Author: Section Editor: Deputy Editor:: Pathogenesis of Systemic Sclerosis (Scleroderma)

You might also like

Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 24

Pathogenesis of systemic sclerosis (scleroderma)

Author:
Christopher P Denton, MD
Section Editor:
John S Axford, DSc, MD, FRCP, FRCPCH
Deputy Editor:
Monica Ramirez Curtis, MD, MPH

Contributor Disclosures

All topics are updated as new evidence becomes available and our peer review process is complete.
Literature review current through: Dec 2018. | This topic last updated: Jun 12, 2018.

INTRODUCTION — The pathogenesis of systemic sclerosis (SSc; scleroderma) is complex and


remains incompletely understood. Immune activation, vascular damage, and excessive synthesis
of extracellular matrix with deposition of increased amounts of structurally normal collagen are
all known to be important in the development of this illness [1,2]. These mechanisms result from
cell-cell, cell-cytokine, and cell-matrix interactions. The heterogeneity in the clinical features of
patients with SSc is most likely a reflection of the variable contributions from each of these
pathogenic factors.

Most hypotheses of the pathogenesis of SSc focus on the interplay between early immunological
events and vascular changes, which result in the generation of a population of activated fibrogenic
fibroblasts generally considered to be the effector cell in the disease (figure 1) [2-4]. There is no
doubt that vascular and immunologic processes are central to the pathogenesis of scleroderma,
although it is unclear what the initial events are and how different processes respectively trigger,
amplify, and facilitate the development of the skin- and organ-based fibrosis with vasculopathy
that is the hallmark of the disease.

Considering the clinical differences between scleroderma and other autoimmune rheumatic
diseases and the relatively modest effects that have been observed in clinical trials of
immunosuppressive agents (eg, cyclophosphamide), it is perhaps surprising that genetic and
serologic approaches to understanding scleroderma pathogenesis have highlighted the importance
of cellular and humoral immunity. Many of the genetic loci associated with scleroderma
susceptibility in large-scale genetic analyses are also associated with systemic lupus and other
autoimmune conditions [5]. Moreover, even when non-immune genes are associated in
subphenotype analyses of genetic loci, these associations are often defined by hallmark
scleroderma autoantibodies [6], again supporting the key role of the immune system in the
development and clinical features of the disease.

The multiple factors felt to be involved in the pathogenesis of SSc are reviewed here. The possible
causes of SSc, as well as the clinical manifestations, diagnosis, and treatment of this disorder, are
discussed separately. (See "Risk factors for and possible causes of systemic sclerosis
(scleroderma)" and "Overview of the clinical manifestations of systemic sclerosis (scleroderma)
in adults" and "Diagnosis and differential diagnosis of systemic sclerosis (scleroderma) in
adults" and "Overview of the treatment and prognosis of systemic sclerosis (scleroderma) in
adults".)

VASCULAR AND ENDOTHELIAL CHANGES — Vascular and endothelial cell changes,


primarily mediating vascular tone, appear to precede other features of systemic sclerosis (SSc).

Vasoconstriction — The following mediators of vascular tone may be important in the


pathogenesis of SSc [7-9]:

●Endothelins (ETs)
●Nitric oxide (NO, or endothelium-derived relaxing factor)
●Endothelium-derived constricting factors (EDCF)
●Neural, humoral, and inflammatory mediators
●Hypoxia
●Physical stress

Derangements of endothelins, NO, and superoxide anions (which are inflammatory mediators) are
felt to be the most significant mediators of altered vascular tone in SSc.

Endothelin — ET-1, a potent vasoconstrictor, is also fibrogenic and is potentially of great


importance in the pathogenesis of SSc [8]. The following observations are consistent with this
hypothesis:

●ET-1 may contribute both to vascular dysfunction and to the development of the fibrotic
lesion in SSc [10].
●The basal secretion of ET-1 from the endothelial cell may provide an important early link
between endothelial cell damage and fibroblast activation.
●Significantly elevated circulating levels of ET-1 have been found both in the minimally
fibrotic subset of SSc patients with primary vascular disease and associated pulmonary
hypertension and in the diffuse fibrotic subset in whom widespread fibrosis is the major
hallmark of the disease [11-13].

ET-1 may play an important role in the initiation of fibrosis in SSc as suggested by
immunohistochemical and autoradiographic studies. ET-1 has been found in association with the
following structures using these special techniques [14]:

●Superficial vessels in both clinically involved and uninvolved skin.


●Its putative receptor, to which it is bound, in the “yet to be involved” skin. This binding
decreased with increasing tissue fibrosis.

Nitric oxide — NO balances the vasoconstrictive action of ET-1 in normal blood vessels; a change
in the relative amounts of these two compounds is postulated to play a pathogenic role in SSc.
However, conflicting data exist concerning the role of nitric oxide. In a preliminary study, total
NO-producing compounds were decreased in the plasma of patients with SSc [15]; furthermore,
the increase in plasma NO seen after exposure of normal subjects to a cold challenge was absent
in these patients. In addition, levels of exhaled nitric oxide correlated inversely with the severity
of elevation of pulmonary artery pressure in patients with SSc lung disease [16]. In another study,
however, levels of plasma NO were significantly elevated in SSc patients compared with those in
controls [17].

NO synthase (NOS) is present in some neurons, as well as within endothelial cells. A progressive
decline in cutaneous neurons containing NOS has been noted in patients with diffuse and limited
SSc; however, it is uncertain whether this is a cause or a result of diminished tissue perfusion [18].

Superoxide anions — Superoxide anions released from the endothelium may damage the
endothelium by being able to neutralize NO and by oxidizing circulating low-density lipoproteins
(LDLs). Oxidized LDLs may be the source of the cytotoxicity of stored SSc serum on cultured
endothelial cells [19]. Studies have shown that LDLs in patients with SSc are much more
susceptible to oxidation (perhaps via free radical attack) than are those from patients with primary
Raynaud phenomenon or other rheumatic diseases [20]. In addition, increased urinary and serum
levels of a markers of oxidative free radical injury (F2-isoprostanes and 8-isoprostane,
respectively) have been found in patients with SSc when compared with healthy controls [21,22].

One adverse effect of superoxide anions, inactivation of NO, has been mentioned above. Another
is nitrosylation of proteins by peroxynitrite, a product of the reaction of NO and superoxide.
Significantly increased concentrations of nitrosylated proteins have been noted in the plasma of
patients with diffuse skin involvement but have not been noted in those with limited skin disease
or in people with primary Raynaud phenomenon [23].

Defective vasculogenesis — A deficiency of circulating endothelial cell precursors and a defect


in the ability of such cells to proliferate and mature into endothelial cells may be an important
pathogenic mechanism in SSc. Preliminary evidence suggests that there are significantly fewer
circulating endothelial cell precursors in such patients than in healthy controls or in those with
rheumatoid arthritis [24]. In addition, cells with surface markers typically associated with a
vasculogenic potential (ie, CD34+CD133+ and vascular endothelial growth factor type 2 bearing)
that are present in SSc patients less often differentiate into endothelial cells in culture than cells
with the same markers obtained from controls.

VASCULAR CYTOTOXIC FACTORS — The sera from some patients with systemic sclerosis
(SSc) have been found to be cytotoxic to endothelial cells. The following antibodies, cytokines,
proteases, and complement factors may be responsible for this activity:

●Twenty to 30 percent of patients with SSc have circulating antiendothelial cell antibodies.
These antibodies by themselves are not thought to be critical, since they are found in the sera
of patients with other rheumatic diseases. However, they are capable of upregulating the
expression of adhesion molecules on endothelial cells and (in in vitro and in vivo animal
experiments) of inducing apoptosis of these cells [25-28].
●The vascular cytotoxicity of some SSc sera is blocked by monoclonal antibodies to tumor
necrosis factor (TNF)-alpha or -beta.
●The activity of some SSc sera is blocked by prior incubation with protease inhibitors. The
source of these proteases and their nature are unknown, but retroviral proteases and the
granzyme family of proteases produced by activated T cells are thought to be possible
candidates [29]. Proteolytic activity of granzymes may also degrade angiogenic plasmin and
may increase the levels of angiostatin [30].
●Deposition of the complement membrane attack complex (C5b-C9) has been demonstrated
immunohistochemically in the microvasculature of involved skin of patients with early and
late SSc [31].

ADHESION MOLECULES — Adhesion molecules such as intercellular adhesion molecule-1


(ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and endothelial leukocyte adhesion
molecule-1 (E-selectin) are upregulated in response to cytokines and other factors following
inflammation and damage to the vascular endothelium. These endothelial adhesion molecules bind
to specific integrins on T and B cells, neutrophils, monocytes, natural killer (NK) cells, and
platelets [32]. This interaction results in adhesion and subsequent migration of these cells through
leaky endothelium and into the extracellular matrix. (See "Leukocyte-endothelial adhesion in the
pathogenesis of inflammation".)

