Next Article in Journal
The Central Role of the NAD+ Molecule in the Development of Aging and the Prevention of Chronic Age-Related Diseases: Strategies for NAD+ Modulation
Next Article in Special Issue
Immune Mechanisms of Pulmonary Fibrosis with Bleomycin
Previous Article in Journal
Role of Uric Acid in Vascular Remodeling: Cytoskeleton Changes and Migration in VSMCs
Previous Article in Special Issue
Wilms Tumor 1-Driven Fibroblast Activation and Subpleural Thickening in Idiopathic Pulmonary Fibrosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Molecular Mechanisms of Systemic Sclerosis-Associated Lung Fibrosis

by
Joe E. Mouawad
1,2 and
Carol Feghali-Bostwick
1,*
1
Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
2
Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC 29425, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2963; https://doi.org/10.3390/ijms24032963
Submission received: 9 November 2022 / Revised: 30 January 2023 / Accepted: 31 January 2023 / Published: 3 February 2023

Abstract

:
Systemic sclerosis (SSc), also known as scleroderma, is an autoimmune disorder that affects the connective tissues and has the highest mortality rate among the rheumatic diseases. One of the hallmarks of SSc is fibrosis, which may develop systemically, affecting the skin and virtually any visceral organ in the body. Fibrosis of the lungs leads to interstitial lung disease (ILD), which is currently the leading cause of death in SSc. The identification of effective treatments to stop or reverse lung fibrosis has been the main challenge in reducing SSc mortality and improving patient outcomes and quality of life. Thus, understanding the molecular mechanisms, altered pathways, and their potential interactions in SSc lung fibrosis is key to developing potential therapies. In this review, we discuss the diverse molecular mechanisms involved in SSc-related lung fibrosis to provide insights into the altered homeostasis state inherent to this fatal disease complication.

1. Introduction

Systemic sclerosis (SSc), or scleroderma, is an autoimmune connective tissue disease with one of the highest mortality rates among the rheumatic diseases [1]. Fibrosis is recognized to be a defining feature of SSc, affecting the skin and multiple visceral organs [2]. As a result, SSc is considered the prototypic fibrosing disease. For more than 20 years, interstitial lung disease (ILD), characterized by lung fibrosis, has been the leading cause of death in SSc [3,4]. This is largely due to the lack of treatments that can stop or reverse the fibrotic process. Currently, only two drugs are approved by the Food and Drug Administration (FDA) for SSc, but these merely reduce the progression of ILD rather than stop or reverse it [5,6]. SSc thus remains incurable due to progressive lung fibrosis, with lung transplantation as the only viable option, which is impossible on the scale that it is needed [7].
The molecular mechanisms mediating SSc-related lung fibrosis are complex and incompletely understood. While the final outcome is the excessive deposition of extracellular matrix (ECM) resulting in lung fibrosis, it is evident that the pathways leading to this outcome are numerous, involving different molecular and cellular components [8]. Understanding the different pathogenic mechanisms that contribute to lung fibrosis in SSc and their interplay is key to identifying potential molecular targets for therapy. In this review, we describe the different molecular mechanisms currently implicated in SSc-related lung fibrosis in an effort to establish a well-needed comprehensive source for better understanding the disease pathogenesis.

2. Gene Expression Profile of SSc

High-throughput gene expression studies on SSc lung tissues and cells have proven to be valuable in identifying molecular pathways underpinning the pathogenesis of SSc-associated lung fibrosis. A microarray analysis performed on normal versus explanted SSc lung tissues and matching primary pulmonary fibroblasts revealed that the latter showed differentially expressed genes corresponding to ECM components and fibrotic signaling molecules as well as novel genes and pathways that were not previously reported in the SSc lung. Molecular signatures included those that were unique to SSc and those that correlated with the fibrotic phenotype in SSc and idiopathic pulmonary fibrosis (IPF) [9,10]. Pro-fibrotic and ECM signatures were also identified in an independent microarray analysis of SSc lung biopsies [11]. This latter study also identified a macrophage activation signature and interferon signatures [11]. It is worth noting that Hsu et al.’s analysis focused on SSc lungs with primary usual interstitial pneumonia on pathology [9], while Christmann et al. included lung tissues with non-specific interstitial pneumonia on pathology [11]. Moreover, Tyler et al. identified a three-gene network of interacting alleles in a cohort of 416 SSc patients, namely, WNT5A, RBMS3, and MS12, which influenced lung outcomes in SSc. Gene expression profiling has also focused on the fibroblasts in the lung, as they are the effector cells in fibrosis [12,13]. A bulk RNA sequencing (RNA-seq) analysis of fibroblasts derived from healthy versus SSc lung tissues revealed an expression profile similar to that of whole lung tissues, with increased expression of ECM genes, including COL1A1 and COL3A1, and fibrotic genes such as TGFB2, IGF2, IGFBP3, IGFBP5, and WNT5A, while antifibrotic genes such as MMP1, MMP9, CTSL, SFRP1, and IL33 were underexpressed in SSc lung fibroblasts [14]. Taken together, these findings suggest that fibroblasts are the predominant source of the aberrant lung tissue expression profile identified using microarray analysis. These findings also emphasize the notion that fibrosis is a dynamic process caused by the disruption of a balance to favor pro-fibrotic factors over anti-fibrotic pathways. More recently, single-cell RNA-seq (scRNA-seq) technology has proven to be a powerful tool to garner insights into individual cellular contributions to lung fibrosis in SSc [15]. Valenzi et al. demonstrated, using scRNA-seq, that SSc lung tissues show a unique heterogeneity in fibroblast populations, namely, SPINT2hi, MFAPhi, WIFhi, and a new myofibroblast population with high ACTA2 expression, all showing differential expression profiles when compared with the control [13]. The data reinforced that myofibroblast differentiation and proliferation are key drivers of disease pathogenesis [13]. Similarly, using scRNA-seq, Reyfman et al. showed differential gene expression in the fibroblast subpopulations of fibrotic lungs that included but were not limited to SSc-ILD [16]. Their analysis also identified a novel population of pro-fibrotic alveolar macrophages, distinct epithelial cell signatures, and novel airway stem cells that were across fibrosing lung diseases [16]. In activated fibroblasts, the differential expression of ECM and pro-fibrotic genes including COL1A1, COL3A1, POSTN, TAGLN, and ACTA2 was also noted, validating findings of microarray and RNAseq studies [17]. These findings further reinforce the established disease paradigm that fibroblasts are the driving force behind lung tissue fibrosis in SSc. Thus, understanding the various molecular mechanisms of fibroblast activation and proliferation in SSc is essential for mapping the pathology of the disease.

3. Deregulated Molecular Pathways in SSc

3.1. Fibrotic Factors

3.1.1. Transforming Growth Factor Beta (TGFβ)

TGFβ is one of the most widely studied pro-fibrotic factors in the context of fibrosis. The TGFβ superfamily includes TGFβ, bone morphogenetic proteins (BMPs), growth/differentiation factors (GDFs), activins, and inhibins [18]. TGFβ plays a crucial role in transitioning fibroblasts into activated myofibroblasts, which are responsible for the excessive production of ECM in fibrosis [19]. TGFβ signals via interaction with two receptor serine/threonine kinases, known as type I and type II receptors, which form a heterotetrameric complex upon ligand binding [20]. Upon complexing, the autophosphorylation of type I and II receptors mediates the docking and phosphorylation of Smad 2/3, which in turn interact with Smad 4 to create a transcriptional complex that translocates to the nucleus and activates or represses multiple target genes [21]. While TGFβ works mainly via activation of the Smad pathway, it can also activate other non-canonical pathways [18]. In fibroblasts, TGFβ signaling shifts the gene expression profile to a profibrotic phenotype, inducing the expression of profibrotic and ECM genes, while suppressing the antifibrotic and matrix-degrading genes, leading to tissue fibrosis [19,22]. TGFβ can also exert similar effects on other cell types, such as epithelial and endothelial cells, and can induce their transition into alpha smooth muscle actin-expressing myofibroblasts [19,23].
In SSc, TGFβ-regulated genes are differentially expressed in the fibrotic lungs of patients, which is correlated with the severity of the disease [24]. This is consistent with the notion that TGFβ plays a central role in SSc pathology [25]. In fact, we previously demonstrated that fibroblasts derived from the lungs of SSc patients express higher TGFβ1 and TGFβ2 levels than fibroblasts from healthy lungs [26]. This supports previous findings obtained by Christmann et al. indicating that lung tissues from SSc patients show amplified expression of genes regulated by TGFβ [11]. Recent evidence has shown that macrophages polarized to the alternatively activated phenotype (M2) are also a major source of TGFβ [27]. A follow-up study showed that the expression of TGFβ by M2 macrophages is amplified by methyl-CpG-binding domain 2 (MBD2) protein, which suppresses the expression of an inhibitor upstream of TGFβ, and MBD2 was found to be overexpressed in SSc-ILD lung tissues [28]. Zehender et al. recently demonstrated a novel mechanism for TGFβ-induced fibrosis in SSc, which involves a loss of epigenetic control over autophagy via a Smad3-dependent downregulation of the H4K16 histone acetyltransferase MYST1, mediating the activation of fibroblasts [29]. Core regulators of autophagy, BECLIN1 and ATG7, were consequently found to be upregulated in SSc dermal fibroblasts, as well as fibrotic skin and lungs of mice overexpressing TGFBRI, while their knockdown alleviated fibrosis [29].
Efforts to target TGFβ as a therapeutic strategy to reduce SSc lung fibrosis have not been effective, and concerns about potential adverse complications due to the pleiotropic roles of TGFβ in lung physiology have led to efforts focusing on targeting other pro-fibrotic factors and molecular pathways as a therapeutic strategy to treat SSc [1,30,31].
It is worth noting that several members of the TGFβ family have been largely overlooked in SSc lung research to date, although they are likely to play important roles in disease pathogenesis. Unlike TGFβ, whose active form is generated when and where it is needed, activin A and BMP4 are generally readily active and thus possess distinct signaling dynamics from TGFβ-induced fibrosis [32].

