
... . In this in vitro assay, we confirmed that Hs27 cells activated with TGF-β express higher levels of the fibrotic markers Col1 and α-SMA compared to nonactivated (healthy) Hs27 cells. 27 Figure 1C shows a reduction of both Col1 and α-SMA protein expression that was induced by TGF-β. After only 6 h of treatment with COA/NAVI, the amount of both Col1 and α-SMA was reduced by 16 and 31%, as shown in Figure 1D. ...
Article
Pathological fibrosis is distinguished from physiological wound healing by persistent myofibroblast activation, suggesting that therapies that induce myofibroblast apoptosis selectively could prevent progression and potentially reverse the established fibrosis, such as for scleroderma (a heterogeneous autoimmune disease characterized by multiorgan fibrosis). Navitoclax (NAVI) is a BCL-2/BCL-xL inhibitor with antifibrotic properties and has been investigated as a potential therapeutic for fibrosis. NAVI makes myofibroblasts particularly vulnerable to apoptosis. However, despite NAVI's significant potency, clinical translation of BCL-2 inhibitors, NAVI in this case, is hindered due to the risk of thrombocytopenia. Therefore, in this work, we utilized a newly developed ionic liquid formulation of NAVI for direct topical application to the skin, thereby avoiding systemic circulation and off-target-mediated side effects. The ionic liquid composed of choline and octanoic acid (COA) at a 1:2 molar ionic ratio increases skin diffusion and transportation of NAVI and maintains their retention within the dermis for a prolonged duration. Topical administration of NAVI-mediated BCL-xL and BCL-2 inhibition results in the transition of myofibroblast to fibroblast and ameliorates pre-existing fibrosis, as demonstrated in a scleroderma mouse model. We have observed a significant reduction of α-SMA and collagen, which are known as fibrosis marker proteins, as a result of the inhibition of anti-apoptotic proteins BCL-2/BCL-xL. Overall, our findings show that COA-assisted topical delivery of NAVI upregulates apoptosis specific to myofibroblasts, with minimal presence of the drug in the systemic circulation, resulting in an accelerated therapeutic effect with no discernible drug-associated toxicity.
... Microvascular changes with endothelial disfunction, epigenetics, and complex autoimmune responses are the major factors contributing to the development of the disease [1]. In systemic sclerosis, vascular damage plays a pivotal role in the pathogenesis of tissue fibrosis; therefore, identifying markers of endothelial disfunction is crucial to help optimize patients assessments and to develop novel treatment methods aimed at mitigating progressive fibrosis [2][3][4][5]. One of the therapeutic aims is to stabilize the progression of microangiopathy, i.e., the loss of capillaries over time, which correlates with the course of disease [6]. ...
Article
Full-text available
Background: Systemic sclerosis (SSc) is a connective tissue disease manifesting with progressive fibrosis of the skin and internal organs. Its pathogenesis is strictly associated with vascular disfunction and damage. Salusin-α and salusin-β, endogenous peptides regulating secretion of pro-inflammatory cytokines and vascular smooth muscle proliferation, may potentially play a role in SSc pathogenesis. Objectives: The aim of this study was to assess the concentration of salusins in sera of patients with SSc and healthy controls and to evaluate correlations between the salusins levels and selected clinical parameters within the study group. Materials and methods: 48 patients with SSc (44 women; mean age, 56.4, standard deviation, 11.4) and 25 adult healthy volunteers (25 women; mean age, 55.2, standard deviation, 11.2) were enrolled. All patients with SSc were treated with vasodilators and twenty-seven of them (56%) also received immunosuppressive therapy. Results: Circulating salusin-α was significantly elevated in patients with SSc in comparison to healthy controls (U = 350.5, p = 0.004). Patients with SSc receiving immunosuppression had higher serum salusin-α concentrations compared with those without immunosuppressive therapy (U = 176.0, p = 0.026). No correlation was observed between salusins concentrations and skin or internal organ involvement parameters. Conclusions: Salusin-α, a bioactive peptide mitigating the endothelial disfunction, was elevated in patients with systemic sclerosis receiving vasodilators and immunosuppressants. Increased salusin-α concertation may be associated with the initiation of atheroprotective processes in patients with SSc managed pharmacologically, which requires verification in future studies.