Patients with systemic sclerosis (SSc) have both an increase in endothelial cell surface expression
of adhesion molecules and an elevation in circulating levels of their soluble forms [33], as shown
in the following studies:

●Positive immunostaining for E-selectin and ICAM-1 has been demonstrated on endothelial
cells in SSc skin but not in control skin [34-36].
●Increased circulating levels of E-selectin, VCAM-1, and ICAM-1 have also been observed
in small cross-sectional studies of patients with SSc [17,37,38].
●Elevated levels of soluble VCAM-1, E-selectin, and ICAM-1 were found in patients with
SSc renal crisis, but not in those with SSc-associated pulmonary disease [39].

It is unlikely that a single measurement will be of particular clinical use or predictive value.
However, one study of serial serum samples has indicated a correlation between changes in soluble
VCAM-1 and in soluble E-selectin and clinical deterioration or improvement [40]. Such serial
studies need to be expanded, as proven markers for disease activity are not available but are
urgently required.

Increased expression of messenger ribonucleic acid (mRNA) for some adhesion molecules in the
peripheral blood cells of patients with SSc may lead to novel diagnostic tests for SSc. Upregulated
expression of genes that encode ligands for P-selectin (SELPLG gene) and receptors for von
Willebrand factor (ITGA2B and GP1BB genes) are among a total of 382 differentially regulated
genes identified in a study that utilized gene expression profiling of unseparated peripheral blood
cells to distinguish between SSc patients and healthy controls [41].
IMMUNOLOGIC ACTIVITY — The continuing activation of endothelial cells, resulting in the
upregulation of adhesion molecules, leukocyte adhesion, and leukocyte migration out of the
vasculature, probably contributes to the pathogenesis of systemic sclerosis (SSc). It is likely that
both innate immunity and adaptive immunity are perturbed in SSc and share a role in pathogenesis.
The following studies support this conclusion:

●The expression of endothelial leukocyte adhesion molecule-1 (E-selectin) on endothelial


cells correlates with the degree of mononuclear cell infiltration in the early inflammatory
lesion of SSc [40].
●Subpopulations of lymphocytes from patients with SSc,
namely activated/cytotoxic/inducer T cells, natural killer (NK) cells, and some helper T cells,
have a markedly increased ability to adhere to endothelium [42]. The number of highly
adherent cells appears to be diminished in the peripheral blood of patients with SSc, as
suggested by an overall reduction in adhesion of peripheral mononuclear cells to endothelium
in an in vitro assay. It was postulated that this subpopulation of lymphocytes was eliminated
from the circulation through enhanced in vivo adhesion aided by chemotactic forces and by
a highly permeable endothelium.
●The mononuclear cells from patients with SSc which migrate into the extracellular matrix
express differentiation markers of activated T cells including CD3, CD4, CD45, human
leukocyte antigen (HLA)-DR, and lymphocyte function associated antigen-1 (LFA-1) [43].
These cells also have subsets of integrin molecules on their surface, including those of the
beta 1 and beta 2 class, that facilitate binding to other cells including fibroblasts and tissue
components such as type I and IV collagens, fibronectin, and laminin [44,45].
●The peripheral blood of patients with SSc disorders contains a higher proportion of such
activated CD3 and CD4 double-positive cells that migrate in vitro into an endothelial cell-
covered collagen matrix than do healthy controls (71 versus 56 percent) [46].
●There is growing appreciation that the innate immune system is likely to be involved in
pathogenesis of SSc. Genetic and functional studies have provided evidence for infiltration
of cells of the monocyte or macrophage lineages in the skin and lung of patients with SSc, as
well as evidence of altered expression and function of toll-like receptors [47,48].

Further inflammatory agents including histamines, kinins, complement, antibodies, free radicals,
thromboxanes, leukotrienes, oxidized low-density lipoproteins (LDLs), and cytolytic T cells are
all possible additional mediators of the deranged immunologic processes present in SSc.

Autoantibodies — Approximately 95 percent of patients with SSc have circulating autoantibodies


directed against one or more of a number of antigens. These include topoisomerase I (formerly
called Scl-70) (20 to 45 percent) [49], centromere antigens (12 to 44 percent) [49], fibrillarin,
ribonucleic acid (RNA) polymerase, PM-Scl, and fibrillin-1 [50], as well as RNA I, II, and III (20
percent) (table 1) [51]. (See "Diagnosis and differential diagnosis of systemic sclerosis
(scleroderma) in adults", section on 'Laboratory testing'.)

Although not very sensitive, antitopoisomerase-I (Scl-70) antibodies are highly specific for SSc
(98 to 100 percent) and correlate with a higher risk of interstitial lung disease [1]. Higher
concentrations or titers are also associated with more extensive skin involvement and with higher
disease activity [52]. Anticentromere antibodies (ACA) are associated with limited cutaneous
involvement.

The inciting stimulus for the production of these autoantibodies is unknown. One possibility is that
the targets for these autoantibodies are autoantigens that have been fragmented via reactive oxygen
species and specific metals, such as copper or iron [53]. Immunogenic peptides are subsequently
formed which are capable of breaking tolerance, inducing antibodies, and, through epitope
spreading, developing antibodies to other epitopes. In addition, the involvement of metals provided
a rationale for the use of penicillamine, a metal chelator, in the treatment of SSc.

It is also possible that, in patients with cancer-associated SSc, tumors may induce these
autoantibodies by producing somatic mutations, which then induce antibody formation against the
mutated protein. As an example, somatic mutations in the POL3RA gene, which encodes RNA
polymerase III, may induce anti-RNA polymerase III autoantibodies that may be involved in
development of SSc. (See "Overview of the clinical manifestations of systemic sclerosis
(scleroderma) in adults", section on 'Cancer risk'.)

Another possibility is that these antibodies arise in response to an infection and, via molecular
mimicry, crossreact with a native antigen. In one report, for example, antibodies directed against
cytomegalovirus and found in patients with SSc-induced apoptosis in endothelial cells via an
interaction with a cell surface protein complex [54].

Immunoglobulin G (IgG) antibodies generally mirror T-cell response to antigen, and most workers
believe it is the T cells which will be pathogenic. However, antibodies can, under different
circumstances, be directly pathogenic (as described above), can be additive to T-cell damage, or
can block T cells [1].

Autoantibodies that interact with fibroblasts could potentially play a pathogenic role in SSc. One
study of sera from 69 patients with diffuse and limited SSc, 30 patients with sarcoidosis, and 50
healthy controls found elevated levels of antifibroblast antibodies of IgG-class in 58 percent and
IgM-class in 33 percent of those with SSc, but found the same in only 1 of 30 patients with
sarcoidosis (3 percent) and in none of the healthy controls [55]. Sera-containing antifibroblast
antibodies promoted increased expression of the intercellular adhesion molecule-1 (ICAM-1) on
cultured human fibroblasts and increased both messenger RNA (mRNA) and secretion of a number
of proinflammatory cytokines.

The autoantibodies discussed above are not necessarily mutually exclusive. Indeed, a strong
correlation was noted in one study between the intensity of antifibroblast staining with
immunoglobulins from patients with SSc and reactivity of Scl-70 antibodies [56]. Further
investigation demonstrated that affinity-purified antitopoisomerase antibodies were able to bind to
the cell surface of cultured fibroblasts. Some data suggest that topoisomerase I on the fibroblast
surface mediates antifibroblast activity [57].
The presence of antibodies to receptors for platelet-derived growth factor (PGDF) in the serum of
patients with SSc may be a relatively specific finding, and profibrotic effects of agonistic antibody
binding to the fibroblast PGDF receptor suggest a potentially pathophysiological role for such
antibodies. This was illustrated in a study of 46 patients with SSc, 75 controls, and 10 patients who
underwent allogeneic bone marrow transplants and who developed graft-versus-host disease
(GVHD) with skin involvement [58]. A biologic assay that assessed the stimulatory effect of
immunoglobulin G (IgG) was increased (>99 percentile for healthy persons) in all of those with
SSc but in none of those with systemic lupus erythematosus, rheumatoid arthritis, primary
Raynaud phenomenon, or interstitial lung disease. The stimulatory activity was present in Ig
obtained from the 10 patients with GVHD. However, these results have not been confirmed in
three other studies [59].

In one study, the presence of agonist antibodies to vascular receptors for endothelin (ET) and
angiotensin was associated with poor long-term survival, although the contribution to pathogenesis
remains unclear [60].

Other autoantibodies that have been observed include antinucleolar, anti-Th/To, anti-Ku,
antiphospholipid, anti-Sm, anti-RNP, anti-Ro, antineutrophil cytoplasmic antibody (ANCA), anti-
U1 RNA, antinucleosome, anti-fibrillin-1 (FBN1), anti-matrix metalloproteinases 1 and 3, and
anti-endothelial cell specificities [25,61-64].

Graft-versus-host disease or microchimerism — Some investigators have postulated that SSc


is a variant of GVHD, since both disorders share clinical features such as prominent involvement
of the skin, lung, and esophagus. An animal model of GVHD replicates many of the features of
human disease [65,66]. Indirect support for this hypothesis is provided both by an increased
frequency of persistent fetal cells among women with SSc and a history of pregnancy (as compared
with normal women with a history of pregnancy) and by the presence of fetal cells in the involved
skin of such patients [67-70]. Cells derived from the fetus may also be found in other tissues of
women with SSc, most often in the spleen [71]. These associations, however, do not prove
causality [72].