3.1.2. Platelet-Derived Growth Factor (PDGF)

PDGF has also been shown to play a central role in organ fibrosis, since stromal mesenchymal cells, including fibroblasts, express PDGF receptor isoforms that are activated and drive processes implicated in fibrosis, such as proliferation, migration, and ECM deposition [33]. In fact, lung fibrosis of various etiologies, whether environmental exposure, transplant rejection, autoimmune, or idiopathic, have been associated with increased PDGF levels in bronchoalveolar lavage fluid (BALF) or lung tissues [34]. There are two PDGF receptor isoforms, PDGFRα and PDGFRβ, which are tyrosine kinase receptors recognized by four ligand isoforms, PDGF-A, PDGF-B, PDGF-C, and PDGF-D [35]. Upon ligand binding, the homo- or hetero-dimerization of the receptors leads to autophosphorylation events of their cytoplasmic domain, which activates downstream signaling pathways, including phosphatidylinositol 3 kinase (PI3K), Ras-MAPK, Src, and phospholipase Cγ (PLCγ) pathways [35]. More recently, we showed that PDGF can also signal via melanin-concentrating hormone receptor 1 (MCHR1), modulating intracellular cyclic adenosine monophosphate (cAMP) production and inducing TGFβ1 and connective tissue growth factor (CTGF) expression in fibroblasts, thus promoting a fibrotic response [36].
PDGF-A and PDGF-B levels are elevated in the BALF of SSc patients [37]. Interestingly, SSc-derived fibroblasts exhibit unique, positive cross-talk between PDGF and TGFβ signaling, which does not occur in healthy fibroblasts [38]. In addition, microRNA miR-30b, which suppresses the expression of PDGFRβ, is downregulated in the serum of SSc patients [39]. Reducing the expression of PDGFRβ in SSc dermal fibroblasts with miR-30b inhibited collagen synthesis and myofibroblast activation [39]. Since miR-30b levels are decreased in the circulation of SSc patients, it is reasonable to extrapolate these findings to lung fibroblasts. Recently, Svegliati et al. demonstrated that PDGF and anti-PDGFR autoantibodies, which are elevated in SSc patient serum [40,41], stimulated higher growth rate, migration, and expression of collagen in human pulmonary artery smooth muscle cells, which was attributed to the generation of reactive oxygen species, and elevated NOX4 and mammalian target of rapamycin (mTOR) [42]. All these findings have made PDGF an attractive molecular target for therapeutic treatment of SSc lung fibrosis [43]. In fact, nintedanib, a drug that blocks the ATP-binding pocket of PDGFR and other tyrosine kinase receptors, such as fibroblast growth factor receptor (FGFR) and vascular endothelial growth factor receptor (VEGFR), was approved by the FDA for the treatment of SSc-ILD [6,44]. At the cellular level, nintedanib blocks the PDGF-induced differentiation of lung fibroblasts into myofibroblasts, reduces their proliferation and migration, and suppresses the expression of collagen and fibronectin, supporting the antifibrotic outcome of blocking PDGF signaling.

3.1.3. Fibroblast Growth Factor (FGF)

FGFs are a family of signaling proteins that can act in an endocrine, paracrine, or even intracrine manner [45]. Under paracrine or endocrine conditions, target cells interact with FGF ligands via four receptor tyrosine kinases, FGFR1, FGFR2, FGFR3, and FGFR4 [45]. Intracrine FGFs are nonsignaling, in that they act independently of FGFRs and mainly serve as cofactors for voltage-gated sodium channels [45,46]. Upon ligand binding, FGFRs activate multiple pathways, including PI3K, Ras-MAPK, PLCγ, and STAT signaling pathways [45]. The roles of the different FGF ligands in fibrosis have been variable in the experimental models, with some promoting lung fibrosis and others protecting against it [47]. For example, members of the FGF family of proteins can activate fibroblasts and induce their proliferation and ECM deposition [48]. In contrast, one member of the family, FGF19, was found to be protective against lung fibrosis in mice, and its levels were decreased in the plasma of IPF patients [49]. However, studies about the specific role of FGFs in SSc-related lung fibrosis are scarce. Recently, Chakraborty et al. demonstrated a mechanistic involvement of FGF9 and its receptor FGFR3 in SSc, both of which are upregulated in SSc fibrotic skin [50]. FGF9 was shown to bind FGFR3 and activate dermal fibroblasts from SSc skin, leading to the downstream stimulation of AKT, p38, extracellular signal-regulated kinase (ERK), and calcium/calmodulin-dependent protein kinase 2 (CAMK2) and promoting cyclic adenosine 3′,5′-monophosphate response element binding protein (CREB) activation, which induced the expression of profibrotic mediators [50]. These findings from SSc skin have not yet been validated in tissues or primary cells derived from lungs of SSc patients.

3.1.4. Wnt/β-Catenin Signaling

Widely known for its role in organ and tissue development, the Wnt/β-catenin signaling pathway has more recently been implicated in fibrotic disorders in different organs [51,52,53,54]. The binding of Wnt ligands to their Frizzled (Fz) receptors triggers downstream effects inhibiting the degradation of β-catenin, stabilizing it in the cytoplasm, and promoting its translocation to the nucleus, where it results in the transcription of Wnt target genes [55]. Earlier studies have confirmed the involvement of the Wnt/β-catenin pathway in the pathogenesis of lung fibrosis and, specifically, in SSc [56,57,58]. In addition, SSc skin fibroblasts express high levels of Wnt proteins such as Wnt1 and Wnt10b, coupled with decreased expression of the Wnt antagonists SFRP1, DKK1, and WIF1 [59]. Our group recently reported similar results in SSc lung fibroblasts, showing increased Wnt5a and decreased SFRP1 expression in SSc lung fibroblasts compared with normal lung fibroblasts [14]. More recently, increased levels of a novel isoform of CD146 that activates myofibroblasts were noted in the serum of SSc patients with pulmonary fibrosis, a process driven by Wnt5a [60]. This is consistent with previous studies showing that the Wnt/β-catenin pathway is activated in SSc lung fibrosis, allowing its downstream pro-fibrotic effects to be promoted [57,61].

3.1.5. Interleukins

Interleukins (ILs) are a group of cytokines with immunoregulatory functions known to be secreted by white blood cells, but also several other cell types, such as epithelial and stromal cells [62,63]. There are more than 40 distinct ILs, each eliciting different functions across multiple different cell types via binding to high-affinity receptors [64]. Several of these ILs have been shown to directly interact with fibroblasts to promote lung fibrosis [65].
IL-6 has been extensively studied in the context of SSc, and its levels are increased in SSc serum, skin, and fibroblasts [66,67,68,69]. Serum levels of IL-6 strongly correlate with the severity of SSc lung fibrosis and are predictive of mortality in SSc patients, suggesting a profibrotic role of IL-6 in lung tissues [70]. Our group confirmed a significant increase in IL-6 expression in lung tissues derived from SSc patients compared with healthy lung tissues [10] and showed that the increase in lung fibroblasts is mechanistically driven, at least in part, by upregulated lysyl oxidase (LOX) via activation of c-Fos [71]. Tocilizumab, a monoclonal antibody targeting the IL-6 receptor, preserved lung function in SSc patients, slowing lung fibrosis progression, and was approved by the FDA for the treatment of SSc-ILD [5,72,73].
The involvement of multiple other ILs in SSc-related lung fibrosis has been documented. Yaseen et al. demonstrated a significant increase in IL-31 in SSc serum coupled with the overexpression of its receptor, IL-31RA, in SSc-derived lung fibroblasts, and confirmed the profibrotic effects of IL-31 in mouse lungs [74]. Moreover, IL-11 was significantly upregulated in SSc lung fibroblasts [75,76]. Follow-up studies confirmed that IL-11 drives the activation of lung fibroblasts. Specifically, mice with conditional fibroblast-specific knockout of the IL-11 receptor were protected from bleomycin-induced lung fibrosis, and IL-11 knockout lung fibroblasts were refractory to TGFβ stimulation [77]. IL-1 induces miR-155, which was overexpressed in SSc lung fibroblasts, leading to increased TGFβ and collagen synthesis, as well as further feed-forward expression of IL-1 driven by inflammasome activation [78]. Other interleukins whose serum levels correlated with ILD severity in SSc patients include IL-13, IL-17, IL-33, and IL-34; however, their exact roles in promoting lung fibrosis are incompletely delineated [79,80,81,82].