Article
Full-text available
Systemic sclerosis (SSc) is a complex autoimmune disease characterized by vascular damage, vasoinstability, and decreased perfusion with ischemia, inflammation, and exuberant fibrosis of the skin and internal organs. Biomarkers are analytic indicators of the biological and disease processes within an individual that can be accurately and reproducibly measured. The field of biomarkers in SSc is complex as recent studies have implicated at least 240 pathways and dysregulated proteins in SSc pathogenesis. Anti-nuclear antibodies (ANA) are classical biomarkers with well-described clinical classifications and are present in more than 90% of SSc patients and include anti-centromere, anti-Th/To, anti-RNA polymerase III, and anti-topoisomerase I antibodies. Transforming growth factor-β (TGF-β) is central to the fibrotic process of SSc and is intimately intertwined with other biomarkers. Tyrosine kinases, interferon-1 signaling, IL-6 signaling, endogenous thrombin, peroxisome proliferator-activated receptors (PPARs), lysophosphatidic acid receptors, and amino acid metabolites are new biomarkers with the potential for developing new therapeutic agents. Other biomarkers implicated in SSc-ILD include signal transducer and activator of transcription 4 (STAT4), CD226 (DNAX accessory molecule 1), interferon regulatory factor 5 (IRF5), interleukin-1 receptor–associated kinase-1 (IRAK1), connective tissue growth factor (CTGF), pyrin domain containing 1 (NLRP1), T-cell surface glycoprotein zeta chain (CD3ζ) or CD247, the NLR family, SP-D (surfactant protein), KL-6, leucine-rich α2-glycoprotein-1 (LRG1), CCL19, genetic factors including DRB1 alleles, the interleukins (IL-1, IL-4, IL-6, IL-8, IL-10 IL-13, IL-16, IL-17, IL-18, IL-22, IL-32, and IL-35), the chemokines CCL (2,3,5,13,20,21,23), CXC (8,9,10,11,16), CX3CL1 (fractalkine), and GDF15. Adiponectin (an indicator of PPAR activation) and maresin 1 are reduced in SSc patients. A new trend has been the use of biomarker panels with combined complex multifactor analysis, machine learning, and artificial intelligence to determine disease activity and response to therapy. The present review is an update of the various biomarker molecules, pathways, and receptors involved in the pathology of SSc.
Preprint
Full-text available
Introduction : Systemic sclerosis is a connective tissue disease with unpredictable symptoms. The GI tract is often affected, with calprotectin levels indicating inflammation. We are examining fecal calprotectin as a simple method to diagnose GI disorders and disease activity. Materials and methods We gathered information from the 39 patients by filling out a UCLA SCTS 2.0 questionnaire regarding their GI symptoms. Additionally, we examined their skin and determined their skin score. Blood samples were taken to assess malnutrition, and stool samples were collected to measure the calprotectin level. We also conducted a non-aggressive Barium Swallow and CT Enterography to check for GI disorders. Results Increased levels of FC didn’t have any correlation with age (P Value = 0.79), disease type (P Value = 0.59), slight intestine wall thickening (P Value = 0.10), esophageal dilatation (P Value = 0.18), skin score (P Value = 0.25), final GI score (P Value = 0.30) and none of the serum tests. In our study, just 5 cases had evidence of wall thickening in CT Enterography, and all those 5 cases had increased levels of calprotectin. Also, as most of our patients take vitamin and mineral supplements, we couldn’t find any signs of micronutrient deficiencies or correlation with FC level. Discussion and Conclusion While no statistical correlation was found between FC level and the variables, the results suggest that FC might be a specific tool for assessing intestinal wall thickness. However, due to the limited sample size, further studies are necessary to validate these findings.
Article
Autoimmune diseases (AID), such as systemic lupus erythematosus (SLE) and systemic sclerosis (SS), are complex conditions involving immune system dysregulation. Diagnosis is challenging, requiring biomarkers for improved detection and prediction of relapses. Lipids have emerged as potential biomarkers due to their role in inflammation and immune response. This study uses an untargeted C18 RP-LC-MS lipidomics approach to comprehensively assess changes in lipid profiles in patients with SLE and SS. By analyzing whole blood and plasma, the study aims to simplify the lipidomic analysis, explore cellular-level lipids, and compare lipid signatures of SLE and SS with healthy controls. Our findings showed variations in the lipid profile of SLE and SS. Sphingomyelin and ceramide molecular species showed significant increases in plasma samples from SS patients, suggesting an atherosclerotic profile and potentially serving as lipid biomarkers. Phosphatidylserine species in whole blood from SLE patients exhibited elevated levels supporting previously reported dysregulated processes of cell death and defective clearance of dying cells in this AID. Moreover, decreased phospholipids bearing PUFA were observed, potentially attributed to the degradation of these species through lipid peroxidation processes. Further studies are needed to better understand the role of lipids in the pathological mechanisms underlying SLE and SS.