GROWTH FACTORS AND CYTOKINES — The cell-cell and cell-matrix interactions


mentioned above may stimulate the production and release of growth factors and cytokines able
to mediate the proliferation and activation of vascular and connective tissue cells, particularly
fibroblasts. The importance of cytokines in the pathogenesis of systemic sclerosis (SSc) has been
strengthened by the realization that the endothelial cell and the fibroblast are both capable of
producing factors essential for the development of this illness (table 2).

It had been previously assumed that the major sources of paracrine factors in SSc were
inflammatory cells and lymphocytes. However, fibroblasts and endothelial cells can also produce
and release cytokines, and they can mobilize growth factors, such as basic fibroblast growth factor,
from the extracellular matrix [73]. These observations raise the possibility of reciprocal
interactions between all of these cell types, which may be central to disease pathogenesis.
Autocrine or paracrine loops of cytokines may also be responsible for the
generation and/or persistence of the SSc phenotype seen after several cell passages in conventional
tissue culture [74,75]. A number of possible interactions involving various mediators and several
cell types may occur (figure 2). Different loops may also operate within clinical subsets and may
be variously expressed at different stages of the disease.

Potential pathogenic cytokines — Large numbers of cytokines have been examined as potential
effectors of fibrosis in SSc (table 3). Included in this group are:

●Transforming growth factor-beta (TGF-beta)


●Platelet-derived growth factor (PDGF)
●Interleukin (IL)-1, IL-4, IL-6, IL-8, and IL-17
●Connective tissue growth factor
●Insulin-like growth factors (IGF)
●Basic fibroblast growth factor (bFGF)
●Interferon-gamma (IFN-gamma)
●CXCL4
●Monocyte chemotactic protein-1 and -3
●Tumor necrosis factor (TNF)

It is highly unlikely that any one mediator can account for the pathogenesis of SSc, since cytokines
are produced by several cell types that may interact through autocrine and paracrine pathways. The
following sections address the biological properties of those growth factors and cytokines felt to
be most likely to contribute to the development of SSc. (See "Role of cytokines in rheumatic
diseases".)

Effects on cell-cell interactions — Cytokines can modulate cell-cell interactions in addition to


the direct cellular effects previously mentioned. As an example, IL-1 and TNF-alpha both enhance
leukocyte adhesions to endothelial cells, their subsequent extravasation into the extracellular
matrix, and the number of activated endothelial cells in SSc lesions.

Effects on adhesion molecules — Cytokines may directly upregulate key adhesion molecules,
including intracellular adhesion molecule (ICAM)-1. As an example, fibroblasts from patients with
SSc bind to lymphocytes in vitro, through interactions between ICAM-1 and lymphocyte function-
associated antigen (LFA)-1 [76]. In addition, IL-1, TNF, and IFN-gamma have been shown to
modulate the expression of ICAM-1 by SSc fibroblasts, an effect enhanced by the female sex
hormone B-estradiol. This may be an important observation in view of the female predominance
of this disease [77].

Effects on the SSc fibroblast phenotype — Many studies have focused on the possible paracrine
factors that may initiate or maintain the typical SSc fibroblast phenotype. Early investigations
demonstrated the inherent ability of SSc cells to grow in low serum environments and their reduced
responsiveness to exogenous PDGF. These findings were explained by the observation that SSc
fibroblasts secrete PDGF, thereby rendering them less sensitive to exogenous growth factors [78].
Subsequent work has shown that there is considerable enhancement of the fibroblasts’ mitogenic
response to other mediators, including TGF-beta, IL-6, and endothelin (ET)-1.

Transforming growth factor-beta — The marked matrix stimulatory properties of the


transforming growth factors, particularly TGF-beta, have implicated these proteins as potentially
important mediators in SSc. However, studies of skin biopsies, bronchoalveolar lavage (BAL),
and blood samples from patients with SSc have produced conflicting results on the levels of TGF-
beta messenger ribonucleic acid (mRNA) and protein, despite the fact that TGF-beta can be readily
demonstrated in both involved skin and the fibrotic lung [79-81]. These unexpected results may
be explained by the different clinical characteristics of the SSc patients enrolled in the two studies.
These patients may have been representative of unique SSc subsets, may have been studied at
different stages of their disease process, or may have had variable rates of disease progression.
These variables make exact comparison of results from different studies exceedingly difficult.

Even if TGF-beta is an important mediator in the pathogenesis of SSc, it is likely to operate in


concert with other cytokines. Moreover, different downstream-signaling pathways may be invoked
for short- or long-term (chronic) TGF-beta-mediated effects. A central role for the Smad family of
proteins is postulated. Smads are evolutionarily conserved proteins that mediate transcriptional
activation by members of the TGF-beta superfamily of cytokines. Some (eg, Smad3) activate
intracellular signaling, whereas others (eg, Smad7) are inhibitory.

The role of TGF-beta in the acquisition and maintenance of the SSc phenotype is also unclear.
Attempts to induce long-term enhancement of collagen-1 production by fibroblasts via regular
pulsed exposure to TGF-beta have failed, as have attempts to demonstrate coordinated regulation
of collagen-1 and TGF-beta by SSc fibroblasts [82-84]. Furthermore, collagen and TGF-beta type
II receptor mRNA appear to be inducible by TGF-beta in both SSc cells and control cells,
indicating that the lack of a sustained elevation in collagen synthesis is not due to lack of
responsiveness by the fibroblasts; it may, however, reflect the transient nature of TGF-beta-
induced fibrogenesis [85].

The following evidence suggests that TGF-beta may indirectly influence other cytokines, primarily
PDGF, to promote fibrogenesis:

●TGF-beta is an indirect mitogen for fibroblasts, acting via interaction with the PDGF-alpha
receptor and inducing expression of other mitogenic growth factors including connective
tissue growth factor (CTGF, also termed CCN-2).
●Fibroblasts from patients with SSc, in contrast to normal adult newborn foreskin fibroblasts,
respond to TGF-beta-1 by expressing greater numbers of PDGF-alpha receptors on their
surface, increasing PDGF-alpha receptor protein, and elevating mRNA levels [86].
●TGF-beta increases the mitogenic responses to platelet-derived growth factor (PDGF)-AA.
PDGF-AA binds only to the PDGF-alpha receptor, in contrast to PDGF-beta, which binds to
both the alpha and beta receptors of SSc cells. PDGF-AA has been found
immunohistochemically in SSc dermis near blood vessels and hair follicles but not in normal
skin.
TGF-beta receptors — The possibility that receptors for TGF-beta on fibroblasts have a role in
SSc pathogenesis has also been explored. Expression of type I and type II TGF-beta receptors were
increased more than twofold in fibroblasts cultivated from the skin of patients with diffuse SSc
when compared with control fibroblasts [87]. Production of collagen mRNA by cultivated
fibroblasts could be decreased by anti-TGF-beta antibodies and by transfection with TGF-beta
receptor antisense oligonucleotides.

Upregulation of type I and type II TGF-beta receptors also occurs in fibroblasts obtained from
lesional skin of patients with localized forms of SSc, such as morphea and linear SSc [88]. The
stimulus and mechanism for the increased expression of these receptors are unknown.

Decreased turnover of TGF-beta receptors and a failure of control over receptor signaling could
also result in an increased sensitivity to TGF effects. This mechanism was suggested by the
finding, in cultured skin fibroblasts of patients with diffuse cutaneous SSc, of impaired ubiquitin-
mediated degradation of TGF-beta receptors; and by an absence of negative feedback via
inhibitory molecules (Smad7) [89]. Defective induction of Smad7 signaling may also contribute
to altered TGF-beta responsiveness in SSc [90].

Increased expression of endoglin, a non-activating TGF-beta receptor, as indicated by modest


elevation in fibroblast endoglin mRNA and membrane-bound protein, occurs in sclerodermatous
skin, and there is preliminary evidence that these levels increase with the duration of disease. [91].
It is unclear what role, if any, endoglin may play in SSc. Endoglin is important in angiogenesis,
and mutations in the endoglin gene are responsible for some forms of hereditary hemorrhagic
telangiectasia. (See "Clinical manifestations and diagnosis of hereditary hemorrhagic
telangiectasia (Osler-Weber-Rendu syndrome)", section on 'Pathophysiology'.)

Platelet-derived growth factor — These results suggest that TGF-beta may increase the
expression of fibroblast populations in SSc by activating the PDGF-AA ligand or PDGF-alpha
receptor pathway. PDGF, in turn, has been shown to increase the rate of transcription of bFGF in
normal fibroblasts. Both PDGF and bFGF are deposited around blood vessels in the lower dermis
of early lesions, suggesting that these growth factors may be related to endothelial injury in SSc.

PDGF also regulates the expression of genes that are important in the mitogenic response. As an
example, an increase in ras oncogene product in early lesional skin was noted in association with
vascular endothelium and mononuclear cells [92]. Other studies have established that fibroblasts
expressing the ras oncogene produce more bFGF, suggesting that bFGF may also play an autocrine
role leading to growth stimulation.