3.1.6. Insulin-Like Growth Factors (IGFs) and Their Binding Proteins (IGFBPs)

IGFs and IGFBPs have been implicated in the pathogenesis of SSc lung fibrosis. IGF-I levels are increased in the BALF of SSc patients, and the protein itself promotes the proliferation of fibroblasts [83]. IGF-II expression is also increased in fibrotic SSc lung tissues and fibroblasts and induces ECM deposition via the activation of PI3K and Jun N-terminal kinase pathways [84]. A follow-up study demonstrated that IGF-II signaled via type 1 IGF receptor (IGF1R), insulin receptor (IR), and a hybrid IGF1R/IR complex receptor, promoting fibrosis through multiple mechanisms: by directly activating myofibroblasts, by increasing ECM production while reducing its degradation, and by stimulating the expression of TGFβ2 and TGFβ3 [26]. We showed that IGFBP-3 and IGFBP-5 are profibrotic proteins that stimulate the production of ECM by lung fibroblasts [85,86,87]. IGFBP-5 is significantly overexpressed in SSc lung tissues and fibroblasts and induces the expression of collagen type I, fibronectin, CTGF, LOX, and DOK5 [88,89]. Mice expressing human IGFBP-5 showed sustained increased expression of ECM genes [90]. Recently, IGFBP-2 serum levels were found to have prognostic value for assessing the development of ILD in SSc patients, but further studies are needed to understand its potential role in lung disease [91].

3.1.7. YAP/TAZ

Yes-associated protein (YAP) and transcription coactivator with PDZ-binding motif (TAZ) are key components of the Hippo pathway [92]. Mammalian STE20-like (MST) and large tumor suppressor kinase (LATS) are the core proteins upstream of YAP/TAZ in the Hippo pathway [93]. When the Hippo pathway is activated, the sequential phosphorylation of MST, LATS, and then YAP/TAZ occurs, causing the retention of YAP/TAZ in the cytoplasm [94]. YAP/TAZ signaling mediates its effect via cross-talk with multiple other pathways, including the TGFβ and Wnt/β-catenin pathways [94]. YAP/TAZ was shown to promote myofibroblast proliferation, contraction, and ECM synthesis [95]. Myofibroblast-specific YAP/TAZ deficiency ameliorates fibrosis across multiple organs, including lungs, kidneys, and liver, supporting previous findings on the critical role that YAP/TAZ signaling plays in fibrogenesis [96]. Further delineation of the role of YAP/TAZ signaling in SSc-related fibrosis is warranted. Toyoma et al. demonstrated that targeting YAP/TAZ with dimethyl fumarate is a viable therapeutic strategy for dermal fibrosis in SSc [97]. More recently, Wu et al. showed that skin fibroblasts and serum from SSc patients had increased levels of YAP and TAZ compared with healthy controls, and they demonstrated that knockdown of YAP/TAZ in mice alleviated bleomycin-induced lung fibrosis [98].

3.1.8. Chemokines

Chemokines are a family of small proteins that play a critical role in maintaining homeostasis and regulating inflammation and tissue-specific leukocyte migration [99]. Upon tissue damage, chemokines initiate and maintain the inflammatory process, and the fine-tuning of their expression is imperative for the resolution of inflammation, culminating in tissue repair and wound healing [100]. Chemokines can activate fibroblasts and perpetuate their ECM production and deposition [100]. This has led to research on the roles of certain chemokines in SSc lung disease. Specifically, C-C motif ligand-2 (CCL2) levels were elevated in the BALF of SSc patients compared with healthy controls [101]. Furthermore, CCL2 protein was overexpressed in fibroblasts from fibrotic lungs of SSc patients [102]. Wu et al. demonstrated that CCL2 plasma levels can serve as a biomarker and a potential therapeutic target for ILD progression in SSc patients, as higher CCL2 levels predicted a faster decline in forced vital capacity (FVC%) over time [103]. Another chemokine, CXCL4, was reported by van Bon et al. to be elevated in SSc patients and correlated with the severity of lung fibrosis [104]. A follow-up study by Affandi et al. demonstrated that CXCL4 is required for bleomycin-induced lung fibrosis in mice, where it promoted myofibroblast transformation, leading to excessive ECM deposition [105]. Multiple other chemokines have been implicated in SSc lung fibrosis, such as CCL5, CCL7, CCL18, CXCL3, and CXCL8, mostly as potential biomarkers of ILD [100,106,107]. Further studies are still needed to elucidate which of these chemokines are central to the pathogenesis of SSc lung fibrosis.

3.2. Anti-Fibrotic Factors

While most studies focus on the pro-fibrotic molecular contributors to SSc lung fibrosis, reduced levels and/or blunted activity of anti-fibrotic pathways constitute a crucial component of uncontrolled fibrosis. Here, we describe anti-fibrotic proteins and pathways whose reduced expression or activity has been implicated in SSc lung fibrosis.
Cathepsins are lysosomal proteases widely known for their role in intracellular housekeeping for the maintenance of cellular homeostasis, such as antigen processing and the degradation of proteases and chemokines [108]. However, cathepsins are not limited to the lysosome and have been shown to play important extracellular functions, particularly in remodeling and degrading the ECM [109,110]. Cathepsin S (CTSS) levels were significantly decreased in the serum of SSc patients compared with healthy controls, and the reduced levels were reflective of the severity of SSc lung fibrosis [111]. More recently, we reported that Cathepsin L (CTSL) expression was significantly reduced in lung fibroblasts and tissues derived from SSc patients, in part due to constitutive suppression by the TGFβ/Smad pathway, and that the lack of availability of CTSL in the extracellular milieu prevented the release of the anti-fibrotic protein endostatin [14]. Endostatin is released by proteolytic cleavage from the C-terminus of collagen-XVIII and has shown potent anti-fibrotic effects on human fibroblasts and human tissues as well murine models of fibrosis [112]. Endostatin is in fact detected in the circulation and BALF of SSc patients and patients with other lung fibrosing diseases, but its levels do not reach therapeutic levels, suggesting a blunted anti-fibrotic response in SSc-associated fibrosis [113,114].
Matrix metalloproteinases (MMPs) are ECM-degrading enzymes that play an important role in maintaining tissue matrix homeostasis [115]. We showed, using bulk RNAseq, that the expression of multiple MMPs, including MMP1, MMP9, MMP15, and MMP28, is reduced in lung fibroblasts derived from SSc patients compared with healthy controls [14]. MMP19-deficient mice had an augmented lung fibrotic response to bleomycin when compared with wildtype mice, and their lung fibroblasts overexpressed collagen type I and alpha smooth muscle actin, a marker of myofibroblast activation [116,117]. In addition, E4, an anti-fibrotic peptide derived from the C-terminus of endostatin, activates the urokinase pathway in primary lung fibroblasts and lung tissues, leading to the induction of MMP1 and MMP3 and consequently promoting matrix degradation and fibrosis resolution [118].
Multiple other proteins have been shown to play a protective role against SSc-related lung fibrosis. For example, Chu et al. demonstrated that deacetylase Sirtuin 1 was underexpressed in the peripheral blood mononuclear cells (PBMCs) of SSc patients with lung fibrosis, and its activation in bleomycin-treated mice or overexpression in human lung fibroblasts reduced collagen production and ameliorated the fibrotic response [119]. Sirtuin 3 has also been implicated in SSc skin and lung fibrosis [120]. Further, decreased serum levels of Sirtuins 1 and 3 correlated with lung fibrosis in SSc [121]. The expression of other sirutins, notably, Sirtuin 7, is also decreased in SSc lung fibroblasts, resulting in increased Smad3 levels [122]. As we note above, two members of the IGFBP family, IGFBP-3 and -5, are increased in SSc skin and lung and promote fibrosis [85,86,87]. In contrast, another member of the family, IGFBP-4, is underexpressed in SSc lung fibroblasts and exhibits anti-fibrotic activity via the suppression of CTGF and C-X-C chemokine receptor 4 (CXCR4) [123]. Antagonism of Wnt signaling is another anti-fibrotic pathway. The expression of secreted frizzled proteins (SFRPs) 1 and 4, which are Wnt antagonists, was suppressed upon bleomycin administration in mice via the hypermethylation of the promoter region, which induced Wnt overactivity and progression of lung fibrosis [124]. Another Wnt antagonist, DKK1, was also suppressed via the hypermethylation of its gene promoter in SSc fibroblasts [125]. This is supported by findings showing that methyl cap binding protein-2 (MeCP2), which methylates the SFRP1 and DKK1 promoter regions, is overexpressed in SSc fibroblasts, resulting in reduced expression of its target genes [126,127]. In addition, miR27a-3p, which targets SFRP1 expression, is upregulated in SSc fibroblasts, leading to reduced SFRP1 levels in the circulation of SSc patients [128]. While the latter studies were mainly focused on SSc dermal fibroblasts, our group confirmed the reduced expression of SFRP1 and DKK1, coupled with Wnt overexpression, in SSc lung fibroblasts, supporting the protective role of these Wnt antagonists also in SSc lung fibrosis [14]. Taken together, the reduced expression of anti-fibrotic genes in SSc-associated lung fibrosis supports the concept of a blunted or suppressed anti-fibrotic response in SSc.

3.3. Extracellular Molecules as Pro- and Anti-Fibrotic Mediators

In addition to increased production of ECM components and the enzymes responsible for their crosslinking in fibrosis, matricryptins have emerged as important players in fibrosis. Matricryptins are cleavage products of extracellular matrix proteins and glycosaminoglycans that exert biological activity. For example, endostatin is cleaved from Collagen XVIII and exerts anti-fibrotic effects [112]. In contrast, endotrophin, released from Collagen VI, has pro-fibrotic effects [129]. In addition, enzymes extensively studied for their matrix crosslinking function can harbor additional functions relevant to fibrosis. As an example, LOX has moonlighting functions that include the induction of IL-6 and the increased production of ECM components [71]. Thus, approaches to mitigate lung fibrosis in SSc should take into consideration the levels and activity of the various enzymes that cleave molecules in the ECM, resulting in the release of matricryptins [130], and those implicated in ECM stabilization that have additional novel functions in promoting fibrosis.