Article
Full-text available
Significant advances have been made in understanding the genetic basis of systemic sclerosis (SSc) in recent years. Genomewide association and other large-scale genetic studies have identified 30 largely immunity-related genes which are significantly associated with SSc. We review these studies, along with genomewide expression studies, proteomic studies, genetic mouse models, and insights from rare sclerodermatous diseases. Collectively, these studies have begun to identify pathways that are relevant to SSc pathogenesis. The findings presented in this review illustrate how both genetic and genomic aberrations play important roles in the development of SSc. However, despite these recent discoveries, there remain major gaps between current knowledge of SSc, a unified understanding of pathogenesis, and effective treatment. To this aim, we address the important issue of SSc heterogeneity and discuss how future research needs to address this in order to develop a clearer understanding of this devastating and complex disease.
Article
Full-text available
Objective Earlier detection of pulmonary arterial hypertension (PAH), a leading cause of death in systemic sclerosis (SSc), facilitates earlier treatment. The objective of this study was to develop the first evidence-based detection algorithm for PAH in SSc. Methods In this cross-sectional, international study conducted in 62 experienced centres from North America, Europe and Asia, adults with SSc at increased risk of PAH (SSc for >3 years and predicted pulmonary diffusing capacity for carbon monoxide <60%) underwent a broad panel of non-invasive assessments followed by diagnostic right heart catheterisation (RHC). Univariable and multivariable analyses selected the best discriminatory variables for identifying PAH. After assessment for clinical plausibility and feasibility, these were incorporated into a two-step, internally validated detection algorithm. Nomograms for clinical practice use were developed. Results Of 466 SSc patients at increased risk of PAH, 87 (19%) had RHC-confirmed PAH. PAH was mild (64% in WHO functional class I/II). Six simple assessments in Step 1 of the algorithm determined referral to echocardiography. In Step 2, the Step 1 prediction score and two echocardiographic variables determined referral to RHC. The DETECT algorithm recommended RHC in 62% of patients (referral rate) and missed 4% of PAH patients (false negatives). By comparison, applying European Society of Cardiology/European Respiratory Society guidelines to these patients, 29% of diagnoses were missed while requiring an RHC referral rate of 40%. Conclusions The novel, evidence-based DETECT algorithm for PAH detection in SSc is a sensitive, non-invasive tool which minimises missed diagnoses, identifies milder disease and addresses resource usage.
Article
Full-text available
OBJECTIVE: Earlier detection of pulmonary arterial hypertension (PAH), a leading cause of death in systemic sclerosis (SSc), facilitates earlier treatment. The objective of this study was to develop the first evidence-based detection algorithm for PAH in SSc. METHODS: In this cross-sectional, international study conducted in 62 experienced centres from North America, Europe and Asia, adults with SSc at increased risk of PAH (SSc for >3 years and predicted pulmonary diffusing capacity for carbon monoxide <60%) underwent a broad panel of non-invasive assessments followed by diagnostic right heart catheterisation (RHC). Univariable and multivariable analyses selected the best discriminatory variables for identifying PAH. After assessment for clinical plausibility and feasibility, these were incorporated into a two-step, internally validated detection algorithm. Nomograms for clinical practice use were developed. RESULTS: Of 466 SSc patients at increased risk of PAH, 87 (19%) had RHC-confirmed PAH. PAH was mild (64% in WHO functional class I/II). Six simple assessments in Step 1 of the algorithm determined referral to echocardiography. In Step 2, the Step 1 prediction score and two echocardiographic variables determined referral to RHC. The DETECT algorithm recommended RHC in 62% of patients (referral rate) and missed 4% of PAH patients (false negatives). By comparison, applying European Society of Cardiology/European Respiratory Society guidelines to these patients, 29% of diagnoses were missed while requiring an RHC referral rate of 40%. CONCLUSIONS: The novel, evidence-based DETECT algorithm for PAH detection in SSc is a sensitive, non-invasive tool which minimises missed diagnoses, identifies milder disease and addresses resource usage.