Connective tissue growth factor — There is growing evidence for upregulation of the TGF-beta-
induced matricellular protein known as CTGF in various forms of fibrosis [93]. Increased
production by SSc fibroblasts has been consistently found. Mean plasma levels of an N-terminal
fragment of CTGF are markedly elevated in patients with diffuse skin involvement and are
moderately increased in those with limited cutaneous disease when compared with healthy controls
[94].
Initially, it was suggested that CTGF may be important in sustaining a profibrotic fibroblast
phenotype as a secondary cytokine induced by TGF-beta. However, a body of evidence suggests
a more complex activity as a matricellular protein exerting some direct effects on fibroblasts by
regulating activity or bioavailability of TGF-beta [95] or effects on cell adhesion and fibroblast-
matrix interaction [96].

Interleukin-6 and related cytokines — There has been an increased focus on IL-6 as a mediator
in SSc based upon the promising early clinical data for use of anti-IL-6 antibody in diffuse SSc
[97] and emerging clinical data suggesting that higher levels of serum IL-6 may associated with
more severe skin disease [98] or greater progression of lung fibrosis [99]. SSc cells appear to
produce much more IL-6 than controls [100], and IL-6 may represent one of the soluble mitogenic
factors transferable in culture medium derived from such cells [101]. The role of IL-6 is unclear,
but it may modulate activity of other key profibrotic pathways in fibroblasts as well as its effect
on immune cells. This may be especially relevant to polarization of lymphocytes, favoring Th17
differentiation or macrophages. Interestingly, attenuation of a fibrogenic M2 macrophage
signature in skin was associated with possible clinical response to blocking IL-6 in a clinical trial
[97]. Other members of the IL-6 family have also been suggested as possible mediators in SSc
pathogenesis, including IL-31, oncostatin M, and IL-11 [102].

Interleukin-1 — Interest has also been directed to the production and regulation of the cytokines
IL-1 by SSc fibroblasts. IL-1a is also produced excessively by SSc fibroblast and may contribute,
via autocrine or paracrine pathways, to their abnormal properties in culture [103,104]. Some
evidence suggests that keratinocyte-derived IL-1a is important in regulating fibroblast properties
in SSc [105].

Interleukin-4 — IL-4 may have a profibrotic effect on scleroderma dermal fibroblasts. These cells
have IL-4 receptors, and, when cultured in IL-4 containing media, the production of collagen is
significantly increased [106].

Chemokines — A number of chemokines have been reported to be elevated in the serum and
biopsies of SSc skin. CXCL4, also known as platelet-activating factor 4, belongs to the CXC
family of chemokines. CXCL4 is a protein that is a potent antiangiogenic chemokine that
influences angiogenesis through an integrin-dependent mechanism [107]. It also inhibits the
expression of the antifibrotic cytokine IFN-gamma, and upregulates profibrotic cytokines IL-4 and
IL-13 [108].

Further, CXCL4 has been implicated in the pathogenesis of SSc based on findings that plasma
levels are elevated in patients with SSc, and high plasma levels correlate with progression of both
skin and pulmonary fibrosis, as well as pulmonary arterial hypertension [109]. This was
demonstrated in a proteome-wide analysis in which CXCL4 levels were measured in patients with
SSc and compared with controls, including patients with rheumatoid arthritis, ankylosing
spondylitis, and systemic lupus erythematosus. CXCL4 was the predominant protein secreted by
plasmacytoid dendritic cells in patients with SSc, both in the circulation and the skin. CXCL4 was
also the only chemokine that correlated with faster progression of skin and lung fibrosis. Although
interesting, these observations require further confirmation to assess whether targeting this
pathway may be helpful in managing the disorder.

The role of plasmacytoid dendritic cells has been an area of interest in the pathogenesis of SSc as
they are the major source of type I interferon, which has been implicated in the pathogenesis of
several autoimmune diseases [110-113]. However, this may suggest that altered function of these
cells is a generic feature of autoimmunity, and further studies are needed to delineate scleroderma-
specific importance.

It has been suggested that CC motif chemokine receptor 2 (CCR2) overexpression of monocyte
chemotactic protein (MCP)-1 (CC motif ligand 2 [CCL2]) associates with early-stage disease and
may promote fibrosis [114]. Interactions with other cytokines including IL-4 have also been
demonstrated [106].

mRNA for MCP-3, a member of the CC chemokine family, is overexpressed by dermal fibroblasts
cultured from tight-skin mice, a model of SSc [115]. Immunostaining for this protein in the skin
of patients and healthy controls demonstrated more MCP-3 in the lesional skin of those with early
onset diffuse disease than in those with limited cutaneous involvement or in healthy controls. This
chemokine could contribute to fibrosis through effects on extracellular matrix production as a
chemoattractant for leukocytes. Chemokine expression in lung tissue may be relevant to the
recruitment of fibroblast precursor cells and to the development of lung fibrosis in SSc [116-118].

Summary of the effects of growth factors and cytokines — It seems likely that the interplay
between a number of cytokines and growth factors will be found to underlie the pathogenesis of
SSc. Increased levels of circulating IL-1, IL-2, IL-2R, IL-4, IL-8, IL-17, TNF-alpha, interferon,
and antibodies to IL-6 and IL-8 have been found in patients with SSc [119-122]. However, the
presence of high levels of a particular cytokine does not necessarily imply a causative role. In
addition, it has become clear that the interpretation of these studies is dependent upon the assay
used (bioassay or immunoassay) and upon a realization that many factors interfere with each
system. Despite these reservations, it may be inferred that the increased levels of the various
cytokines support a cellular immune mechanism in SSc and an ongoing expansion of T cells and
of their secreted products.

It is important to remember that, because cytokines influence each other, the pattern of total
cytokine production may be more important than any individual cytokine. The soluble cytokines
are not exclusive to SSc, being found in many other diseases. However, their presence, pattern of
expression, and known interactions may provide clues to the correct cell-type and to relevant
tissues involved in disease development and may aid in the development of directed therapy.

FIBROBLAST ACTIVITY AND COLLAGEN SYNTHESIS — The development of the


fibroblast capable of excess matrix production and deposition is the hallmark of systemic sclerosis
(SSc) (table 4). Fibroblast cultures from both the papillary and the reticular dermis of patients with
SSc have been found to produce increased amounts of collagen and other extracellular matrix
components; this has been demonstrated in vivo, in biopsy specimens, and in vitro in fibroblast
cultures derived from lesional SSc skin. These abnormalities persist through many passages in
tissue culture. Normal dermal fibroblasts, in contrast, are relatively quiescent, showing little
tendency to proliferate or to elaborate extracellular matrix in the absence of positive stimuli.

Abnormal fibroblast activity in SSc can also be identified in organs other than skin. As an example,
increased collagen synthesis by fibroblasts isolated from lung biopsy specimens of patients with
SSc lung disease has been demonstrated, thereby suggesting a similar pathogenic mechanism in
lung and skin [123].

Increased collagen production is not a feature of all SSc skin fibroblast strains or of all fibroblasts
within a culture population; it may, therefore, be limited to discrete subpopulations [124]. Indeed,
differences between normal and SSc populations are often more apparent when individual
subpopulations are studied rather than mass populations. In situ hybridization studies have
demonstrated that high collagen-producing subpopulations of fibroblasts are frequently observed
in close proximity to mononuclear cells and adjacent to blood vessels in patients with SSc
[125,126].

Activating and inhibitory signals — Increased matrix production by fibroblasts may result from
a net excess of positive activating signals and/or from a reduction in inhibitory signals. Positive
paracrine mediators may be derived from immune cells, from endothelial cells, or from the
fibroblasts themselves. In turn, inhibitory influences may arise from interactions with noncellular
extracellular matrix components such as collagen fibers and fibronectin. Such interactions appear
to be potent down-regulators of collagen synthesis by normal dermal fibroblasts and also modulate
cell morphology, proliferative capacity, and cytokine responsiveness. It has been suggested that
down-regulation of nuclear hormone receptors, including peroxisome proliferator-activated
receptor gamma [127] and others, may provide a mechanism by which there is a failure of
appropriate feedback inhibitory signals in activated fibroblasts in SSc [128].

The SSc fibroblast — One potential mechanism for the development of the activated fibroblast
places the initiating event in the vascular bed, leading to growth factor and cytokine production
with resulting fibroblast activation and with subsequent fibrosis. Other studies have added
complexity by demonstrating that fibroblasts involved in tissue repair may be derived from diverse
origins, including bone marrow-derived progenitor cells, local epithelial cells, or resident
progenitor or stem cell populations; however, a unifying mechanism may be the dependence of
these cell types on transforming growth factor-beta (TGF-beta) and its regulated pathways and
mediators [129]. Thus, deletion of TGF-beta responses by genetic modification in animal models
appears to attenuate fibrosis and scar formation [130,131]. This may have implications for novel
treatment strategies [132]. While this unifies several of the different pathogenic events seen in SSc,
it cannot completely explain the abnormal phenotype of the SSc fibroblast as indicated by the
following observations:

●Although dermal mononuclear cell infiltrates are common in vivo, they are not universal,
and in the later stages of the disease there is ongoing dermal fibrosis in the absence of apparent
immune cell activity. Furthermore, SSc fibroblasts in vitro are often less dependent upon
serum supplementation and other growth promoting factors than normal, healthy fibroblast
cultures [78].
●In another study, the collagen-secreting ability of SSc fibroblasts continued at the same rate
whatever their feeding conditions, unlike control fibroblasts which increased their collagen
output briskly in response to frequent serum replenishment [133].