4. Conclusions

ILD remains the leading cause of death in SSc due to the lack of effective treatments that halt or reverse fibrosis. Understanding the multitude of molecular pathways involved in SSc-driven lung fibrosis and building a comprehensive view of their interconnections are critical to identify effective targets for anti-fibrotic therapies. The dynamic nature of fibrosis is the result of an abundance of pro-fibrotic factors and paucity of anti-fibrotic ones (Figure 1). While most therapeutic approaches to SSc-ILD have focused on targeting profibrotic molecular pathways, strategies to induce the endogenous anti-fibrotic response could serve as a viable and more effective therapeutic strategy, since endogenous molecules are less likely to demonstrate toxicity or elicit an autoimmune response. The end goals are to restore homeostasis, halt the progression lung fibrosis, and even reverse it, thus reducing mortality in SSc and other fibrosing lung diseases such as IPF.

Author Contributions

J.E.M. wrote the first draft; J.E.M. and C.F.-B. edited and approved the article. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a pre-doctoral fellowship to J.M. from National Scleroderma Foundation and by NIH grants K24AR060297 and R01HL153195 to C.F.-B.

Conflicts of Interest

The authors declare no relevant conflicts of interest.

References

  1. Denton, C.P.; Khanna, D. Systemic sclerosis. Lancet 2017, 390, 1685–1699. [Google Scholar] [CrossRef] [PubMed]
  2. Ho, Y.Y.; Lagares, D.; Tager, A.M.; Kapoor, M. Fibrosis-A lethal component of systemic sclerosis. Nat. Rev. Rheumatol. 2014, 10, 390–402. [Google Scholar] [CrossRef] [PubMed]
  3. Perelas, A.; Silver, R.M.; Arrossi, A.V.; Highland, K.B. Systemic sclerosis-associated interstitial lung disease. Lancet Respir. Med. 2020, 8, 304–320. [Google Scholar] [CrossRef] [PubMed]
  4. Steen, V.D.; Medsger, T.A. Changes in causes of death in systemic sclerosis, 1972–2002. Ann. Rheum. Dis. 2007, 66, 940–944. [Google Scholar] [CrossRef]
  5. Khanna, D.; Lin, C.J.F.; Furst, D.E.; Goldin, J.; Kim, G.; Kuwana, M.; Allanore, Y.; Matucci-Cerinic, M.; Distler, O.; Shima, Y.; et al. Tocilizumab in systemic sclerosis: A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir. Med. 2020, 8, 963–974. [Google Scholar] [CrossRef]
  6. Distler, O.; Highland, K.B.; Gahlemann, M.; Azuma, A.; Fischer, A.; Mayes, M.D.; Raghu, G.; Sauter, W.; Girard, M.; Alves, M.; et al. Nintedanib for Systemic Sclerosis–Associated Interstitial Lung Disease. New Engl. J. Med. 2019, 380, 2518–2528. [Google Scholar] [CrossRef]
  7. Cottin, V.; Brown, K.K. Interstitial lung disease associated with systemic sclerosis (SSc-ILD). Respir. Res. 2019, 20, 13. [Google Scholar] [CrossRef]
  8. Denton, C.P.; Wells, A.U.; Coghlan, J.G. Major lung complications of systemic sclerosis. Nat. Rev. Rheumatol. 2018, 14, 511–527. [Google Scholar] [CrossRef]
  9. Hsu, E.; Shi, H.; Jordan, R.M.; Lyons-Weiler, J.; Pilewski, J.M.; Feghali-Bostwick, C.A. Lung tissues in patients with systemic sclerosis have gene expression patterns unique to pulmonary fibrosis and pulmonary hypertension. Arthritis Rheum. 2011, 63, 783–794. [Google Scholar] [CrossRef]
  10. Renaud, L.; Da Silveira, W.A.; Takamura, N.; Hardiman, G.; Feghali-Bostwick, C. Prominence of IL6, IGF, TLR, and Bioenergetics Pathway Perturbation in Lung Tissues of Scleroderma Patients With Pulmonary Fibrosis. Front. Immunol. 2020, 11, 383. [Google Scholar] [CrossRef]
  11. Christmann, R.B.; Sampaio-Barros, P.; Stifano, G.; Borges, C.L.; De Carvalho, C.R.; Kairalla, R.; Parra, E.R.; Spira, A.; Simms, R.; Capellozzi, V.L.; et al. Association of Interferon- and Transforming Growth Factor β-Regulated Genes and Macrophage Activation With Systemic Sclerosis-Related Progressive Lung Fibrosis. Arthritis Rheumatol. 2014, 66, 714–725. [Google Scholar] [CrossRef] [PubMed]
  12. Garrett, S.M.; Baker Frost, D.; Feghali-Bostwick, C. The mighty fibroblast and its utility in scleroderma research. J. Scleroderma Relat. Disord. 2017, 2, 69–134. [Google Scholar] [CrossRef] [PubMed]
  13. Valenzi, E.; Bulik, M.; Tabib, T.; Morse, C.; Sembrat, J.; Bittar, H.T.; Rojas, M.; Lafyatis, R. Single-cell analysis reveals fibroblast heterogeneity and myofibroblasts in systemic sclerosis-associated interstitial lung disease. Ann. Rheum. Dis. 2019, 78, 1379–1387. [Google Scholar] [CrossRef]
  14. Mouawad, J.E.; Sharma, S.; Renaud, L.; Pilewski, J.M.; Nadig, S.N.; Feghali-Bostwick, C. Reduced Cathepsin L Expression and Secretion into the Extracellular Milieu Contribute to Lung Fibrosis in Systemic Sclerosis. Rheumatology 2022, keac411. [Google Scholar] [CrossRef]
  15. Kolodziejczyk, A.A.; Kim, J.K.; Svensson, V.; Marioni, J.C.; Teichmann, S.A. The Technology and Biology of Single-Cell RNA Sequencing. Mol. Cell 2015, 58, 610–620. [Google Scholar] [CrossRef] [PubMed]
  16. Reyfman, P.A.; Walter, J.M.; Joshi, N.; Anekalla, K.R.; McQuattie-Pimentel, A.C.; Chiu, S.; Fernandez, R.; Akbarpour, M.; Chen, C.I.; Ren, Z.; et al. Single-Cell Transcriptomic Analysis of Human Lung Provides Insights into the Pathobiology of Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2019, 199, 1517–1536. [Google Scholar] [CrossRef] [PubMed]
  17. Peyser, R.; MacDonnell, S.; Gao, Y.; Cheng, L.; Kim, Y.; Kaplan, T.; Ruan, Q.; Wei, Y.; Ni, M.; Adler, C.; et al. Defining the Activated Fibroblast Population in Lung Fibrosis Using Single-Cell Sequencing. Am. J. Respir. Cell Mol. Biol. 2019, 61, 74–85. [Google Scholar] [CrossRef] [PubMed]
  18. Massagué, J. TGFβ signalling in context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef]
  19. Meng, X.M.; Nikolic-Paterson, D.J.; Lan, H.Y. TGF-beta: The master regulator of fibrosis. Nat. Rev. Nephrol. 2016, 12, 325–338. [Google Scholar] [CrossRef]
  20. Rahimi, R.A.; Leof, E.B. TGF-β signaling: A tale of two responses. J. Cell. Biochem. 2007, 102, 593–608. [Google Scholar] [CrossRef]
  21. Massagué, J.; Gomis, R.R. The logic of TGFβ signaling. FEBS Lett. 2006, 580, 2811–2820. [Google Scholar] [CrossRef] [PubMed]
  22. Frangogiannis, N.G. Transforming growth factor–β in tissue fibrosis. J. Exp. Med. 2020, 217, e20190103. [Google Scholar] [CrossRef] [PubMed]
  23. Mack, M.; Yanagita, M. Origin of myofibroblasts and cellular events triggering fibrosis. Kidney Int. 2015, 87, 297–307. [Google Scholar] [CrossRef] [PubMed]
  24. Lafyatis, R. Transforming growth factor beta--at the centre of systemic sclerosis. Nat. Rev. Rheumatol. 2014, 10, 706–719. [Google Scholar] [CrossRef]
  25. Ayers, N.B.; Sun, C.-M.; Chen, S.-Y. Transforming growth factor-β signaling in systemic sclerosis. J. Biomed. Res. 2018, 32, 3–12. [Google Scholar] [CrossRef]
  26. Garrett, S.M.; Hsu, E.; Thomas, J.M.; Pilewski, J.M.; Feghali-Bostwick, C. Insulin-like growth factor (IGF)-II-mediated fibrosis in pathogenic lung conditions. PLoS ONE 2019, 14, e0225422. [Google Scholar] [CrossRef]
  27. Yao, Y.; Wang, Y.; Zhang, Z.; He, L.; Zhu, J.; Zhang, M.; He, X.; Cheng, Z.; Ao, Q.; Cao, Y.; et al. Chop Deficiency Protects Mice Against Bleomycin-induced Pulmonary Fibrosis by Attenuating M2 Macrophage Production. Mol. Ther. 2016, 24, 915–925. [Google Scholar] [CrossRef]