Article
Objectives Extent of systemic sclerosis (SSc)-related interstitial lung disease (ILD) assessed from thoracic high-resolution CT (HRCT) predicts disease course, mortality and treatment response. While quantitative HRCT analyses of extent of lung fibrosis (QLFib) or total interstitial lung disease (QILD) are more sensitive and reproducible than visual HRCT assessments of SSc-ILD, these analyses are not widely available. This study evaluates the relationship between clinical disease parameters and QLFib and QILD scores to identify potential surrogate measures of radiographic extent of ILD. Methods Using baseline data from the Scleroderma Lung Study I (SLS I; N=158), multivariate regression analyses were performed using the best subset selection method to identify one to five variable models that best correlated with QLFib and QILD scores in both whole lung (WL) and the zone of maximal involvement (ZM). These models were subsequently validated using baseline data from SLS II (N=142). Bivariate analyses of the radiographic and clinical variables were also performed using pooled data. SLS I and II did not include patients with clinically significant pulmonary hypertension (PH). Results Diffusing capacity for carbon monoxide (DLCO) was the single best predictor of both QLF and QILD in the WL and ZM in all of the best subset models. Adding other disease parameters to the models did not substantially improve model performance. Forced vital capacity (FVC) did not predict QLF or QILD scores in any of the models. Conclusions In the absence of PH, DLCO provides the best overall estimate of HRCT-measured lung disease in patients from two large SSc cohorts. FVC, although commonly used, may not be the best surrogate measure of extent of SSc-ILD at any point in time. Trial registration numbers SLS I: www.clinicaltrials.gov NCT 00000-4563; SLS II: www.clinicaltrials.gov NCT 00883129.
Article
Objective: Bleomycin-induced fibrosis and the tight skin (TSK/+) mouse are well-established experimental murine models of human systemic sclerosis (SSc). Growing evidence has demonstrated the pivotal role of Toll-like receptors (TLRs) in several autoimmune inflammatory diseases, including SSc. This study was undertaken to determine the role of TLR-4 in the fibrotic processes in these murine models. Methods: We generated a murine model of bleomycin-induced SSc using TLR-4(-/-) mice and TLR-4(-/-) ;TSK/+ mice. The mechanisms by which TLR-4 contributes to pathologic tissue fibrosis were investigated in these 2 models by histologic examination, hydroxyproline assay, enzyme-linked immunosorbent assay, real-time polymerase chain reaction, and flow cytometry. Results: Dermal and lung fibrosis was attenuated in bleomycin-treated TLR-4(-/-) mice compared with their wild-type counterparts. Inflammatory cell infiltration, expression of various inflammatory cytokines, and pathologic angiogenesis induced by bleomycin treatment were suppressed with TLR-4 deletion. Furthermore, the increased expression of interleukin-6 (IL-6) in fibroblasts, endothelial cells, and immune cells in response to bleomycin in vivo and to lipopolysaccharide in vitro was notably abrogated in the absence of TLR-4. Moreover, TLR-4 deletion was associated with alleviated B cell activation and skew toward a Th2/Th17 response against bleomycin treatment. Importantly, in TSK/+ mice, another SSc murine model, TLR-4 abrogation attenuated hypodermal fibrosis. Conclusion: These results indicate the pivotal contribution of TLR-4 to the pathologic tissue fibrosis of SSc murine models. Our results indicate the critical role of TLR-4 signaling in the development of tissue fibrosis, suggesting that biomolecular TLR-4 targeting might be a potential therapeutic approach to SSc.
Article
Rationale: Systemic sclerosis (SSc)-associated pulmonary arterial hypertension (PAH) portends worse outcome than other forms of PAH. Vasoconstrictive and vascular remodeling actions of endothelin (ET) 1 and angiotensin (Ang) II via endothelin receptor type A (ETAR) and Ang receptor type-1 (AT1R) activation are implicated in PAH pathogenesis. Objectives: We hypothesized that stimulating autoantibodies (Abs) targeting and activating AT1R and ETAR may contribute to SSc-PAH pathogenesis, and tested their functional and biomarker relevance. Methods: Anti-AT1R and -ETAR Abs were detected by ELISA in different cohorts of patients and tested in vitro and in an animal model for their pathophysiological effects. Measurements and main results: The Abs were significantly higher and more prevalent in patients with SSc-PAH (n = 81) and connective tissue disease-associated PAH (n = 110) compared with other forms of PAH/pulmonary hypertension (n = 106). High anti-AT1R and anti-ETAR Abs predicted development of SSc-PAH and SSc-PAH-related mortality in a prospective analysis. Both Abs increased endothelial cytosolic Ca(2+) concentrations in isolated perfused rat lungs, which could be blocked by respective specific receptor antagonists. Ab-mediated stimulation of intralobar pulmonary rat artery ring segments increased vasoconstrictive responses to Ang II and ET-1, and implicated cross-talk between both pathways demonstrated by reciprocal blockade with respective antagonists. Transfer of SSc-IgG containing both autoantibodies into healthy C57BL/6J mice led to more abundant vascular and airway α-smooth muscle actin expression and inflammatory pulmonary vasculopathy. Conclusions: Anti-AT1R and -ETAR Abs are more frequent in SSc-PAH/connective tissue disease-PAH compared with other forms of pulmonary hypertension, and serve as predictive and prognostic biomarkers in SSc-PAH. Both antibodies may contribute to SSc-PAH via increased vascular endothelial reactivity and induction of pulmonary vasculopathy.