It therefore appears that immune cell or other influences may lead to the development of a
relatively autonomous activated fibroblast strain. Several of the following possibilities may
explain such behavior:

●Soluble fibroblast products, which maintain the SSc phenotype, may have been induced,
allowing an autocrine feedback loop to become established. However, no such mechanism
has been conclusively shown, although several cytokines have been considered, particularly
interleukin (IL)-1, IL-6, platelet-derived growth factor (PDGF), and TGF-beta.
●A second possibility is the induction of a permanent dysfunction of some intracellular
regulatory gene(s), such as a proto-oncogene or its product, which then acts to increase matrix
synthesis. As an example, cultured fibroblasts from patients with SSc have more
immunoreactive protein kinase C-delta (PKC-delta) than those of healthy controls; increased
transcription of type I collagen genes in cells derived from these patients can be abrogated in
vitro by a selective inhibitor of PKC-delta, rottlerin [134].
●A third potential mechanism is an immune cell-mediated selection of a subpopulation of
fibroblasts already committed to high levels of collagen production or preferential
proliferation. The clonal expansion of such fibroblast subsets under immune, hypoxic, or
other influences may perpetuate the fibrotic processes in vivo as well as the experimental
situation in vitro.
The metabolic heterogeneity of fibroblasts is well-established, and analysis of fibroblast
clonal populations shows marked heterogeneity of cell surface markers, proliferation,
collagen production, collagenase synthesis, and prostaglandin E2 biosynthetic responses to
IL-2 or other mediators [124]. Several studies on clonal selection in SSc fibroblasts have been
performed with varying results.
●A fourth possibility is that the interaction of fibroblasts with the extracellular matrix alters
cellular function, including the control of matrix deposition and the response to cytokines.
The question has been raised as to whether SSc fibroblasts may differ in their adhesion to
matrix. In general, interactions of cells with matrix proteins involve specific cell membrane
proteins that belong to different families of receptors such as integrins, cell surface
proteoglycans, and anchorins. This is a very complex process involving different domains
(eg, RGD [arginine-glycine-aspartate] sequences) and various receptor molecules which then
mediate different cellular signals that are transmitted to the nucleus. It has been suggested
that stiffness of extracellular matrix may be important in generation or persistence of
myofibroblasts phenotype. This is based on observations that suggest that mechanical
stiffness of fibrotic skin or lung may promote further fibroblast activation or myofibroblasts
persistence [135]. It has been suggested that altered matrix proteins may also contribute to
innate immune activation relevant to persistent fibrosis [136].
Microfibrils of fibrillin-1 produced by fibroblasts from uninvolved skin of patients with SSc
may be less stable than fibrils produced by similar cells of those without disease [137]. The
mechanism for the observed instability is uncertain. Findings that suggest that the fibrillar
instability may be pathogenic include the association between SSc and genetic markers near
the fibrillin-1 gene among Choctaw Indians [138], as well as a tandem repeat in the fibrillin
gene of the tight skin mouse [139]. (See "Risk factors for and possible causes of systemic
sclerosis (scleroderma)".)
●A fifth possibility, for which there is some support, is that a stable epigenetic change leads
to a profibrotic phenotype of the fibroblast. Hypermethylation of deoxyribonucleic acid
(DNA) and deacetylation of histone proteins, changes that decrease the transcriptional
activity in the region of a promoter of a gene (FLI1 gene), have been noted in cultured
fibroblasts from patients, and hypermethylation of the promoter was present in samples of
skin from patients but not in three control cell lines [140].

Models of extracellular matrix — Another area of investigation uses models to simulate the
extracellular milieu seen by SSc fibroblasts. The extracellular matrix may be simulated by the use
of three-dimensional collagen gels. When normal fibroblasts are seeded in such gels, they contract
the gels, change their morphology, and modulate several of their specific functions, including
protein synthesis and a marked reduction in collagen-specific mRNA levels. When SSc cells are
seeded in a similar fashion, they contract the gel but do not downregulate collagen synthesis
compared with normal cells.

This impairment in downregulation may reflect one of the following [141]:

●Changes in the a1b1 integrin expression on the fibroblast cell surface


●Altered intracellular signaling secondary to integrin-ligand binding
●Altered regulation of collagen gene transcription in these cells

It has also been suggested that altered stability of the collagen mRNA transcript may be involved
in determining steady-state mRNA levels in three-dimensional collagen gel versus monolayer
fibroblast cultures [142].

Other relevant areas of investigation include studies of the complex regulation of extracellular
matrix degradation. Early reports suggested that matrix degradative processes were not altered in
SSc; however, several studies suggest that this may not be the case [143,144]. As an example, one
product of matrix degradation, endostatin (a fragment of type XVIII collagen and an inhibitor of
angiogenesis), was increased significantly in patients with SS compared with healthy controls
(53.2 versus 9.9 ng/mL) [145].

SUMMARY — The pathogenesis of systemic sclerosis (SSc) is better defined at a molecular level
and involves multiple cell types from the innate or adaptive immune system, vasculature, and
connective tissue. Fibrogenic fibroblasts ultimately mediate the fibrotic pathology of SSc, but the
factors determining inappropriate activation and the relative importance of enhanced activating
influences or reduced down-regulatory signals are not well understood.
Treatment for almost all aspects of SSc remains inadequate. Elucidation of the precise interplay
between the immunologic, vascular, and fibrotic components of SSc is likely to be an important
first step toward more effective therapy. As key growth factors or chemokines are identified, there
may be new opportunities for a more targeted approach to therapy. The ultimate goal of targeted
subset and stage-specific therapy will depend upon progress in the understanding of the complex
pathogenesis of this disease.

Use of UpToDate is subject to the Subscription and License Agreement.