  28. Wang, Y.; Zhang, L.; Wu, G.R.; Zhou, Q.; Yue, H.; Rao, L.Z.; Yuan, T.; Mo, B.; Wang, F.X.; Chen, L.M.; et al. MBD2 serves as a viable target against pulmonary fibrosis by inhibiting macrophage M2 program. Sci. Adv. 2021, 7, eabb6075. [Google Scholar] [CrossRef]
  29. Zehender, A.; Li, Y.N.; Lin, N.Y.; Stefanica, A.; Nüchel, J.; Chen, C.W.; Hsu, H.H.; Zhu, H.; Ding, X.; Huang, J.; et al. TGFβ promotes fibrosis by MYST1-dependent epigenetic regulation of autophagy. Nat. Commun. 2021, 12, 4404. [Google Scholar] [CrossRef]
  30. Saito, A.; Horie, M.; Nagase, T. TGF-β Signaling in Lung Health and Disease. Int. J. Mol. Sci. 2018, 19, 2460. [Google Scholar] [CrossRef] [Green Version]
  31. Györfi, A.H.; Matei, A.E.; Distler, J.H.W. Targeting TGF-β signaling for the treatment of fibrosis. Matrix Biol. J. Int. Soc. Matrix Biol. 2018, 68–69, 8–27. [Google Scholar] [CrossRef]
  32. Miller, D.S.J.; Schmierer, B.; Hill, C.S. TGF-β family ligands exhibit distinct signalling dynamics that are driven by receptor localisation. J. Cell Sci. 2019, 132, jcs234039. [Google Scholar] [CrossRef]
  33. Klinkhammer, B.M.; Floege, J.; Boor, P. PDGF in organ fibrosis. Mol. Aspects Med. 2018, 62, 44–62. [Google Scholar] [CrossRef]
  34. Andrae, J.; Gallini, R.; Betsholtz, C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev. 2008, 22, 1276–1312. [Google Scholar] [CrossRef]
  35. Betsholtz, C. Biology of platelet-derived growth factors in development. Birth Defects Res. C Embryo Today 2003, 69, 272–285. [Google Scholar] [CrossRef]
  36. Takamura, N.; Renaud, L.; da Silveira, W.A.; Feghali-Bostwick, C. PDGF Promotes Dermal Fibroblast Activation via a Novel Mechanism Mediated by Signaling Through MCHR1. Front. Immunol. 2021, 12, 745308. [Google Scholar] [CrossRef]
  37. Ludwicka, A.; Ohba, T.; Trojanowska, M.; Yamakage, A.; Strange, C.; Smith, E.A.; Leroy, E.C.; Sutherland, S.; Silver, R.M. Elevated levels of platelet derived growth factor and transforming growth factor-beta 1 in bronchoalveolar lavage fluid from patients with scleroderma. J. Rheumatol. 1995, 22, 1876–1883. [Google Scholar]
  38. Trojanowska, M. Role of PDGF in fibrotic diseases and systemic sclerosis. Rheumatology 2008, 47, v2–v4. [Google Scholar] [CrossRef]
  39. Tanaka, S.; Suto, A.; Ikeda, K.; Sanayama, Y.; Nakagomi, D.; Iwamoto, T.; Suzuki, K.; Kambe, N.; Matsue, H.; Matsumura, R.; et al. Alteration of circulating miRNAs in SSc: miR-30b regulates the expression of PDGF receptor β. Rheumatology 2013, 52, 1963–1972. [Google Scholar] [CrossRef]
  40. Moroncini, G.; Grieco, A.; Nacci, G.; Paolini, C.; Tonnini, C.; Pozniak, C.; Cuccioloni, M.; Mozzicafreddo, M.; Svegliati, S.; Angeletti, M. Epitope specificity determines pathogenicity and detectability of anti-PDGFRα autoantibodies in systemic sclerosis. Arthritis Rheumatol. 2015, 67, 1891–1903. [Google Scholar] [CrossRef]
  41. Gabrielli, A.; Svegliati, S.; Moroncini, G.; Luchetti, M.; Tonnini, C.; Avvedimento, E.V. Stimulatory autoantibodies to the PDGF receptor: A link to fibrosis in scleroderma and a pathway for novel therapeutic targets. Autoimmun. Rev. 2007, 7, 121–126. [Google Scholar] [CrossRef] [PubMed]
  42. Svegliati, S.; Amico, D.; Spadoni, T.; Fischetti, C.; Finke, D.; Moroncini, G.; Paolini, C.; Tonnini, C.; Grieco, A.; Rovinelli, M.; et al. Agonistic Anti-PDGF Receptor Autoantibodies from Patients with Systemic Sclerosis Impact Human Pulmonary Artery Smooth Muscle Cells Function In Vitro. Front. Immunol. 2017, 8, 75. [Google Scholar] [CrossRef] [PubMed]
  43. Paolini, C.; Agarbati, S.; Benfaremo, D.; Mozzicafreddo, M.; Svegliati, S.; Moroncini, G. PDGF/PDGFR: A Possible Molecular Target in Scleroderma Fibrosis. Int. J. Mol. Sci. 2022, 23, 3904. [Google Scholar] [CrossRef] [PubMed]
  44. Wollin, L.; Maillet, I.; Quesniaux, V.; Holweg, A.; Ryffel, B. Antifibrotic and anti-inflammatory activity of the tyrosine kinase inhibitor nintedanib in experimental models of lung fibrosis. J. Pharmacol. Exp. Ther. 2014, 349, 209–220. [Google Scholar] [CrossRef] [PubMed]
  45. Ornitz, D.M.; Itoh, N. The Fibroblast Growth Factor signaling pathway. WIREs Dev. Biol. 2015, 4, 215–266. [Google Scholar] [CrossRef]
  46. Olsen, S.K.; Garbi, M.; Zampieri, N.; Eliseenkova, A.V.; Ornitz, D.M.; Goldfarb, M.; Mohammadi, M. Fibroblast growth factor (FGF) homologous factors share structural but not functional homology with FGFs. J. Biol. Chem. 2003, 278, 34226–34236. [Google Scholar] [CrossRef]
  47. Sun, C.; Tian, X.; Jia, Y.; Yang, M.; Li, Y.; Fernig, D.G. Functions of exogenous FGF signals in regulation of fibroblast to myofibroblast differentiation and extracellular matrix protein expression. Open. Biol. 2022, 12, 210356. [Google Scholar] [CrossRef]
  48. de Araújo, R.; Lôbo, M.; Trindade, K.; Silva, D.F.; Pereira, N. Fibroblast Growth Factors: A Controlling Mechanism of Skin Aging. Ski. Pharmacol. Physiol. 2019, 32, 275–282. [Google Scholar] [CrossRef]
  49. Justet, A.; Ghanem, M.; Boghanim, T.; Hachem, M.; Vasarmidi, E.; Jaillet, M.; Vadel, A.; Joannes, A.; Mordant, P.; Bonniaud, P.; et al. FGF19 Is Downregulated in Idiopathic Pulmonary Fibrosis and Inhibits Lung Fibrosis in Mice. Am. J. Respir. Cell Mol. Biol. 2022, 67, 173–187. [Google Scholar] [CrossRef]
  50. Chakraborty, D.; Zhu, H.; Jüngel, A.; Summa, L.; Li, Y.-N.; Matei, A.-E.; Zhou, X.; Huang, J.; Trinh-Minh, T.; Chen, C.-W.; et al. Fibroblast growth factor receptor 3 activates a network of profibrotic signaling pathways to promote fibrosis in systemic sclerosis. Sci. Transl. Med. 2020, 12, eaaz5506. [Google Scholar] [CrossRef]
  51. Duspara, K.; Bojanic, K.; Pejic, J.I.; Kuna, L.; Kolaric, T.O.; Nincevic, V.; Smolic, R.; Vcev, A.; Glasnovic, M.; Curcic, I.B.; et al. Targeting the Wnt Signaling Pathway in Liver Fibrosis for Drug Options: An Update. J. Clin. Transl. Hepatol. 2021, 9, 960–971. [Google Scholar] [CrossRef]
  52. Griffin, M.F.; Huber, J.; Evan, F.J.; Quarto, N.; Longaker, M.T. The role of Wnt signaling in skin fibrosis. Med. Res. Rev. 2022, 42, 615–628. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, T.; Gonzalez De Los Santos, F.; Hirsch, M.; Wu, Z.; Phan, S.H. Noncanonical Wnt Signaling Promotes Myofibroblast Differentiation in Pulmonary Fibrosis. Am. J. Respir. Cell Mol. Biol. 2021, 65, 489–499. [Google Scholar] [CrossRef] [PubMed]
  54. Methatham, T.; Tomida, S.; Kimura, N.; Imai, Y.; Aizawa, K. Inhibition of the canonical Wnt signaling pathway by a β-catenin/CBP inhibitor prevents heart failure by ameliorating cardiac hypertrophy and fibrosis. Sci. Rep. 2021, 11, 14886. [Google Scholar] [CrossRef]
  55. Majidinia, M.; Aghazadeh, J.; Jahanban-Esfahlani, R.; Yousefi, B. The roles of Wnt/β-catenin pathway in tissue development and regenerative medicine. J. Cell. Physiol. 2018, 233, 5598–5612. [Google Scholar] [CrossRef] [PubMed]
  56. Akhmetshina, A.; Palumbo, K.; Dees, C.; Bergmann, C.; Venalis, P.; Zerr, P.; Horn, A.; Kireva, T.; Beyer, C.; Zwerina, J.; et al. Activation of canonical Wnt signalling is required for TGF-β-mediated fibrosis. Nat. Commun. 2012, 3, 735. [Google Scholar] [CrossRef]