REFERENCES

1. Black CM. The aetiopathogenesis of systemic sclerosis: thick skin--thin hypotheses. The Parkes
Weber Lecture 1994. J R Coll Physicians Lond 1995; 29:119.
2. Systemic sclerosis: current pathogenetic concepts and future prospects for targeted therapy. Lancet
1996; 347:1453.
3. Piela-Smith TH, Korn JH. Lymphocyte modulation of fibroblast function in systemic sclerosis.
Clin Dermatol 1994; 12:369.
4. Sollberg S, Mauch C, Eckes B, Krieg T. The fibroblast in systemic sclerosis. Clin Dermatol 1994;
12:379.
5. Gorlova O, Martin JE, Rueda B, et al. Identification of novel genetic markers associated with
clinical phenotypes of systemic sclerosis through a genome-wide association strategy. PLoS Genet
2011; 7:e1002178.
6. Radstake TR, Gorlova O, Rueda B, et al. Genome-wide association study of systemic sclerosis
identifies CD247 as a new susceptibility locus. Nat Genet 2010; 42:426.
7. Pearson JD. The endothelium: its role in scleroderma. Ann Rheum Dis 1991; 50 Suppl 4:866.
8. Levin ER. Endothelins. N Engl J Med 1995; 333:356.
9. Flavahan NA, Flavahan S, Liu Q, et al. Increased alpha2-adrenergic constriction of isolated
arterioles in diffuse scleroderma. Arthritis Rheum 2000; 43:1886.
10. Kahaleh MB. Endothelin, an endothelial-dependent vasoconstrictor in scleroderma. Enhanced
production and profibrotic action. Arthritis Rheum 1991; 34:978.
11. Zamora MR, O'Brien RF, Rutherford RB, Weil JV. Serum endothelin-1 concentrations and cold
provocation in primary Raynaud's phenomenon. Lancet 1990; 336:1144.
12. Yamane K, Miyauchi T, Suzuki N, et al. Significance of plasma endothelin-1 levels in patients
with systemic sclerosis. J Rheumatol 1992; 19:1566.
13. Vancheeswaran R, Magoulas T, Efrat G, et al. Circulating endothelin-1 levels in systemic sclerosis
subsets--a marker of fibrosis or vascular dysfunction? J Rheumatol 1994; 21:1838.
14. Vancheeswaran R, Azam A, Black C, Dashwood MR. Localization of endothelin-1 and its binding
sites in scleroderma skin. J Rheumatol 1994; 21:1268.
15. Kahaleh MB, LeRoy EC. Autoimmunity and vascular involvement in systemic sclerosis (SSc).
Autoimmunity 1999; 31:195.
16. Rolla G, Colagrande P, Scappaticci E, et al. Exhaled nitric oxide in systemic sclerosis:
relationships with lung involvement and pulmonary hypertension. J Rheumatol 2000; 27:1693.
17. Andersen GN, Caidahl K, Kazzam E, et al. Correlation between increased nitric oxide production
and markers of endothelial activation in systemic sclerosis: findings with the soluble adhesion
molecules E-selectin, intercellular adhesion molecule 1, and vascular cell adhesion molecule 1.
Arthritis Rheum 2000; 43:1085.
18. Ibba-Manneschi L, Niissalo S, Milia AF, et al. Variations of neuronal nitric oxide synthase in
systemic sclerosis skin. Arthritis Rheum 2006; 54:202.
19. Blake DR, Winyard P, Scott DG, et al. Endothelial cell cytotoxicity in inflammatory vascular
diseases--the possible role of oxidised lipoproteins. Ann Rheum Dis 1985; 44:176.
20. Bruckdorfer KR, Hillary JB, Bunce T, et al. Increased susceptibility to oxidation of low-density
lipoproteins isolated from patients with systemic sclerosis. Arthritis Rheum 1995; 38:1060.
21. Stein CM, Tanner SB, Awad JA, et al. Evidence of free radical-mediated injury (isoprostane
overproduction) in scleroderma. Arthritis Rheum 1996; 39:1146.
22. Ogawa F, Shimizu K, Muroi E, et al. Serum levels of 8-isoprostane, a marker of oxidative stress,
are elevated in patients with systemic sclerosis. Rheumatology (Oxford) 2006; 45:815.
23. Dooley A, Gao B, Bradley N, et al. Abnormal nitric oxide metabolism in systemic sclerosis:
increased levels of nitrated proteins and asymmetric dimethylarginine. Rheumatology (Oxford)
2006; 45:676.
24. Kuwana M, Okazaki Y, Yasuoka H, et al. Defective vasculogenesis in systemic sclerosis. Lancet
2004; 364:603.
25. Carvalho D, Savage CO, Black CM, Pearson JD. IgG antiendothelial cell autoantibodies from
scleroderma patients induce leukocyte adhesion to human vascular endothelial cells in vitro.
Induction of adhesion molecule expression and involvement of endothelium-derived cytokines. J
Clin Invest 1996; 97:111.
26. Sgonc R, Gruschwitz MS, Boeck G, et al. Endothelial cell apoptosis in systemic sclerosis is
induced by antibody-dependent cell-mediated cytotoxicity via CD95. Arthritis Rheum 2000;
43:2550.
27. Worda M, Sgonc R, Dietrich H, et al. In vivo analysis of the apoptosis-inducing effect of anti-
endothelial cell antibodies in systemic sclerosis by the chorionallantoic membrane assay. Arthritis
Rheum 2003; 48:2605.
28. Ahmed SS, Tan FK, Arnett FC, et al. Induction of apoptosis and fibrillin 1 expression in human
dermal endothelial cells by scleroderma sera containing anti-endothelial cell antibodies. Arthritis
Rheum 2006; 54:2250.
29. Kahaleh MB, Fan PS. Mechanism of serum-mediated endothelial injury in scleroderma:
identification of a granular enzyme in scleroderma skin and sera. Clin Immunol Immunopathol
1997; 83:32.
30. Mulligan-Kehoe MJ, Drinane MC, Mollmark J, et al. Antiangiogenic plasma activity in patients
with systemic sclerosis. Arthritis Rheum 2007; 56:3448.
31. Sprott H, Müller-Ladner U, Distler O, et al. Detection of activated complement complex C5b-9
and complement receptor C5a in skin biopsies of patients with systemic sclerosis (scleroderma). J
Rheumatol 2000; 27:402.
32. Springer TA. Adhesion receptors of the immune system. Nature 1990; 346:425.
33. Sato S. Abnormalities of adhesion molecules and chemokines in scleroderma. Curr Opin
Rheumatol 1999; 11:503.
34. Claman HN, Giorno RC, Seibold JR. Endothelial and fibroblastic activation in scleroderma. The
myth of the "uninvolved skin". Arthritis Rheum 1991; 34:1495.
35. Sollberg S, Peltonen J, Uitto J, Jimenez SA. Elevated expression of beta 1 and beta 2 integrins,
intercellular adhesion molecule 1, and endothelial leukocyte adhesion molecule 1 in the skin of
patients with systemic sclerosis of recent onset. Arthritis Rheum 1992; 35:290.
36. Gruschwitz M, von den Driesch P, Kellner I, et al. Expression of adhesion proteins involved in
cell-cell and cell-matrix interactions in the skin of patients with progressive systemic sclerosis. J
Am Acad Dermatol 1992; 27:169.
37. Sfikakis PP, Tesar J, Baraf H, et al. Circulating intercellular adhesion molecule-1 in patients with
systemic sclerosis. Clin Immunol Immunopathol 1993; 68:88.
38. Carson CW, Beall LD, Hunder GG, et al. Serum ELAM-1 is increased in vasculitis, scleroderma,
and systemic lupus erythematosus. J Rheumatol 1993; 20:809.
39. Stratton RJ, Coghlan JG, Pearson JD, et al. Different patterns of endothelial cell activation in renal
and pulmonary vascular disease in scleroderma. QJM 1998; 91:561.
40. Denton CP, Bickerstaff MC, Shiwen X, et al. Serial circulating adhesion molecule levels reflect
disease severity in systemic sclerosis. Br J Rheumatol 1995; 34:1048.
41. Tan FK, Zhou X, Mayes MD, et al. Signatures of differentially regulated interferon gene
expression and vasculotrophism in the peripheral blood cells of systemic sclerosis patients.
Rheumatology (Oxford) 2006; 45:694.
42. Rudnicka L, Majewski S, Blaszczyk M, et al. Adhesion of peripheral blood mononuclear cells to
vascular endothelium in patients with systemic sclerosis (scleroderma). Arthritis Rheum 1992;
35:771.
43. Prescott RJ, Freemont AJ, Jones CJ, et al. Sequential dermal microvascular and perivascular
changes in the development of scleroderma. J Pathol 1992; 166:255.
44. Hynes RO. Integrins: versatility, modulation, and signaling in cell adhesion. Cell 1992; 69:11.
45. Gullberg D, Tingström A, Thuresson AC, et al. Beta 1 integrin-mediated collagen gel contraction
is stimulated by PDGF. Exp Cell Res 1990; 186:264.
46. Stummvoll GH, Aringer M, Grisar J, et al. Increased transendothelial migration of scleroderma
lymphocytes. Ann Rheum Dis 2004; 63:569.
47. York MR. Novel insights on the role of the innate immune system in systemic sclerosis. Expert
Rev Clin Immunol 2011; 7:481.
48. Bhattacharyya S, Wei J, Varga J. Understanding fibrosis in systemic sclerosis: shifting paradigms,
emerging opportunities. Nat Rev Rheumatol 2011; 8:42.
49. Reveille JD, Solomon DH, American College of Rheumatology Ad Hoc Committee of
Immunologic Testing Guidelines. Evidence-based guidelines for the use of immunologic tests:
anticentromere, Scl-70, and nucleolar antibodies. Arthritis Rheum 2003; 49:399.
50. Tan FK, Arnett FC, Antohi S, et al. Autoantibodies to the extracellular matrix microfibrillar
protein, fibrillin-1, in patients with scleroderma and other connective tissue diseases. J Immunol
1999; 163:1066.
51. Bunn CC, Black CM. Systemic sclerosis: an autoantibody mosaic. Clin Exp Immunol 1999;
117:207.
52. Hu PQ, Fertig N, Medsger TA Jr, Wright TM. Correlation of serum anti-DNA topoisomerase I
antibody levels with disease severity and activity in systemic sclerosis. Arthritis Rheum 2003;
48:1363.
53. Casciola-Rosen L, Wigley F, Rosen A. Scleroderma autoantigens are uniquely fragmented by
metal-catalyzed oxidation reactions: implications for pathogenesis. J Exp Med 1997; 185:71.
54. Lunardi C, Bason C, Navone R, et al. Systemic sclerosis immunoglobulin G autoantibodies bind
the human cytomegalovirus late protein UL94 and induce apoptosis in human endothelial cells.
Nat Med 2000; 6:1183.
55. Chizzolini C, Raschi E, Rezzonico R, et al. Autoantibodies to fibroblasts induce a proadhesive and
proinflammatory fibroblast phenotype in patients with systemic sclerosis. Arthritis Rheum 2002;
46:1602.
56. Hénault J, Tremblay M, Clément I, et al. Direct binding of anti-DNA topoisomerase I
autoantibodies to the cell surface of fibroblasts in patients with systemic sclerosis. Arthritis Rheum
2004; 50:3265.
57. Hénault J, Robitaille G, Senécal JL, Raymond Y. DNA topoisomerase I binding to fibroblasts
induces monocyte adhesion and activation in the presence of anti-topoisomerase I autoantibodies
from systemic sclerosis patients. Arthritis Rheum 2006; 54:963.
58. Baroni SS, Santillo M, Bevilacqua F, et al. Stimulatory autoantibodies to the PDGF receptor in
systemic sclerosis. N Engl J Med 2006; 354:2667.
59. Dragun D, Distler JH, Riemekasten G, Distler O. Stimulatory autoantibodies to platelet-derived
growth factor receptors in systemic sclerosis: what functional autoimmunity could learn from
receptor biology. Arthritis Rheum 2009; 60:907.
60. Riemekasten G, Philippe A, Näther M, et al. Involvement of functional autoantibodies against
vascular receptors in systemic sclerosis. Ann Rheum Dis 2011; 70:530.
61. Cepeda EJ, Reveille JD. Autoantibodies in systemic sclerosis and fibrosing syndromes: clinical
indications and relevance. Curr Opin Rheumatol 2004; 16:723.
62. Sato S, Kodera M, Hasegawa M, et al. Antinucleosome antibody is a major autoantibody in
localized scleroderma. Br J Dermatol 2004; 151:1182.
63. Assous N, Allanore Y, Batteux F, et al. Prevalence of antiphospholipid antibodies in systemic
sclerosis and association with primitive pulmonary arterial hypertension and endothelial injury.
Clin Exp Rheumatol 2005; 23:199.
64. Arnett FC. Is scleroderma an autoantibody mediated disease? Curr Opin Rheumatol 2006; 18:579.
65. Ruzek MC, Jha S, Ledbetter S, et al. A modified model of graft-versus-host-induced systemic
sclerosis (scleroderma) exhibits all major aspects of the human disease. Arthritis Rheum 2004;
50:1319.
66. Chakravarty E. Pre-disease pregnancy complications and systemic sclerosis: pathogenic or pre-
clinical? Arthritis Res Ther 2012; 14:102.
67. Artlett CM, Smith JB, Jimenez SA. Identification of fetal DNA and cells in skin lesions from
women with systemic sclerosis. N Engl J Med 1998; 338:1186.
68. Nelson JL, Furst DE, Maloney S, et al. Microchimerism and HLA-compatible relationships of
pregnancy in scleroderma. Lancet 1998; 351:559.
69. Evans PC, Lambert N, Maloney S, et al. Long-term fetal microchimerism in peripheral blood
mononuclear cell subsets in healthy women and women with scleroderma. Blood 1999; 93:2033.
70. Artlett CM, Cox LA, Jimenez SA. Detection of cellular microchimerism of male or female origin
in systemic sclerosis patients by polymerase chain reaction analysis of HLA-Cw antigens. Arthritis
Rheum 2000; 43:1062.
71. Johnson KL, Nelson JL, Furst DE, et al. Fetal cell microchimerism in tissue from multiple sites in
women with systemic sclerosis. Arthritis Rheum 2001; 44:1848.
72. Nelson JL. Microchimerism and autoimmune disease. N Engl J Med 1998; 338:1224.
73. Bashkin P, Doctrow S, Klagsbrun M, et al. Basic fibroblast growth factor binds to subendothelial
extracellular matrix and is released by heparitinase and heparin-like molecules. Biochemistry
1989; 28:1737.
74. Leroy EC. Connective tissue synthesis by scleroderma skin fibroblasts in cell culture. J Exp Med
1972; 135:1351.
75. Uitto J, Bauer EA, Eisen AZ. Scleroderma: increased biosynthesis of triple-helical type I and type
III procollagens associated with unaltered expression of collagenase by skin fibroblasts in culture.
J Clin Invest 1979; 64:921.
76. Abraham D, Lupoli S, McWhirter A, et al. Expression and function of surface antigens on
scleroderma fibroblasts. Arthritis Rheum 1991; 34:1164.
77. Shi-Wen X, Panesar M, Vancheeswaran R, et al. Expression and shedding of intercellular adhesion
molecule 1 and lymphocyte function-associated antigen 3 by normal and scleroderma fibroblasts.
Effects of interferon-gamma, tumor necrosis factor alpha, and estrogen. Arthritis Rheum 1994;
37:1689.
78. LeRoy EC, Mercurio S, Sherer GK. Replication and phenotypic expression of control and
scleroderma human fibroblasts: responses to growth factors. Proc Natl Acad Sci U S A 1982;
79:1286.
79. Dziadzio M, Smith RE, Abraham DJ, et al. Circulating levels of active transforming growth factor
beta1 are reduced in diffuse cutaneous systemic sclerosis and correlate inversely with the modified
Rodnan skin score. Rheumatology (Oxford) 2005; 44:1518.
80. Higley H, Persichitte K, Chu S, et al. Immunocytochemical localization and serologic detection of
transforming growth factor beta 1. Association with type I procollagen and inflammatory cell
markers in diffuse and limited systemic sclerosis, morphea, and Raynaud's phenomenon. Arthritis
Rheum 1994; 37:278.
81. Corrin B, Butcher D, McAnulty BJ, et al. Immunohistochemical localization of transforming
growth factor-beta 1 in the lungs of patients with systemic sclerosis, cryptogenic fibrosing
alveolitis and other lung disorders. Histopathology 1994; 24:145.
82. Varga J, Rosenbloom J, Jimenez SA. Transforming growth factor beta (TGF beta) causes a
persistent increase in steady-state amounts of type I and type III collagen and fibronectin mRNAs
in normal human dermal fibroblasts. Biochem J 1987; 247:597.
83. Krieg T, Meurer M. Systemic scleroderma. Clinical and pathophysiologic aspects. J Am Acad
Dermatol 1988; 18:457.
84. Smith EA, LeRoy EC. A possible role for transforming growth factor-beta in systemic sclerosis. J
Invest Dermatol 1990; 95:125S.
85. McWhirter A, Colosetti P, Rubin K, et al. Collagen type I is not under autocrine control by
transforming growth factor-beta 1 in normal and scleroderma fibroblasts. Lab Invest 1994; 71:885.
86. Yamakage A, Kikuchi K, Smith EA, et al. Selective upregulation of platelet-derived growth factor
alpha receptors by transforming growth factor beta in scleroderma fibroblasts. J Exp Med 1992;
175:1227.
87. Ihn H, Yamane K, Kubo M, Tamaki K. Blockade of endogenous transforming growth factor beta
signaling prevents up-regulated collagen synthesis in scleroderma fibroblasts: association with
increased expression of transforming growth factor beta receptors. Arthritis Rheum 2001; 44:474.
88. Kubo M, Ihn H, Yamane K, Tamaki K. Up-regulated expression of transforming growth factor
beta receptors in dermal fibroblasts in skin sections from patients with localized scleroderma.
Arthritis Rheum 2001; 44:731.
89. Asano Y, Ihn H, Yamane K, et al. Impaired Smad7-Smurf-mediated negative regulation of TGF-
beta signaling in scleroderma fibroblasts. J Clin Invest 2004; 113:253.
90. Dong C, Zhu S, Wang T, et al. Deficient Smad7 expression: a putative molecular defect in
scleroderma. Proc Natl Acad Sci U S A 2002; 99:3908.
91. Leask A, Abraham DJ, Finlay DR, et al. Dysregulation of transforming growth factor beta
signaling in scleroderma: overexpression of endoglin in cutaneous scleroderma fibroblasts.
Arthritis Rheum 2002; 46:1857.
92. Gay S, Trabandt A, Moreland LW, Gay RE. Growth factors, extracellular matrix, and oncogenes
in scleroderma. Arthritis Rheum 1992; 35:304.
93. Leask A, Holmes A, Abraham DJ. Connective tissue growth factor: a new and important player in
the pathogenesis of fibrosis. Curr Rheumatol Rep 2002; 4:136.
94. Dziadzio M, Usinger W, Leask A, et al. N-terminal connective tissue growth factor is a marker of
the fibrotic phenotype in scleroderma. QJM 2005; 98:485.
95. Holmes A, Abraham DJ, Sa S, et al. CTGF and SMADs, maintenance of scleroderma phenotype
is independent of SMAD signaling. J Biol Chem 2001; 276:10594.
96. Shi-wen X, Stanton LA, Kennedy L, et al. CCN2 is necessary for adhesive responses to
transforming growth factor-beta1 in embryonic fibroblasts. J Biol Chem 2006; 281:10715.
97. Khanna D, Denton CP, Jahreis A, et al. Safety and efficacy of subcutaneous tocilizumab in adults
with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial. Lancet 2016;
387:2630.
98. Khan K, Xu S, Nihtyanova S, et al. Clinical and pathological significance of interleukin 6
overexpression in systemic sclerosis. Ann Rheum Dis 2012; 71:1235.
99. De Lauretis A, Sestini P, Pantelidis P, et al. Serum interleukin 6 is predictive of early functional
decline and mortality in interstitial lung disease associated with systemic sclerosis. J Rheumatol
2013; 40:435.
100. Feghali CA, Bost KL, Boulware DW, Levy LS. Human recombinant interleukin-4 induces
proliferation and interleukin-6 production by cultured human skin fibroblasts. Clin Immunol
Immunopathol 1992; 63:182.
101. Kawaguchi Y. IL-1 alpha gene expression and protein production by fibroblasts from
patients with systemic sclerosis. Clin Exp Immunol 1994; 97:445.
102. O'Reilly S, Ciechomska M, Cant R, et al. Interleukin-6, its role in fibrosing conditions.
Cytokine Growth Factor Rev 2012; 23:99.
103. Kawaguchi Y, Hara M, Wright TM. Endogenous IL-1alpha from systemic sclerosis
fibroblasts induces IL-6 and PDGF-A. J Clin Invest 1999; 103:1253.
104. Kawaguchi Y, McCarthy SA, Watkins SC, Wright TM. Autocrine activation by interleukin
1alpha induces the fibrogenic phenotype of systemic sclerosis fibroblasts. J Rheumatol 2004;
31:1946.
105. Aden N, Nuttall A, Shiwen X, et al. Epithelial cells promote fibroblast activation via IL-
1alpha in systemic sclerosis. J Invest Dermatol 2010; 130:2191.
106. Distler JH, Jüngel A, Caretto D, et al. Monocyte chemoattractant protein 1 released from
glycosaminoglycans mediates its profibrotic effects in systemic sclerosis via the release of
interleukin-4 from T cells. Arthritis Rheum 2006; 54:214.
107. Aidoudi S, Bujakowska K, Kieffer N, Bikfalvi A. The CXC-chemokine CXCL4 interacts
with integrins implicated in angiogenesis. PLoS One 2008; 3:e2657.
108. Romagnani P, Maggi L, Mazzinghi B, et al. CXCR3-mediated opposite effects of CXCL10
and CXCL4 on TH1 or TH2 cytokine production. J Allergy Clin Immunol 2005; 116:1372.
109. van Bon L, Affandi AJ, Broen J, et al. Proteome-wide analysis and CXCL4 as a biomarker
in systemic sclerosis. N Engl J Med 2014; 370:433.
110. Rönnblom L, Pascual V. The innate immune system in SLE: type I interferons and
dendritic cells. Lupus 2008; 17:394.
111. van der Pouw Kraan TC, Wijbrandts CA, van Baarsen LG, et al. Rheumatoid arthritis
subtypes identified by genomic profiling of peripheral blood cells: assignment of a type I interferon
signature in a subpopulation of patients. Ann Rheum Dis 2007; 66:1008.
112. Eloranta ML, Franck-Larsson K, Lövgren T, et al. Type I interferon system activation and
association with disease manifestations in systemic sclerosis. Ann Rheum Dis 2010; 69:1396.
113. Kim D, Peck A, Santer D, et al. Induction of interferon-alpha by scleroderma sera
containing autoantibodies to topoisomerase I: association of higher interferon-alpha activity with
lung fibrosis. Arthritis Rheum 2008; 58:2163.
114. Carulli MT, Ong VH, Ponticos M, et al. Chemokine receptor CCR2 expression by systemic
sclerosis fibroblasts: evidence for autocrine regulation of myofibroblast differentiation. Arthritis
Rheum 2005; 52:3772.
115. Ong VH, Evans LA, Shiwen X, et al. Monocyte chemoattractant protein 3 as a mediator of
fibrosis: Overexpression in systemic sclerosis and the type 1 tight-skin mouse. Arthritis Rheum
2003; 48:1979.
116. Antonelli A, Ferri C, Fallahi P, et al. CXCL10 (alpha) and CCL2 (beta) chemokines in
systemic sclerosis--a longitudinal study. Rheumatology (Oxford) 2008; 47:45.
117. Tiev KP, Hua-Huy T, Kettaneh A, et al. Serum CC chemokine ligand-18 predicts lung
disease worsening in systemic sclerosis. Eur Respir J 2011; 38:1355.
118. Schmidt K, Martinez-Gamboa L, Meier S, et al. Bronchoalveoloar lavage fluid cytokines
and chemokines as markers and predictors for the outcome of interstitial lung disease in systemic
sclerosis patients. Arthritis Res Ther 2009; 11:R111.
119. Needleman BW, Wigley FM, Stair RW. Interleukin-1, interleukin-2, interleukin-4,
interleukin-6, tumor necrosis factor alpha, and interferon-gamma levels in sera from patients with
scleroderma. Arthritis Rheum 1992; 35:67.
120. Takemura H, Suzuki H, Yoshizaki K, et al. Anti-interleukin-6 autoantibodies in rheumatic
diseases. Increased frequency in the sera of patients with systemic sclerosis. Arthritis Rheum 1992;
35:940.
121. Furuse S, Fujii H, Kaburagi Y, et al. Serum concentrations of the CXC chemokines
interleukin 8 and growth-regulated oncogene-alpha are elevated in patients with systemic sclerosis.
J Rheumatol 2003; 30:1524.
122. Kurasawa K, Hirose K, Sano H, et al. Increased interleukin-17 production in patients with
systemic sclerosis. Arthritis Rheum 2000; 43:2455.
123. Shi-Wen X, Denton CP, McWhirter A, et al. Scleroderma lung fibroblasts exhibit elevated
and dysregulated type I collagen biosynthesis. Arthritis Rheum 1997; 40:1237.
124. Korn JH, Thrall RS, Wilbur DC, et al. Fibroblast heterogeneity: clonal selection of fibrobl
asts as a model for fibrotic disease. In: Pulmonary fibroblast heterogeneity, Phipps RP (Ed), CRC
Press, Boca Raton, Florida 1992.
125. Kähäri VM, Sandberg M, Kalimo H, et al. Identification of fibroblasts responsible for
increased collagen production in localized scleroderma by in situ hybridization. J Invest Dermatol
1988; 90:664.
126. Scharffetter K, Lankat-Buttgereit B, Krieg T. Localization of collagen mRNA in normal
and scleroderma skin by in-situ hybridization. Eur J Clin Invest 1988; 18:9.
127. Wei J, Ghosh AK, Sargent JL, et al. PPARγ downregulation by TGFß in fibroblast and
impaired expression and function in systemic sclerosis: a novel mechanism for progressive
fibrogenesis. PLoS One 2010; 5:e13778.
128. Palumbo-Zerr K, Zerr P, Distler A, et al. Orphan nuclear receptor NR4A1 regulates
transforming growth factor-β signaling and fibrosis. Nat Med 2015; 21:150.
129. Gilbane AJ, Denton CP, Holmes AM. Scleroderma pathogenesis: a pivotal role for
fibroblasts as effector cells. Arthritis Res Ther 2013; 15:215.
130. Denton CP, Khan K, Hoyles RK, et al. Inducible lineage-specific deletion of TbetaRII in
fibroblasts defines a pivotal regulatory role during adult skin wound healing. J Invest Dermatol
2009; 129:194.
131. Hoyles RK, Derrett-Smith EC, Khan K, et al. An essential role for resident fibroblasts in
experimental lung fibrosis is defined by lineage-specific deletion of high-affinity type II
transforming growth factor β receptor. Am J Respir Crit Care Med 2011; 183:249.
132. Denton CP, Ong VH. Targeted therapies for systemic sclerosis. Nat Rev Rheumatol 2013;
9:451.
133. Trojanowska M, Wu LT, LeRoy EC. Elevated expression of c-myc proto-oncogene in
scleroderma fibroblasts. Oncogene 1988; 3:477.
134. Jimenez SA, Gaidarova S, Saitta B, et al. Role of protein kinase C-delta in the regulation
of collagen gene expression in scleroderma fibroblasts. J Clin Invest 2001; 108:1395.
135. Shiwen X, Stratton R, Nikitorowicz-Buniak J, et al. A Role of Myocardin Related
Transcription Factor-A (MRTF-A) in Scleroderma Related Fibrosis. PLoS One 2015;
10:e0126015.
136. Bhattacharyya S, Tamaki Z, Wang W, et al. FibronectinEDA promotes chronic cutaneous
fibrosis through Toll-like receptor signaling. Sci Transl Med 2014; 6:232ra50.
137. Wallis DD, Tan FK, Kielty CM, et al. Abnormalities in fibrillin 1-containing microfibrils
in dermal fibroblast cultures from patients with systemic sclerosis (scleroderma). Arthritis Rheum
2001; 44:1855.
138. Tan FK, Stivers DN, Foster MW, et al. Association of microsatellite markers near the
fibrillin 1 gene on human chromosome 15q with scleroderma in a Native American population.
Arthritis Rheum 1998; 41:1729.
139. Siracusa LD, McGrath R, Ma Q, et al. A tandem duplication within the fibrillin 1 gene is
associated with the mouse tight skin mutation. Genome Res 1996; 6:300.
140. Wang Y, Fan PS, Kahaleh B. Association between enhanced type I collagen expression
and epigenetic repression of the FLI1 gene in scleroderma fibroblasts. Arthritis Rheum 2006;
54:2271.
141. Ivarsson M, McWhirter A, Black CM, Rubin K. Impaired regulation of collagen pro-alpha
1(I) mRNA and change in pattern of collagen-binding integrins on scleroderma fibroblasts. J Invest
Dermatol 1993; 101:216.
142. Eckes B, Mauch C, Hüppe G, Krieg T. Downregulation of collagen synthesis in fibroblasts
within three-dimensional collagen lattices involves transcriptional and posttranscriptional
mechanisms. FEBS Lett 1993; 318:129.
143. Takeda K, Hatamochi A, Ueki H, et al. Decreased collagenase expression in cultured
systemic sclerosis fibroblasts. J Invest Dermatol 1994; 103:359.
144. Kirk TZ, Mark ME, Chua CC, et al. Myofibroblasts from scleroderma skin synthesize
elevated levels of collagen and tissue inhibitor of metalloproteinase (TIMP-1) with two forms of
TIMP-1. J Biol Chem 1995; 270:3423.
145. Hebbar M, Peyrat JP, Hornez L, et al. Increased concentrations of the circulating
angiogenesis inhibitor endostatin in patients with systemic sclerosis. Arthritis Rheum 2000;
43:889.

You might also like