  57. Lam, A.P.; Flozak, A.S.; Russell, S.; Wei, J.; Jain, M.; Mutlu, G.M.; Budinger, G.R.; Feghali-Bostwick, C.A.; Varga, J.; Gottardi, C.J. Nuclear β-catenin is increased in systemic sclerosis pulmonary fibrosis and promotes lung fibroblast migration and proliferation. Am. J. Respir. Cell Mol. Biol. 2011, 45, 915–922. [Google Scholar] [CrossRef]
  58. Ulsamer, A.; Wei, Y.; Kim, K.K.; Tan, K.; Wheeler, S.; Xi, Y.; Thies, R.S.; Chapman, H.A. Axin pathway activity regulates in vivo pY654-β-catenin accumulation and pulmonary fibrosis. J. Biol. Chem. 2012, 287, 5164–5172. [Google Scholar] [CrossRef]
  59. Bergmann, C.; Distler, J.H.W. Canonical Wnt signaling in systemic sclerosis. Lab. Investig. 2016, 96, 151–155. [Google Scholar] [CrossRef]
  60. Nollet, M.; Bachelier, R.; Joshkon, A.; Traboulsi, W.; Mahieux, A.; Moyon, A.; Muller, A.; Somasundaram, I.; Simoncini, S.; Peiretti, F.; et al. Involvement of Multiple Variants of Soluble CD146 in Systemic Sclerosis: Identification of a Novel Profibrotic Factor. Arthritis Rheumatol. 2022, 74, 1027–1038. [Google Scholar] [CrossRef]
  61. Beyer, C.; Schramm, A.; Akhmetshina, A.; Dees, C.; Kireva, T.; Gelse, K.; Sonnylal, S.; de Crombrugghe, B.; Taketo, M.M.; Distler, O.; et al. β-catenin is a central mediator of pro-fibrotic Wnt signaling in systemic sclerosis. Ann. Rheum. Dis. 2012, 71, 761–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Akdis, M.; Aab, A.; Altunbulakli, C.; Azkur, K.; Costa, R.A.; Crameri, R.; Duan, S.; Eiwegger, T.; Eljaszewicz, A.; Ferstl, R.; et al. Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: Receptors, functions, and roles in diseases. J. Allergy Clin. Immunol. 2016, 138, 984–1010. [Google Scholar] [CrossRef] [PubMed]
  63. Brocker, C.; Thompson, D.; Matsumoto, A.; Nebert, D.W.; Vasiliou, V. Evolutionary divergence and functions of the human interleukin (IL) gene family. Hum. Genom. 2010, 5, 30–55. [Google Scholar] [CrossRef] [PubMed]
  64. Vaillant, J.A.A.; Qurie, A. Interleukin; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  65. She, Y.X.; Yu, Q.Y.; Tang, X.X. Role of interleukins in the pathogenesis of pulmonary fibrosis. Cell Death Discov. 2021, 7, 52. [Google Scholar] [CrossRef]
  66. Feghali, C.A.; Bost, K.L.; Boulware, D.W.; Levy, L.S. Control of Il-6 Expression and Response in Fibroblasts from Patients with Systemic Sclerosis. Autoimmunity 1994, 17, 309–318. [Google Scholar] [CrossRef]
  67. Hasegawa, M.; Sato, S.; Fujimoto, M.; Ihn, H.; Kikuchi, K.; Takehara, K. Serum levels of interleukin 6 (IL-6), oncostatin M, soluble IL-6 receptor, and soluble gp130 in patients with systemic sclerosis. J. Rheumatol. 1998, 25, 308–313. [Google Scholar] [PubMed]
  68. Sato, S.; Hasegawa, M.; Takehara, K. Serum levels of interleukin-6 and interleukin-10 correlate with total skin thickness score in patients with systemic sclerosis. J. Dermatol. Sci. 2001, 27, 140–146. [Google Scholar] [CrossRef]
  69. Feghali, C.A.; Bost, K.L.; Boulware, D.W.; Levy, L.S. Mechanisms of pathogenesis in scleroderma. I. Overproduction of interleukin 6 by fibroblasts cultured from affected skin sites of patients with scleroderma. J. Rheumatol. 1992, 19, 1207–1211. [Google Scholar]
  70. Yousif, M.; Habib, R.; Esaely, H.; Yasin, R.; Sonbol, A. Interleukin-6 in systemic sclerosis and potential correlation with pulmonary involvement. Egypt. J. Chest Dis. Tuberc. 2015, 64, 237–241. [Google Scholar] [CrossRef]
  71. Nguyen, X.X.; Nishimoto, T.; Takihara, T.; Mlakar, L.; Bradshaw, A.D.; Feghali-Bostwick, C. Lysyl oxidase directly contributes to extracellular matrix production and fibrosis in systemic sclerosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2021, 320, L29–L40. [Google Scholar] [CrossRef]
  72. Roofeh, D.; Lin, C.J.F.; Goldin, J.; Kim, G.H.; Furst, D.E.; Denton, C.P.; Huang, S.; Khanna, D. Tocilizumab Prevents Progression of Early Systemic Sclerosis-Associated Interstitial Lung Disease. Arthritis Rheumatol. 2021, 73, 1301–1310. [Google Scholar] [CrossRef]
  73. Khanna, D.; Lin, C.J.F.; Furst, D.E.; Wagner, B.; Zucchetto, M.; Raghu, G.; Martinez, F.J.; Goldin, J.; Siegel, J.; Denton, C.P. Long-Term Safety and Efficacy of Tocilizumab in Early Systemic Sclerosis-Interstitial Lung Disease: Open-Label Extension of a Phase 3 Randomized Controlled Trial. Am. J. Respir. Crit. Care Med. 2022, 205, 674–684. [Google Scholar] [CrossRef]
  74. Yaseen, B.; Lopez, H.; Taki, Z.; Zafar, S.; Rosario, H.; Abdi, B.A.; Vigneswaran, S.; Xing, F.; Arumalla, N.; Black, S.; et al. Interleukin-31 promotes pathogenic mechanisms underlying skin and lung fibrosis in scleroderma. Rheumatology 2020, 59, 2625–2636. [Google Scholar] [CrossRef]
  75. Lindahl, G.E.; Stock, C.J.; Shi-Wen, X.; Leoni, P.; Sestini, P.; Howat, S.L.; Bou-Gharios, G.; Nicholson, A.G.; Denton, C.P.; Grutters, J.C.; et al. Microarray profiling reveals suppressed interferon stimulated gene program in fibroblasts from scleroderma-associated interstitial lung disease. Respir. Res. 2013, 14, 80. [Google Scholar] [CrossRef]
  76. Cook, S.A.; Schafer, S. Hiding in Plain Sight: Interleukin-11 Emerges as a Master Regulator of Fibrosis, Tissue Integrity, and Stromal Inflammation. Annu. Rev. Med. 2020, 71, 263–276. [Google Scholar] [CrossRef]
  77. Ng, B.; Dong, J.; Viswanathan, S.; Widjaja, A.A.; Paleja, B.S.; Adami, E.; Ko, N.S.J.; Wang, M.; Lim, S.; Tan, J.; et al. Fibroblast-specific IL11 signaling is required for lung fibrosis and inflammation. FASEB J. 2020, 34, 11802–11815. [Google Scholar] [CrossRef]
  78. Artlett, C.M.; Sassi-Gaha, S.; Hope, J.L.; Feghali-Bostwick, C.A.; Katsikis, P.D. Mir-155 is overexpressed in systemic sclerosis fibroblasts and is required for NLRP3 inflammasome-mediated collagen synthesis during fibrosis. Arthritis Res. Ther. 2017, 19, 144. [Google Scholar] [CrossRef]
  79. Ahmed, S.; Misra, D.P.; Agarwal, V. Interleukin-17 pathways in systemic sclerosis-associated fibrosis. Rheumatol. Int. 2019, 39, 1135–1143. [Google Scholar] [CrossRef]
  80. Kuzumi, A.; Yoshizaki, A.; Toyama, S.; Fukasawa, T.; Ebata, S.; Nakamura, K.; Yamashita, T.; Saigusa, R.; Miura, S.; Hirabayashi, M.; et al. Serum interleukin-34 levels in patients with systemic sclerosis: Clinical association with interstitial lung disease. J. Dermatol. 2018, 45, 1216–1220. [Google Scholar] [CrossRef]
  81. Versace, A.G.; Bitto, A.; Ioppolo, C.; Aragona, C.O.; La Rosa, D.; Roberts, W.N.; D′Angelo, T.; Cinquegrani, A.; Cirmi, S.; Irrera, N.; et al. IL-13 and IL-33 Serum Levels Are Increased in Systemic Sclerosis Patients With Interstitial Lung Disease. Front. Med. 2022, 9, 825567. [Google Scholar] [CrossRef]
  82. Wakhlu, A.; Sahoo, R.; Parida, J.; Rai, M.; Misra, D.; Agrawal, V.; Agarwal, V. Serum Interleukin-6, Interleukin-17A, and transforming growth factor beta are raised in systemic sclerosis with interstitial lung disease. Indian J. Rheumatol. 2018, 13, 107–112. [Google Scholar] [CrossRef]
  83. Harrison, N.K.; Cambrey, A.D.; Myers, A.R.; Southcott, A.M.; Black, C.M.; du Bois, R.M.; Laurent, G.J.; McAnulty, R.J. Insulin—Like Growth Factor-1 is Partially Responsible for Fibroblast Proliferation Induced by Bronchoalveolar Lavage Fluid from Patients with Systemic Sclerosis. Clin. Sci. 1994, 86, 141–148. [Google Scholar] [CrossRef] [PubMed]
  84. Hsu, E.; Feghali-Bostwick, C.A. Insulin-like growth factor-II is increased in systemic sclerosis-associated pulmonary fibrosis and contributes to the fibrotic process via Jun N-terminal kinase- and phosphatidylinositol-3 kinase-dependent pathways. Am. J. Pathol. 2008, 172, 1580–1590. [Google Scholar] [CrossRef] [PubMed]
  85. Pilewski, J.M.; Liu, L.; Henry, A.C.; Knauer, A.V.; Feghali-Bostwick, C.A. Insulin-Like Growth Factor Binding Proteins 3 and 5 Are Overexpressed in Idiopathic Pulmonary Fibrosis and Contribute to Extracellular Matrix Deposition. Am. J. Pathol. 2005, 166, 399–407. [Google Scholar] [CrossRef]
  86. Brissett, M.; Veraldi, K.L.; Pilewski, J.M.; Medsger, T.A., Jr.; Feghali-Bostwick, C.A. Localized expression of tenascin in systemic sclerosis-associated pulmonary fibrosis and its regulation by insulin-like growth factor binding protein 3. Arthritis Rheum. 2012, 64, 272–280. [Google Scholar] [CrossRef]
  87. Ruiz, X.D.; Mlakar, L.R.; Yamaguchi, Y.; Su, Y.; Larregina, A.T.; Pilewski, J.M.; Feghali-Bostwick, C.A. Syndecan-2 is a novel target of insulin-like growth factor binding protein-3 and is over-expressed in fibrosis. PLoS ONE 2012, 7, e43049. [Google Scholar] [CrossRef]
  88. Nguyen, X.X.; Muhammad, L.; Nietert, P.J.; Feghali-Bostwick, C. IGFBP-5 Promotes Fibrosis via Increasing Its Own Expression and That of Other Pro-fibrotic Mediators. Front. Endocrinol. 2018, 9, 601. [Google Scholar] [CrossRef]
  89. Yasuoka, H.; Yamaguchi, Y.; Feghali-Bostwick, C.A. The Pro-Fibrotic Factor IGFBP-5 Induces Lung Fibroblast and Mononuclear Cell Migration. Am. J. Respir. Cell Mol. Biol. 2009, 41, 179–188. [Google Scholar] [CrossRef]
  90. Nguyen, X.X.; Sanderson, M.; Helke, K.; Feghali-Bostwick, C. Phenotypic Characterization of Transgenic Mice Expressing Human IGFBP-5. Int. J. Mol. Sci. 2020, 22, 335. [Google Scholar] [CrossRef]
  91. Guiot, J.; Njock, M.S.; André, B.; Gester, F.; Henket, M.; De Seny, D.; Moermans, C.; Malaise, M.G.; Louis, R. Serum IGFBP-2 in systemic sclerosis as a prognostic factor of lung dysfunction. Sci. Rep. 2021, 11, 10882. [Google Scholar] [CrossRef]
  92. Zhao, B.; Li, L.; Lei, Q.; Guan, K.L. The Hippo-YAP pathway in organ size control and tumorigenesis: An updated version. Genes Dev. 2010, 24, 862–874. [Google Scholar] [CrossRef] [PubMed]
  93. Kwon, H.; Kim, J.; Jho, E.-H. Role of the Hippo pathway and mechanisms for controlling cellular localization of YAP/TAZ. FEBS J. 2022, 289, 5798–5818. [Google Scholar] [CrossRef] [PubMed]
  94. Mauviel, A.; Nallet-Staub, F.; Varelas, X. Integrating developmental signals: A Hippo in the (path)way. Oncogene 2012, 31, 1743–1756. [Google Scholar] [CrossRef] [PubMed]
  95. Liu, F.; Lagares, D.; Choi, K.M.; Stopfer, L.; Marinković, A.; Vrbanac, V.; Probst, C.K.; Hiemer, S.E.; Sisson, T.H.; Horowitz, J.C.; et al. Mechanosignaling through YAP and TAZ drives fibroblast activation and fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 308, L344–L357. [Google Scholar] [CrossRef] [PubMed]
  96. He, X.; Tolosa, M.F.; Zhang, T.; Goru, S.K.; Ulloa Severino, L.; Misra, P.S.; McEvoy, C.M.; Caldwell, L.; Szeto, S.G.; Gao, F.; et al. Myofibroblast YAP/TAZ activation is a key step in organ fibrogenesis. JCI Insight 2022, 7, e146243. [Google Scholar] [CrossRef] [PubMed]
  97. Toyama, T.; Looney, A.P.; Baker, B.M.; Stawski, L.; Haines, P.; Simms, R.; Szymaniak, A.D.; Varelas, X.; Trojanowska, M. Therapeutic Targeting of TAZ and YAP by Dimethyl Fumarate in Systemic Sclerosis Fibrosis. J. Investig. Dermatol. 2018, 138, 78–88. [Google Scholar] [CrossRef]
  98. Wu, D.; Wang, W.; Li, X.; Yin, B.; Ma, Y. Single-cell sequencing reveals the antifibrotic effects of YAP/TAZ in systemic sclerosis. Int. J. Biochem. Cell Biol. 2022, 149, 106257. [Google Scholar] [CrossRef]
  99. Rot, A.; von Andrian, U.H. Chemokines in innate and adaptive host defense: Basic chemokinese grammar for immune cells. Annu. Rev. Immunol. 2004, 22, 891–928. [Google Scholar] [CrossRef]
  100. Codullo, V.; Baldwin, H.M.; Singh, M.D.; Fraser, A.R.; Wilson, C.; Gilmour, A.; Hueber, A.J.; Bonino, C.; McInnes, I.B.; Montecucco, C.; et al. An investigation of the inflammatory cytokine and chemokine network in systemic sclerosis. Ann. Rheum. Dis. 2011, 70, 1115. [Google Scholar] [CrossRef]
  101. Schmidt, K.; Martinez-Gamboa, L.; Meier, S.; Witt, C.; Meisel, C.; Hanitsch, L.G.; Becker, M.O.; Huscher, D.; Burmester, G.R.; Riemekasten, G. 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. [Google Scholar] [CrossRef]
  102. Assassi, S.; Wu, M.; Tan, F.K.; Chang, J.; Graham, T.A.; Furst, D.E.; Khanna, D.; Charles, J.; Ferguson, E.C.; Feghali-Bostwick, C.; et al. Skin gene expression correlates of severity of interstitial lung disease in systemic sclerosis. Arthritis Rheum. 2013, 65, 2917–2927. [Google Scholar] [CrossRef] [PubMed]
  103. Wu, M.; Baron, M.; Pedroza, C.; Salazar, G.A.; Ying, J.; Charles, J.; Agarwal, S.K.; Hudson, M.; Pope, J.; Zhou, X.; et al. CCL2 in the Circulation Predicts Long-Term Progression of Interstitial Lung Disease in Patients With Early Systemic Sclerosis: Data From Two Independent Cohorts. Arthritis Rheumatol. 2017, 69, 1871–1878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. van Bon, L.; Affandi, A.J.; Broen, J.; Christmann, R.B.; Marijnissen, R.J.; Stawski, L.; Farina, G.A.; Stifano, G.; Mathes, A.L.; Cossu, M.; et al. Proteome-wide Analysis and CXCL4 as a Biomarker in Systemic Sclerosis. N. Engl. J. Med. 2013, 370, 433–443. [Google Scholar] [CrossRef]
  105. Affandi, A.J.; Carvalheiro, T.; Ottria, A.; de Haan, J.J.; Brans, M.A.D.; Brandt, M.M.; Tieland, R.G.; Lopes, A.P.; Fernández, B.M.; Bekker, C.P.J.; et al. CXCL4 drives fibrosis by promoting several key cellular and molecular processes. Cell Rep. 2022, 38, 110189. [Google Scholar] [CrossRef] [PubMed]
  106. Weigold, F.; Günther, J.; Pfeiffenberger, M.; Cabral-Marques, O.; Siegert, E.; Dragun, D.; Philippe, A.; Regensburger, A.K.; Recke, A.; Yu, X.; et al. Antibodies against chemokine receptors CXCR3 and CXCR4 predict progressive deterioration of lung function in patients with systemic sclerosis. Arthritis Res. Ther. 2018, 20, 52. [Google Scholar] [CrossRef] [PubMed]
  107. Elhai, M.; Hoffmann-Vold, A.M.; Avouac, J.; Pezet, S.; Cauvet, A.; Leblond, A.; Fretheim, H.; Garen, T.; Kuwana, M.; Molberg, Ø.; et al. Performance of Candidate Serum Biomarkers for Systemic Sclerosis-Associated Interstitial Lung Disease. Arthritis Rheumatol. 2019, 71, 972–982. [Google Scholar] [CrossRef]
  108. Yadati, T.; Houben, T.; Bitorina, A.; Shiri-Sverdlov, R. The Ins and Outs of Cathepsins: Physiological Function and Role in Disease Management. Cells 2020, 9, 1679. [Google Scholar] [CrossRef]
  109. Vidak, E.; Javoršek, U.; Vizovišek, M.; Turk, B. Cysteine Cathepsins and their Extracellular Roles: Shaping the Microenvironment. Cells 2019, 8, 264. [Google Scholar] [CrossRef]
  110. Vizovišek, M.; Fonović, M.; Turk, B. Cysteine cathepsins in extracellular matrix remodeling: Extracellular matrix degradation and beyond. Matrix Biol. J. Int. Soc. Matrix Biol. 2019, 75–76, 141–159. [Google Scholar] [CrossRef]
  111. Toyama, S.; Yamashita, T.; Saigusa, R.; Miura, S.; Nakamura, K.; Hirabayashi, M.; Miyagawa, T.; Fukui, Y.; Omatsu, J.; Yoshizaki, A.; et al. Decreased serum cathepsin S levels in patients with systemic sclerosis-associated interstitial lung disease. J. Dermatol. 2020, 47, 1027–1032. [Google Scholar] [CrossRef]
  112. Yamaguchi, Y.; Takihara, T.; Chambers, R.A.; Veraldi, K.L.; Larregina, A.T.; Feghali-Bostwick, C.A. A peptide derived from endostatin ameliorates organ fibrosis. Sci. Transl. Med. 2012, 4, 136ra171. [Google Scholar] [CrossRef]
  113. Gigante, A.; Margiotta, D.; Navarini, L.; Barbano, B.; Gasperini, M.L.; D′Agostino, C.; Amoroso, A.; Afeltra, A.; Rosato, E. Serum level of endostatin and digital ulcers in systemic sclerosis patients. Int. Wound J. 2018, 15, 424–428. [Google Scholar] [CrossRef]
  114. Richter, A.G.; McKeown, S.; Rathinam, S.; Harper, L.; Rajesh, P.; McAuley, D.F.; Heljasvaara, R.; Thickett, D.R. Soluble endostatin is a novel inhibitor of epithelial repair in idiopathic pulmonary fibrosis. Thorax 2009, 64, 156–161. [Google Scholar] [CrossRef]
  115. de Almeida, L.G.; Thode, H.; Eslambolchi, Y.; Chopra, S.; Young, D.; Gill, S.; Devel, L.; Dufour, A. Matrix metalloproteinases: From molecular mechanisms to physiology, pathophysiology, and pharmacology. Pharmacol. Rev. 2022, 74, 714–770. [Google Scholar]
  116. Jara, P.; Calyeca, J.; Romero, Y.; Plácido, L.; Yu, G.; Kaminski, N.; Maldonado, V.; Cisneros, J.; Selman, M.; Pardo, A. Matrix metalloproteinase (MMP)-19-deficient fibroblasts display a profibrotic phenotype. Am. J. Physiology. Lung Cell. Mol. Physiol. 2015, 308, L511–L522. [Google Scholar] [CrossRef]
  117. Yu, G.; Kovkarova-Naumovski, E.; Jara, P.; Parwani, A.; Kass, D.; Ruiz, V.; Lopez-Otín, C.; Rosas, I.O.; Gibson, K.F.; Cabrera, S.; et al. Matrix metalloproteinase-19 is a key regulator of lung fibrosis in mice and humans. Am. J. Respir. Crit. Care Med. 2012, 186, 752–762. [Google Scholar] [CrossRef]
  118. Sharma, S.; Watanabe, T.; Nishimoto, T.; Takihara, T.; Mlakar, L.; Nguyen, X.-X.; Sanderson, M.; Su, Y.; Chambers, R.A.; Feghali-Bostwick, C. E4 engages uPAR and enolase-1 and activates urokinase to exert antifibrotic effects. JCI Insight 2021, 6, e144935. [Google Scholar] [CrossRef]
  119. Chu, H.; Jiang, S.; Liu, Q.; Ma, Y.; Zhu, X.; Liang, M.; Shi, X.; Ding, W.; Zhou, X.; Zou, H.; et al. Sirtuin1 Protects against Systemic Sclerosis–related Pulmonary Fibrosis by Decreasing Proinflammatory and Profibrotic Processes. Am. J. Respir. Cell Mol. Biol. 2017, 58, 28–39. [Google Scholar] [CrossRef]
  120. Akamata, K.; Wei, J.; Bhattacharyya, M.; Cheresh, P.; Bonner, M.Y.; Arbiser, J.L.; Raparia, K.; Gupta, M.P.; Kamp, D.W.; Varga, J. SIRT3 is attenuated in systemic sclerosis skin and lungs, and its pharmacologic activation mitigates organ fibrosis. Oncotarget 2016, 7, 69321–69336. [Google Scholar] [CrossRef]
  121. Manetti, M.; Rosa, I.; Fioretto, B.S.; Matucci-Cerinic, M.; Romano, E. Decreased Serum Levels of SIRT1 and SIRT3 Correlate with Severity of Skin and Lung Fibrosis and Peripheral Microvasculopathy in Systemic Sclerosis. J. Clin. Med. 2022, 11, 1362. [Google Scholar] [CrossRef]
  122. Wyman, A.E.; Noor, Z.; Fishelevich, R.; Lockatell, V.; Shah, N.G.; Todd, N.W.; Atamas, S.P. Sirtuin 7 is decreased in pulmonary fibrosis and regulates the fibrotic phenotype of lung fibroblasts. Am. J. Physiology. Lung Cell. Mol. Physiol. 2017, 312, L945–L958. [Google Scholar] [CrossRef]
  123. Su, Y.; Nishimoto, T.; Hoffman, S.; Nguyen, X.-X.; Pilewski, J.M.; Feghali-Bostwick, C. Insulin-like growth factor binding protein-4 exerts antifibrotic activity by reducing levels of connective tissue growth factor and the C-X-C chemokine receptor 4. FASEB Bioadvances 2019, 1, 167–179. [Google Scholar] [CrossRef]
  124. Zhou, J.; Yi, Z.; Fu, Q. Dynamic decreased expression and hypermethylation of secreted frizzled-related protein 1 and 4 over the course of pulmonary fibrosis in mice. Life Sci. 2019, 218, 241–252. [Google Scholar] [CrossRef]
  125. Dees, C.; Schlottmann, I.; Funke, R.; Distler, A.; Palumbo-Zerr, K.; Zerr, P.; Lin, N.-Y.; Beyer, C.; Distler, O.; Schett, G.; et al. The Wnt antagonists DKK1 and SFRP1 are downregulated by promoter hypermethylation in systemic sclerosis. Ann. Rheum. Dis. 2014, 73, 1232–1239. [Google Scholar] [CrossRef]
  126. Henderson, J.; Brown, M.; Horsburgh, S.; Duffy, L.; Wilkinson, S.; Worrell, J.; Stratton, R.; O’Reilly, S. Methyl cap binding protein 2: A key epigenetic protein in systemic sclerosis. Rheumatology 2018, 58, 527–535. [Google Scholar] [CrossRef]
  127. Castro-Piedras, I.; Vartak, D.; Sharma, M.; Pandey, S.; Casas, L.; Molehin, D.; Rasha, F.; Fokar, M.; Nichols, J.; Almodovar, S.; et al. Identification of Novel MeCP2 Cancer-Associated Target Genes and Post-Translational Modifications. Front. Oncol. 2020, 10, 576362. [Google Scholar] [CrossRef]
  128. Henderson, J.; Wilkinson, S.; Przyborski, S.; Stratton, R.; O’Reilly, S. microRNA27a-3p mediates reduction of the Wnt antagonist sFRP-1 in systemic sclerosis. Epigenetics 2021, 16, 808–817. [Google Scholar] [CrossRef]
  129. Williams, L.; Layton, T.; Yang, N.; Feldmann, M.; Nanchahal, J. Collagen VI as a driver and disease biomarker in human fibrosis. Febs J. 2022, 289, 3603–3629. [Google Scholar] [CrossRef]
  130. De Castro Brás, L.E.; Frangogiannis, N.G. Extracellular matrix-derived peptides in tissue remodeling and fibrosis. Matrix Biol. 2020, 91–92, 176–187. [Google Scholar] [CrossRef]
Figure 1. Lung fibrosis in SSc is the result of an imbalance between anti-fibrotic and pro-fibrotic factors, favoring the latter, and resulting in the activation of fibroblasts and the excessive deposition of ECM. ES, endostatin; MMP, matrix metalloproteinase; CTSL, Cathepsin L; TGFβ, Transforming Growth Factor Beta; PDGF, Platelet-Derived Growth Factor; WNT, Wingless-Related Integration Site; ECM, extracellular matrix; SSc, systemic sclerosis.
Figure 1. Lung fibrosis in SSc is the result of an imbalance between anti-fibrotic and pro-fibrotic factors, favoring the latter, and resulting in the activation of fibroblasts and the excessive deposition of ECM. ES, endostatin; MMP, matrix metalloproteinase; CTSL, Cathepsin L; TGFβ, Transforming Growth Factor Beta; PDGF, Platelet-Derived Growth Factor; WNT, Wingless-Related Integration Site; ECM, extracellular matrix; SSc, systemic sclerosis.
Ijms 24 02963 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mouawad, J.E.; Feghali-Bostwick, C. The Molecular Mechanisms of Systemic Sclerosis-Associated Lung Fibrosis. Int. J. Mol. Sci. 2023, 24, 2963. https://doi.org/10.3390/ijms24032963

AMA Style

Mouawad JE, Feghali-Bostwick C. The Molecular Mechanisms of Systemic Sclerosis-Associated Lung Fibrosis. International Journal of Molecular Sciences. 2023; 24(3):2963. https://doi.org/10.3390/ijms24032963

Chicago/Turabian Style

Mouawad, Joe E., and Carol Feghali-Bostwick. 2023. "The Molecular Mechanisms of Systemic Sclerosis-Associated Lung Fibrosis" International Journal of Molecular Sciences 24, no. 3: 2963. https://doi.org/10.3390/ijms24032963

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop