Next Article in Journal
Advances in Oocyte Maturation In Vivo and In Vitro in Mammals
Previous Article in Journal
Investigating Mammalian Formins with SMIFH2 Fifteen Years in: Novel Targets and Unexpected Biology
Previous Article in Special Issue
Anti-Spike Antibodies Present in the Milk of SARS-CoV-2 Vaccinated Mothers Are Complement-Activating
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Towards a Unified Approach in Autoimmune Fibrotic Signalling Pathways

Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Piazza Giulio Cesare 1, I-70124 Bari, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(10), 9060; https://doi.org/10.3390/ijms24109060
Submission received: 7 April 2023 / Revised: 12 May 2023 / Accepted: 19 May 2023 / Published: 21 May 2023
(This article belongs to the Special Issue State-of-the-Art Molecular Immunology in Italy)

Abstract

:
Autoimmunity is a chronic process resulting in inflammation, tissue damage, and subsequent tissue remodelling and organ fibrosis. In contrast to acute inflammatory reactions, pathogenic fibrosis typically results from the chronic inflammatory reactions characterizing autoimmune diseases. Despite having obvious aetiological and clinical outcome distinctions, most chronic autoimmune fibrotic disorders have in common a persistent and sustained production of growth factors, proteolytic enzymes, angiogenic factors, and fibrogenic cytokines, which together stimulate the deposition of connective tissue elements or epithelial to mesenchymal transformation (EMT) that progressively remodels and destroys normal tissue architecture leading to organ failure. Despite its enormous impact on human health, there are currently no approved treatments that directly target the molecular mechanisms of fibrosis. The primary goal of this review is to discuss the most recent identified mechanisms of chronic autoimmune diseases characterized by a fibrotic evolution with the aim to identify possible common and unique mechanisms of fibrogenesis that might be exploited in the development of effective antifibrotic therapies.

1. Introduction

The pathophysiology of autoimmune diseases involves an unbalanced interplay between the innate and adaptive immunity, culminating in immune responses mounted against self-antigens [1]. All autoimmune diseases are believed to go through sequential initiation, effector, and resolution phases. In contrast to normal inflammation, in autoimmune diseases there is sustained cellular activation, resulting in chronic inflammation. Recently, increasing evidence show that the abnormal inflammatory response is closely associated with many chronic autoimmune diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic sclerosis, Sjӧgren’s syndrome, and diabetes, with concomitant tissue damage, remodelling, and, as end result, organ fibrosis [2,3]. Fibrosis is a pathological feature of most chronic inflammatory autoimmune diseases. The underlying molecular and cellular events of fibrotic autoimmune diseases share many functional similarities, despite differences in aetiology and clinical evolution. Although fibrosis is initially beneficial, the repair process can become pathogenic, and the continuous accumulation of fibrotic proteins leads to permanent tissue remodelling and significant organ dysfunction and failure. There are multiple versions of fibrotic autoimmune disorders that affecting a variety of organs, and it has been estimated that a significant percentage of deaths can be attributed to organ fibrotic transformation [4]. The molecular events and processes which contribute to the onset and development of autoimmune-associated fibrosis must be elucidated in order to develop novel efficacious treatments. This review was born with the intention of collecting the most recent contributions that can clarify the cellular and molecular mechanisms at the basis of the fibrotic evolution of various autoimmune diseases. The review focuses on the intertwined pathophysiological role of fibrosis and chronic inflammation in different tissues and autoimmune diseases, with the aim of identifying possible molecular bridges between the various fibrotic mechanisms identified in autoimmune diseases.

2. Molecular Mechanisms Involved in the Fibrotic Evolution during Autoimmune Diseases

Within the recent research topics, several experimental works on the cellular and molecular mediators of fibrosis in autoimmune diseases were produced, and the intertwined pathophysiological roles of fibrosis and chronic inflammation were demonstrated. The following paragraphs illustrate the mechanisms identified thus far for the fibrotic evolution during autoimmune diseases, in order to identify mediators common to several mechanisms and build connecting bridges between them.

2.1. Growth Factors and Associated Signalling Pathways

Inputs from growth factors converge on several signalling pathways to promote fibrosis inducing fibroblast activation, epithelial cell apoptosis, epithelial to mesenchymal transformation (EMT), and endothelial to mesenchymal transition (EndMT). The growth factors involved in these fibrotic pathways are represented by TGF-β factors, platelet-derived growth factors (PDGFs), fibroblast growth factors (FGFs), vascular endothelial growth factor (VEGF), and connective tissue growth factor (CTGF). Subsequent downstream signalling pathways are activated such as phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), Janus kinase (JAK)/signal transducer and activator of transcription (STAT), ADAM17 metalloproteinases, and WNT/β-catenin. The following paragraphs will illustrate the fibrotic mechanisms dependent on the activation of growth factors involved in the pathogenesis of various autoimmune diseases. The recently identified signalling pathways involved in fibrosis are depicted in Figure 1.

2.1.1. TGF-β Signalling Pathway

TGF-βs are the key cytokines in most fibrosis pathways. TGF-β is secreted from cells in a latent tripartite complex consisting of its dimeric inactive form (named latency-associated peptide [LAP]) plus a latent TGF-β-binding protein (LTBP) [5]. This TGF-β/LAP/LTBP inactive complex binds to the ECM components [5]. After cleavage by various proteases, the active form of TGF-β is released [5] and is able to bind to TGFβR2 and TGFβR1 receptors [6]. TGF-β expression is promoted by epidermal growth factor (EGF), IL-1, and TNF-α [7]; moreover, integrin αv/β6 could determine the activation of the TGF-β gene, due to the presence of an arginine–glycine–aspartate (RGD) motif in the precursors of TGF-β [8]. Experimental results demonstrated that by blocking integrin αv/β6 using a specific antibody, pulmonary fibrosis in mice was prevented and the inflammatory situation did not undergo evolution [9]. Two signalling pathways, known as Smad-mediated canonical and non-canonical, are activated by TGF-βs to regulate fibrotic evolution [7,10]. In the canonical pathway, after TGFβR1 activation, the Smad2 and Smad3 intracellular effector are phosphorylated; the subsequent interaction with Smad4 determines the nuclear transcription of target genes [11]. TGF-β can also activate Smad-independent non-canonical signalling pathways, through the activation of PI3K/AKT, mitogen-activated protein kinases (MAPKs), and JAK/STAT proteins [12]. Although fibroblasts are major sources and targets of TGF-β, some fibrogenic transformations reflect the activation of other cell types such as macrophages and epithelial cells [13]; in addition, the promotion of TGF-β-dependent fibrosis in autoimmune diseases involves the activation of diverse mechanisms that will be clarified in the next paragraphs.

TGF-β-Mediated Fibrosis through Activation of Resident Fibroblasts

The canonical TGF-β1/Smad3 signalling pathway initially determines the recruitment of inflammatory cells and fibroblasts into sites of injury and promotes the differentiation of fibroblasts to myofibroblasts which display exaggerated ECM production [14]. Several authors demonstrated that reactive oxygen species (ROS) mediate TGF-β-induced activation and transformation of fibroblasts. This involves the activation of NADPH oxidase (Nox) enzymes responsible for ROS formation [15]. Nox expression is, in turn, mediated by TGF-β. Nox4 expression, for example, can be induced by TGF-β in a variety of cells [15]. In primary human cardiac fibroblasts, TGF-β1 treatment increased the level of Nox4 and alpha-smooth muscle actin (α-SMA), a myofibroblast marker, whereas depletion of Nox4 decreased TGF-β1-stimulated α-SMA expression, demonstrating how the expression and activation of Nox and TGF-β are dependent on each other [16]. Although there is no certain evidence of the involvement of this ROS-mediated TGF-β1-induced fibrosis in autoimmune myocarditis, numerous preliminary studies seem to point towards this discovery.

TGF-β Determines Fibrosis through the Induction of Apoptosis

TGF-β1-dependent fibrosis and apoptosis are juxtaposed although the mechanisms might differ between different cell types. Endothelial cell apoptosis is mediated by ROS production which is dependent on TGF-β activation and mediated by p38 [17]. TGF-β1 could also induce apoptosis of mesangial cells in the kidney via p53 phosphorylation and up-regulation of the pro-apoptotic protein Bcl-2-associated protein X (Bax) [18]. In addition, there is evidence that fibroblasts from fibrotic tissues are resistant to apoptosis, and that TGF-β may confer resistance to apoptosis by classic death pathways such as the Fas–caspase activation cascade [19,20]. This mechanism may be mediated through the activation of signalling pathways, such as p38 MAPK and PI3K–Akt [21], and involve regulation of “inhibitor of apoptosis” (IAP) family members [22,23]. TGF-β may also promote fibroblast resistance to apoptosis through cell-cycle regulators such as p14ARF [24]. Inhibition of fibroblast and myofibroblast apoptosis through these pathways may explain the accumulation of these cells during fibrosis. Myofibroblasts are integral in a feedback loop that perpetuates fibrosis through the stiffening of the extracellular matrix. Lagares et al. determined that stiffness-activated myofibroblasts show an increased expression of pro-apoptotic proteins, and these cells become dependent on antiapoptotic protein expression to prevent their death [25]. A drug that mimics a pro-apoptotic protein blocking the anti-apoptotic protein BCL-XL was demonstrated to induce apoptosis in fibroblasts from patients with scleroderma, (also known as systemic sclerosis), a systemic autoimmune disease that often leads to fibrosis in the skin, heart, vasculature, and lungs [26]. In fact, the inhibition of anti-apoptotic BCL-2 proteins with a specific drug (ABT-263) resulted in effective reversal of fibrosis in a mouse model of scleroderma [27]. Based on these results, the block of anti-apoptotic proteins to induce myofibroblast apoptosis could be an effective strategy to treat fibrosis. A schematic representation of TGF-β mechanisms leading to the activation of fibrosis is shown in Figure 2.

TGF-β Regulates Fibrosis through the Activation of EMT

The most common type of EMT program activated in fibrotic evolution is type 2 EMT. Type 2 EMT, mainly triggered by chronic inflammation, is closely linked to the tissue damage repair response and gives rise to myofibroblasts through epithelial cell transition [28]. TGF-β is the main actor in this process, encouraging fibroblast proliferation and migration, promoting fibroblast phenotypic modifications into myofibroblasts and regulating type 2 EMT-dependent fibrosis [7]. TGF-β-mediated EMT-dependent fibrosis is regulated by various factors. Oxidative stress induced by TGF-β is a key event in the EMT process. ROS initiate several effects of TGF-β, influencing several downstream TGF-β signal transduction mediators, including Smads, MAPKs, and NF-κB [7]; TGF-β, on the other hand, increases the level of ROS by upregulating the expression of Nox4 and activating ERK and mTOR signalling-dependent EMT and fibrosis [29]. PI3K/AKT signals also mediate TGF-β-induced EMT [30]. PI3K/Akt and MAPK are two Smad-independent pathways induced by TGF-β1 that contribute to EMT-dependent fibrosis [30]. With this large and complicated scenario in autoimmunity, it is currently widely accepted that TGF-β1-mediated EMT-dependent fibrosis represents the evolution of various chronic inflammatory autoimmune diseases. Various researchers have demonstrated an altered expression of pro-fibrotic molecules in the joints of patients affected by RA [31]. In RA, TGF-β1 acts both as a pro-angiogenic molecule, predominantly in the synovial membrane [32], and as an activator of the synthesis of pro-inflammatory cytokines [32], metalloproteinases [33], and various fibrinolytic factors responsible for tissue remodelling, such as aggrecanase [34] and urokinase-type plasminogen activator [35]. In addition, in RA patients, the loss of inhibitory Smad7 was associated with marked activation of TGFβ/Smad3-dependent EMT [36]. Confirming the inhibitory role of Smad7 in tissue fibrosis and inflammation, the Smad7 deficiency observed in RA patients led to enhanced NF-κB activity, Th1/Th17 cell differentiation, and exacerbation of synovial inflammation, probably through the hyperactivation of the TGF-β/Smad3–IL-6 molecular pathway [36]. In RA, the link between TGF-β expression and EMT was recently confirmed through the observation that the synovial membrane or synovial fluid showed increased levels of TGF-β [37]; furthermore, EMT features, such as more aggressive and invasive cell phenotypes and resistance to apoptosis, that causes pannus tissue invasion and destruction in RA were detected [38]. Some highly innovative studies reported that, in RA, TGF-β is up-regulated by transglutaminase 2 (TG2), an enzyme that regulates ECM composition and degradation and, consequently, EMT through the activation of the transcription factors Snail/Slug, Twist, Zeb, and E47 [39]. In recent years, the activation of an EMT program related to fibrotic evolution has been demonstrated for various other autoimmune diseases, such as inflammatory bowel disease (IBD) [40], ulcerative colitis [41], and Crohn’s disease [42]. Additionally, renal fibrosis features are often linked to SLE nephritis [43]. Similarly, in autoimmune diabetes, complications that involve the lung are present, characterized by the induction and progression of fibrosis into the pulmonary tissue [44]. Diabetes, through the persistent effects of hyperglycaemia, can induce EMT-dependent fibrosis that is the result of inflammatory cell infiltrations into the lung and of the expression of elevated levels of ECM proteins, which induce an inflammatory process leading, consequently, to fibrosis and tissue injury [44]. This process is modulated mainly by the overexpression of TGF-β1 and the EMT transcriptional factor Snail; the elevated accumulation of ECM proteins in the pulmonary tissue is accompanied by the downregulation of epithelial markers such as ZO-1 and cadherin [45]. In autoimmune diabetes, EMT-induced fibrosis occurs via the activation of both SMAD-dependent and SMAD-independent pathways, as demonstrated by the increased levels of expression of the TGF-β1 receptor and SMAD2/3 protein in the diabetic cells, as well as increased levels of p38 and ERK [44]. Additionally, in multiple sclerosis, Troletti and colleagues hypothesized that TGF-β1-induced brain endothelial cell dysfunction might be due to brain endothelial cell trans-differentiation through EndoMT [45]. In multiple sclerosis lesions, TGF-β1 significantly boosted Snail mRNA and protein levels as well as the levels of mesenchymal markers such as fibronectin and vimentin [45,46]. On the contrary, the mRNA levels of junctional factors, such as claudin-1 and claudin-5, were significantly reduced. The triggering of EMT-dependent fibrosis in a situation of chronic inflammation, was recently demonstrated in the chronic inflammatory autoimmune disease Sjögren’s syndrome that affects salivary glands [10,47]. Salivary gland biopsies of patients affected by Sjögren’s syndrome show increased expression of TGF-β1. TGF-β1 stimulates salivary gland epithelial cells leading to phosphorylation and activation of Smad2/3, which form heterocomplexes with Smad4, triggering the canonical Smad-mediated EMT cascade. Interestingly, blocking canonical TGF-β1/Smad2/3 signal transduction had no effect on the activation of the non-canonical TGF-β1/Erk1/2/EMT pathway, suggesting that, in Sjögren’s syndrome, both the canonical and non-canonical signalling pathways are activated independently to induce EMT-dependent fibrosis [10,47]. Figure 3 shows the TGF-β-mediated mechanisms involved in EMT-dependent fibrotic evolution of autoimmune diseases.

2.1.2. PDGFs/PDGFRs

PDGFs are stimulators of cell division that are required for cell growth and proliferation of specific tissues. PDGF ligands bind to their receptors PDGFRαα, PDGFRαβ and PDGFRββ [48] activating downstream signals (RAS/MAPK, PI3K/AKT, and JAK/STAT pathways) [49]. PDGFs are mainly produced by macrophages, endothelial cells, and fibroblasts [50]. PDGFs and PDGF receptors are increased in the fibrotic skin lesions observed in autoimmune systemic diseases [51,52]. Furthermore, both PDGF-B and PDGF-D act as potent factors for hepatic stellate cell proliferation and migration, determining ECM deposition during liver fibrogenesis [53]. However, interestingly, PDGF-C seems to not be involved in liver fibrosis or functional liver impairment, or, as reported by some authors, PDGF-C and PDGF-D seem to be involved in the late stage of hepatic fibrogenesis [54]. Several recent studies demonstrated that PDGFs contribute to the formation of heart and lung fibrosis also via stimulating the activation of fibroblasts [55]. The mechanisms linking PDGF to the chronic inflammation observed in autoimmune conditions still remain to be clarified. Indeed, tissue fibrosis accompanying intractable chronic inflammation is associated with increased PDGF signalling and with proliferation and accumulation of PDGFR-positive mesenchymal cells or fibroblasts in the synovium of RA patients, in which a greater expression of phosphorylated active PDGFRαβ receptors was detected [56]. Interestingly, recent papers reported the presence of brain mesenchymal perivascular aggregates of PDGFRβ-positive cells in multiple sclerosis patients, where scar-forming cells can persist chronically in a condition of active inflammation and demyelination which characterize the disease [57].

2.1.3. FGFs/FGFRs

The fibroblast growth factor (FGF) family consists of signalling ligands that bind with variable affinity to four FGF receptors (FGFRs) [58]; FGFs can induce the dimerization, activation, and autophosphorylation of FGFRs and subsequently the activation of the RAS–extracellular signal-regulated kinase (ERK), PI3K–AKT, and JAK/STAT pathways [59]. The role of the FGF family in fibrosis development is not completely clear but was studied in the liver [60,61]. In particular, two isoforms of FGF2 showed very interesting behaviours during hepatic fibrogenesis; the form of FGF2 with a low molecular weight, when used to treat hepatic stellate cells, attenuated fibrosis by down-regulation of Delta-like 1 protein expression through the p38 MAPK pathway. In contrast, the isoform of FGF2 with a high molecular weight promoted hepatic fibrogenesis [62]. In the case of idiopathic pulmonary fibrosis, on the other hand, FGF1/FGFR signalling is aberrantly increased and may promote fibroblast migration via increased MAPK signalling, leading to the pathogenesis of lung fibrosis. Recent studies confirmed that the inhibition of FGF/FGFR signalling can reduce fibrosis in experimental animal models and the inhibition of FGF signalling is important in treating pulmonary fibrosis [63]. In RA, FGF-1 and FGF-2 have been implicated in abnormal synoviocyte proliferation and apoptosis resistance [64]. In addition, the FGF pathway seems to be implicated in interstitial (or diffuse parenchymal) lung diseases (ILDs) that represent a large, heterogeneous group of rare pulmonary pathologies, characterized by damaged parenchyma and mediated by varying degrees of chronic inflammation and fibrosis [65]. ILDs can represent the pulmonary complication of RA [66] and systemic sclerosis [67]. In the case of ILDs, the aetiology is unknown [68].

2.1.4. VEGFs/VEGFRs

The VEGF family comprises six members: VEGF-A, -B, -C, -D, and -E, and placental growth factor (PIGF) [69]. VEGFs regulate vasculogenesis, angiogenesis, and immunological responses [70]. VEGF-A, which exerts its biological functions by binding to VEGFR1 and VEGFR2, is widely studied as an angiogenesis regulator in homeostasis and diseases [71,72]. VEGF-A was decreased in autoimmune idiopathic pulmonary fibrosis patients, and the overexpression of VEGF-A protected lung tissue from damage and fibrosis [73]; Murray et al. [73] have proposed a non-cell autonomous function mediated by the endothelium to explain this epithelial-protective function of VEGF-A. However, there are conflicting reports as to whether VEGF-A is a contributing or protective factor against fibrosis because several clinical studies support the notion that VEGF-A might facilitate pulmonary fibrogenesis, depending on the specific type of VEGF-A isoform expressed in the tissue [74,75]. Systemic sclerosis is a rare autoimmune disease marked by the fibrosis of the skin and involvement of internal organs, especially the vascular system, lungs, kidneys, and gastrointestinal system, and is caused by excessive collagen deposition, immunological disturbances, and accompanying vascular changes. Microvascular damage and dysfunction of angiogenesis are the identified abnormalities in this disease. Tissue fibrosis results from a series of events, including endothelial dysfunction, inflammation, increased vascular permeability, and platelet aggregation [76]. Altered angiogenesis biomarker expression and microvascular damage are detected in the digit ulcers of systemic sclerosis patients [77]. Higher levels of VEGF in comparison to healthy controls were detected in both the early and established stages of systemic sclerosis [78]. Dysregulated tissue remodelling with aberrant fibrosis is one of the pathological hallmarks of autoimmune rheumatic diseases and interstitial lung disease is an important cause of disease-related morbidity across this group of disorders, particularly within connective tissue diseases such as systemic sclerosis [79]. Interstitial lung disease is the leading cause of disease-related mortality in systemic sclerosis [80]. The induction of VEGF pathways by hypoxia [81] has led researchers to consider its potential role in the pathogenesis of systemic sclerosis. Subsequent work examining VEGF-A splice isoforms provided a plausible explanation, having associated increased plasma levels of the VEGF-A165b splice variant with severe nailfold capillary loss [82]. In addition, when interstitial lung disease is associated with systemic sclerosis, lower VEGF-A BALF levels were detected compared to both healthy controls and systemic sclerosis patients without lung involvement [83]. Additionally, in rheumatoid arthritis, circulating VEGF-A is increased in patients’ sera, particularly in those patients with extra-articular manifestations (including pulmonary fibrosis) [84].

2.1.5. CTGF Signalling Pathway

Connective tissue growth factor (CTGF) is a secreted peptide involved in cell proliferation, angiogenesis, and wound healing; it has also been implicated in tumour development and tissue fibrosis [85]. CTGF is considered a factor that enhances PDGF-B signalling, especially in the PDGF-related regulation of the proliferation and chemotaxis of fibroblasts [86,87]. CTGF synthesis is induced by many pro-fibrotic cytokines such as TGFβ and VEGF [88]; CTGF can, in turn, combine with other factors to promote pro-fibrotic effects. TGF-β-mediated endogenous CTGF induction leads to negative regulation of Smad7 gene transcription; since Smad7 is an inhibitor of the canonical Smad-mediated TGF-β pathway, by blocking the inhibitory effect of Smad7, CTGF induces a persistent activation of pro-fibrotic TGF-β signalling [89]. CTGF regulates hepatic stellate cell adhesion, a critical event during fibrogenesis in hepatic fibrosis [90]. In addition, CTGF activates myofibroblast formation by trans-differentiating resident fibroblasts and epithelial cells through the activation of the EMT program [91]. Among various fibrotic diseases, CTGF has been extensively studied in autoimmune idiopathic pulmonary fibrosis [92] and cardiac [93], liver [94], and renal fibrosis [95]. In idiopathic pulmonary fibrosis, the overexpression of CTGF, in cooperation with TGFβ, is profibrotic and exacerbates ECM deposition in mouse lung tissues [96]. The severity of fibrosis was markedly attenuated by CTGF inhibition, confirming the pro-fibrotic activity of CTGF [97]. Although the involvement of CTGF has been well-documented in systemic sclerosis fibrosis, the therapeutic potential in targeting CTGF is still being studied in this autoimmune disease. The data collected evidenced a role for Angiotensin II which seems to induce skin fibrosis that was mitigated after CTGF gene silencing [98]. In CTGF knock out mice, the number of cells expressing PDGFRβ, procollagen, αSMA, pSmad2, CD45, and Fsp1 in the dermis was significantly reduced, suggesting a key role for CTGF in the fibrotic evolution in systemic sclerosis [99].

2.2. ADAM17 Activation and Fibrosis in Autoimmune Diseases

ADAM17 is a disintegrin and metalloproteinase (ADAM) family member that acts as a sheddase of various membrane proteins. Over the recent decades, ADAM17 has been reported to be a key factor in several biological pathways regulating proliferation, migration, and the immune response [100]. Therefore, it is not surprising that ADAM17, involved in the pathophysiology of numerous human diseases, is critically implicated in EMT and EMT-dependent fibrosis [101]. Since ADAM17 mediates the ectodomain shedding of various pro-inflammatory molecules, it is of no surprise that ADAM17 has attracted attention as a potential driver of inflammation and is repurposed pathologically during fibrosis [102]. Supporting this concept, high ADAM17 expression was detected in numerous human chronic inflammatory diseases, and it has been hypothesized that ADAM17 represents a convergence point between inflammation and the progression of degenerative EMT-dependent fibrotic diseases [103]. In fact, several recent studies have shown correlations between the increased levels of ADAM17 expression and the severity of fibrosis in patients with degenerative fibrotic diseases [103]. The specific role of ADAM17 in the pathophysiology of chronic inflammatory autoimmune diseases and fibrotic diseases is not fully understood and appears to depend on the cellular context. ADAM17 is a crucial modulator of the pathological airway remodelling in lung diseases, including asthma, chronic obstructive pulmonary disease, and cystic fibrosis [104]. In addition, the overexpression of ADAM-17 was detected in Sjӧgren’s syndrome in which pathological neovascularization was regulated by VEGF-A-stimulated ADAM17-dependent crosstalk between VEGFR2 and NF-κB [105] and ADAM17 may be involved in a cascade regulating the salivary gland fibrosis observed in Sjögren’s syndrome.

2.3. Phosphatidylinositol 3-Kinase (PI3K)/Protein Kinase B (PKB/AKT) Signalling Pathway

The PI3K/AKT signalling pathway regulates cell growth, proliferation, motility, metabolism, and survival [106]. PI3K is a group of lipid kinases associated with the plasma membrane [107], while AKT is a serine/threonine protein kinase activated in response to upstream PI3K [108]. Recently, the PI3K/AKT signalling pathway was implicated as a master regulator for idiopathic pulmonary fibrosis, a disease with an autoimmune aetiology, characterized by a chronic progressive interstitial fibrosis [109,110]. It seems that the overexpression of alpha-smooth muscle actin (α-SMA) in lung fibrosis was related to the activation of PI3K/AKT12, and the interaction between TGF-β and PI3K/AKT promoted lung fibrosis [111]. The activation of PI3K/AKT can determine pulmonary fibrosis by its downstream regulators of mammalian metabolism such as the target of rapamycin (mTOR), hypoxia inducible factor-1α (HIF-1α), and FOX family proteins [108,112]. Recently, the PI3K–AKT pathway was demonstrated to control the release of profibrotic mediators and to disturb the balance between profibrotic and anti-fibrotic mediators [113]. This response was accompanied by abnormal EMT activation, fibroblast proliferation, and fibroblast to myofibroblast transformation [114]. Additionally, myofibroblasts secrete ECM, mainly collagen, which leads to chaotic lung remodelling, and ultimately, progressive pulmonary fibrosis and loss of function. In this context, a pivotal role was exerted by epithelial cells; it is common to observe abnormal epithelial cells, such as bronchial epithelial cells and proliferative type II alveolar epithelial cells, in fibrotic areas in IPF lung biopsies, and studies have shown that alveolar epithelial cell damage is sufficient to cause pulmonary fibrosis [115]. Alveolar epithelial cell apoptosis is frequent in regions with high myofibroblast activity and fibrosis; in addition, these cells produce CTGF, PDGF, and TGF-β which are key fibrogenic mediators [116,117]. The PI3K/AKT signalling pathway has also been reported to be related to liver fibrosis in the course of autoimmune hepatitis, a chronic inflammatory disorder of the liver, characterized by elevation of serum immunoglobulin G (IgG), the presence of autoantibodies, and interface hepatitis on liver histology [118]. Recently, researchers found that PDGF-dependent overexpression of Sparc/osteonectin and kazal-like domain proteoglycan 1 (SPOCK1) promoted hepatic stellate cell activation and proliferation by activating the PI3K/Akt signalling pathway [119]. PI3K/AKT could also regulate angiogenesis by modulating the expression of angiogenic factors such as nitric oxide and angiopoietins and increasing VEGF/VEGFR signalling [120] and enhanced VEGFA/VEGFR2 signalling in liver fibrosis and angiogenesis [121]. In addition, JAK)/STAT-mediated transduction depends on the activation of PI3K/AKT/mTOR signalling [122] and JAK/STAT signals together with TGF-β1/Smad signals promote the EMT process in fibrosis [123].

2.4. WNT/β-Catenin

Wingless-related integration site (Wnt) proteins are secreted ligands that signal through the interaction with Frizzled receptors and low-density lipoprotein receptors. Upon binding to their receptors, Wnt proteins induce the stabilization of the transcription factor β-catenin that regulates Wnt target genes transcription [124]. The pro-fibrotic Wnt/β-catenin signalling seems to be active in autoimmune myocarditis [125]. In humans, infections with viruses or parasites, such as Trypanosoma cruzi, often induces heart-specific autoimmune responses, resulting in heart tissue inflammation; this inflammatory condition finally leads to dilated cardiomyopathy, with fibrotic changes in the myocardium and reduced myocardial contractility [126]. Published data have implicated Angiotensin II signalling in the development of autoimmune-mediated myocarditis [127]. Angiotensin II molecular activation was demonstrated to enhance the production of profibrotic TGF-β [128] and Angiotensin II has been implicated in the activation of the pro-fibrotic Wnt that, in turn, activates β-catenin, a transcription factor whose expression is mainly regulated by WNT proteins [125,129,130]. The WNT/β-catenin pathway activates and synergizes with TGF-β1 to mediate myofibroblast activation in autoimmune lung fibrosis [131]. In liver fibrosis, WNT/β-catenin also regulates the expression of vimentin, collagen 1, and fibronectin in hepatic stellate cells induced by TGF-β [132]; however, recently, there seems to be a turnaround derived from the demonstration that constitutively active canonical Wnt/β-catenin signalling confers tolerogenicity to hepatic dendritic cells under steady-state conditions and, therefore, deficiency of canonical Wnt/β-catenin signalling in these cells seems to be responsible for the triggering of autoimmune hepatitis [133].

2.5. Peroxisome Proliferator-Activated Receptors (PPARs) Signalling Pathway

Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors of the nuclear hormone receptor family [134] which regulate the activation of targeted genes related to lipid and glucose metabolism [135]. There are three PPARs: PPARα, PPARγ, and PPARβ/δ [136]. PPARα is predominantly expressed in brown adipose tissue and the liver [137]. PPAR-α and PPAR-γ activators are involved in the evolution of fibrotic diseases, particularly in cardiac fibrosis [138], renal fibrosis [139], and pulmonary fibrosis [140]. Systemic sclerosis is still a serious disease which is characterized by microvascular dysfunction, autoimmune reactivity, and organ fibrosis [141]. Fibrosis in multiple organs is a final common occurrence in systemic sclerosis [142]. The underlying mechanism of the uncontrolled fibrosis progression in systemic sclerosis remains unclear. However, the altered PPAR-γ expression or function in systemic sclerosis may partly explain the fibrotic program activation in this disease [143]. Several years ago, researchers demonstrated the expression of PPARγ in normal dermal fibroblasts and found that PPARγ inhibition could abrogate the TGFβ-induced collagen gene expression, inhibit myofibroblast differentiation, and repress Smad-dependent gene transcription; in addition, a reduction in PPAR-γ expression was observed in systemic sclerosis [143]. Kohno et al. confirmed the involvement of PPAR with fibrotic pathways because they found correlations between PPARγ ligand expression, the reduction of dermal sclerosis, and the decreased expression levels of CTGF and TGFβ in experimentally induced systemic sclerosis [144]. A further confirmation was derived from the studies conducted by Wu et al. that demonstrated that synthetic PPARγ ligand administration could attenuate inflammation and dermal fibrosis in an experimental animal model of scleroderma [145]. Mice with a knockout of PPARγ in fibroblasts became more susceptible to developing skin fibrosis, as indicated by increased collagen deposition and enhanced inflammation and susceptibility of fibroblasts to pro-fibrotic TGF-β1 signalling [146]. All of these studies established the role of PPARγ in regulating TGF-β-dependent fibrogenesis. The correlation of PPARs with liver fibrosis has been well established. Treatment with PPARα ligands attenuated liver fibrosis in autoimmune hepatitis [147]. The release of TGF-β1 and other inflammatory cytokines are mainly modulated by NF-κB in the development of liver fibrosis. The activation of PPARγ in hepatic stellate cells could block NF-κB by inhibiting the translocation of NF-κB to the nucleus, reducing NF-κB-dependent fibrosis [148].

3. Conclusions

Autoimmune diseases are a significant clinical problem because of their chronic nature, which compromises the quality of life, their prevalence in young populations, and the associated healthcare costs. Current therapies, such as the use of specific cytokine antagonists, have shown great promise; however, most of the current therapeutic agents target the terminal phase of the chronic inflammation of these diseases and do not address the fundamental problems that are responsible for the initiation and progression of the autoimmune process which often leads to organ fibrosis. Although controlling the inflammatory state is the most straightforward way to prevent tissue fibrosis, this can be a challenge, as the precise triggers of the inflammation remain unclear. Currently, no effective anti-fibrotic drug is yet available for clinical use in autoimmune patients. In conclusion, the present research topic has collected the most recent findings regarding novel and interconnected molecular pathways of multifaceted chronic inflammatory and fibrotic autoimmune diseases. The increasing elucidation of the molecular and cellular bases for chronic inflammation-associated organ fibrosis could paving the way towards multi-target therapeutic strategies as well as to novel treatments adaptable to various organs sharing similar fibrotic pathogenetic pathways.

Author Contributions

All authors were involved in drafting the article or critically revising it for important intellectual content, and all authors approved the final version for publication. M.S. and S.L. had full access to the data collected in the review, take responsibility for their integrity and performed a critical reading. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAM17a disintegrin and metalloprotease 17
AKTprotein-chinasi B
α-SMAalpha-smooth muscle actin
BaxBcl-2 Associated protein X
BCL-XLB-cell lymphoma-extra large
CTGFconnective tissue growth factor
ECMextracellular matrix
EGFepidermal growth factor
EMTepithelial mesenchymal transition
EndMTendothelial mesenchymal transition
ERKextracellular signal-regulated kinase
FGFsfibroblast growth factors
FOXforkhead box
FSP1ferroptosis suppressor protein 1
HIF-1αhypoxia inducible factor-1α
IAPinhibitor of apoptosis
IBDinflammatory bowel disease
IL-1interleukin-1
ILDsinterstitial lung diseases
IPFidiopathic pulmonary fibrosis
JAKJanus kinase
LAPlatency-associated peptide
LTBPlatent TGF-β binding protein
MAPKsmitogen-activated protein kinases
mTORmammalian/mechanistic target of rapamycin
NF-κBnuclear factor kappa-light-chain-enhancer of activated B cells
NoxNADPH oxidase
PDGFsplatelet-derived growth factors
PI3Kphosphatidylinositol 3-kinase
PPARSperoxisome proliferator-activated receptors
RArheumatoid arthritis
RGDarginine-glycine-aspartate
ROSreactive oxygen species
SLEsystemic lupus erythematosus
Smadsuppressor of mothers against decapentaplegic
SNAILzinc finger protein SNAI
SPOCK1kazal-like domain proteoglycan 1
STATsignal transducer and activator of transcription
SSSjӧgren’s syndrome
SScsystemic sclerosis
TG2transglutaminase 2
TGF-βtumor growth factor Beta
TNF-αtumor necrosis factor
TWISTTwist-related protein 1
VEGFvascular endothelial growth factor
WNTwingless/Integrated
ZEBzinc Finger E-Box Binding Homeobox
ZO-1zonula occludens-1

References

  1. Ahsan, H. Origins and history of autoimmunity. A brief review. Rheumatol. Autoimmun. 2023, 3, 9–14. [Google Scholar] [CrossRef]
  2. Wang, L.; Wang, F.S.; Gershwin, M.E. Human autoimmune diseases: A comprehensive update. J. Intern. Med. 2015, 278, 369–395. [Google Scholar] [CrossRef] [PubMed]
  3. Sisto, M.; Ribatti, D.; Lisi, S. Molecular Mechanisms Linking Inflammation to Autoimmunity in Sjögren’s Syndrome: Identification of New Targets. Int. J. Mol. Sci. 2022, 23, 13229. [Google Scholar] [CrossRef]
  4. Ueha, S.; Shand, F.H.; Matsushima, K. Cellular and molecular mechanisms of chronic inflammation-associated organ fibrosis. Front. Immunol. 2012, 3, 71. [Google Scholar] [CrossRef]
  5. Munger, J.S.; Sheppard, D. Cross talk among TGF-β signalling pathways, integrins, and the extracellular matrix. Cold Spring Harb. Perspect. Biol. 2011, 3, a005017. [Google Scholar] [CrossRef] [PubMed]
  6. Sanjabi, S.; Oh, S.A.; Li, M.O. Regulation of the Immune Response by TGF-β: From Conception to Autoimmun-ity and Infection. Cold Spring Harb. Perspect. Biol. 2017, 9, a022236. [Google Scholar] [CrossRef]
  7. Chung, J.Y.F.; Chan, M.K.K.; Li, J.S.F.; Chan, A.S.W.; Tang, P.C.T.; Leung, K.T.; To, K.F.; Lan, H.Y.; Tang, P.M.K. TGF-β Signalling: From Tissue Fibrosis to Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 7575. [Google Scholar] [CrossRef]
  8. Mamuya, F.A.; Duncan, M.K. aV integrins and TGF-β-induced EMT: A circle of regulation. J. Cell Mol. Med. 2012, 16, 445–455. [Google Scholar] [CrossRef]
  9. Horan, G.S.; Wood, S.; Ona, V.; Li, D.J.; Lukashev, M.E.; Weinreb, P.H.; Simon, K.J.; Hahm, K.; Allaire, N.E.; Rinaldi, N.J.; et al. Partial inhibition of integrin alpha(v)beta6 prevents pulmonary fibrosis without exacerbating inflammation. Am. J. Respir. Crit. Care Med. 2008, 177, 56–65. [Google Scholar] [CrossRef]
  10. Sisto, M.; Ribatti, D.; Lisi, S. SMADS-Mediate Molecular Mechanisms in Sjögren’s Syndrome. Int. J. Mol. Sci. 2021, 22, 3203. [Google Scholar] [CrossRef]
  11. 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]
  12. Finnson, K.W.; Almadani, Y.; Philip, A. Non-canonical (non-SMAD2/3) TGF-beta signalling in fibrosis: Mechanisms and targets. Semin. Cell Dev. Biol. 2020, 101, 115–122. [Google Scholar] [CrossRef] [PubMed]
  13. Wynn, T.A.; Ramalingam, T.R. Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef]
  14. Zhao, M.; Wang, L.; Wang, M.; Zhou, S.; Lu, Y.; Cui, H.; Racanelli, A.C.; Zhang, L.; Ye, T.; Ding, B.; et al. Targeting fibrosis: Mechanisms and clinical trials. Sig. Transduct. Target Ther. 2022, 7, 206. [Google Scholar] [CrossRef]
  15. Liu, R.M.; Desai, L.P. Reciprocal regulation of TGF-β and reactive oxygen species: A perverse cycle for fibrosis. Redox Biol. 2015, 6, 565–577. [Google Scholar] [CrossRef]
  16. Cucoranu, I.; Clempus, R.; Dikalova, A.; Phelan, P.J.; Ariyan, S.; Dikalov, S.; Sorescu, D. NAD(P)H oxidase 4 mediates transforming growth factor-beta1-induced differentiation of cardiac fibroblasts into myofibroblasts. Circ. Res. 2005, 97, 900–907. [Google Scholar] [CrossRef] [PubMed]
  17. Edlund, S.; Bu, S.; Schuster, N.; Aspenström, P.; Heuchel, R.; Heldin, N.E.; ten Dijke, P.; Heldin, C.H.; Landström, M. Transforming growth factor-beta1 (TGF-beta)-induced apoptosis of prostate cancer cells involves Smad7-dependent activation of p38 by TGF-beta-activated kinase 1 and mitogen-activated protein kinase kinase 3. Mol. Biol. Cell. 2003, 14, 529–544. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, B.; Luo, Y.; Zhou, X.; Li, R. Trifluoperazine induces apoptosis through the upregulation of Bax/Bcl-2 and downregulated phosphorylation of AKT in mesangial cells and improves renal function in lupus nephritis mice. Int. J. Mol. Med. 2018, 41, 3278–3286. [Google Scholar] [CrossRef]
  19. Tanaka, T.; Yoshimi, M.; Maeyama, T.; Hagimoto, N.; Kuwano, K.; Hara, N. Resistance to Fas-mediated apoptosis in human lung fibroblast. Eur. Respir. J. 2002, 20, 359–368. [Google Scholar] [CrossRef] [PubMed]
  20. Thannickal, V.J. Mechanistic links between aging and lung fibrosis. Biogerontology 2013, 14, 609–615. [Google Scholar] [CrossRef] [PubMed]
  21. Kulasekaran, P.; Scavone, C.A.; Rogers, D.S.; Arenberg, D.A.; Thannickal, V.J.; Horowitz, J.C. Endothelin-1 and transforming growth factor-β1 independently induce fibroblast resistance to apoptosis via AKT activation. Am. J. Respir. Cell Mol. Biol. 2009, 41, 484–493. [Google Scholar] [CrossRef] [PubMed]
  22. Horowitz, J.C.; Ajayi, I.O.; Kulasekaran, P.; Rogers, D.S.; White, J.B.; Townsend, S.K.; White, E.S.; Nho, R.S.; Higgins, P.D.; Huang, S.K. Survivin expression induced by endothelin-1 promotes myofibroblast resistance to apoptosis. Int. J. Biochem. Cell Biol. 2012, 44, 158–169. [Google Scholar] [CrossRef] [PubMed]
  23. Sisson, T.H.; Ajayi, I.O.; Subbotina, N.; Dodi, A.E.; Rodansky, E.S.; Chibucos, L.N.; Kim, K.K.; Keshamouni, V.G.; White, E.S.; Zhou, Y.; et al. Inhibition of myocardin-related transcription factor/serum response factor signalling decreases lung fibrosis and promotes mesenchymal cell apoptosis. Am. J. Pathol. 2015, 185, 969–986. [Google Scholar] [CrossRef]
  24. Cisneros, J.; Hagood, J.; Checa, M.; Ortiz-Quintero, B.; Negreros, M.; Herrera, I.; Ramos, C.; Pardo, A.; Selman, M. Hypermethylation-mediated silencing of p14ARF in fibroblasts from idiopathic pulmonary fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 303, L295–L303. [Google Scholar] [CrossRef]
  25. Lagares, D.; Santos, A.; Grasberger, P.E.; Liu, F.; Probst, C.K.; Rahimi, R.A.; Sakai, N.; Kuehl, T.; Ryan, J.; Bhola, P.; et al. Targeted apoptosis of myofibroblasts with the BH3 mimetic ABT-263 reverses established fibrosis. Sci. Transl. Med. 2017, 9, 3765. [Google Scholar] [CrossRef]
  26. Jafarinejad-Farsangi, S.; Farazmand, A.; Mahmoudi, M.; Gharibdoost, F.; Karimizadeh, E.; Noorbakhsh, F.; Faridani, H.; Jamshidi, A.R. MicroRNA-29a induces apoptosis via increasing the Bax:Bcl-2 ratio in dermal fibroblasts of patients with systemic sclerosis. Autoimmunity 2015, 48, 369–378. [Google Scholar] [CrossRef]
  27. Cooley, J.C.; Javkhlan, N.; Wilson, J.A.; Foster, D.G.; Edelman, B.L.; Ortiz, L.A.; Schwartz, D.A.; Riches, D.W.; Redente, E.F. Inhibition of antiapoptotic BCL-2 proteins with ABT-263 induces fibroblast apoptosis, reversing persistent pulmonary fibrosis. J. Clin. Investig. 2023, 8, e163762. [Google Scholar] [CrossRef]
  28. Marconi, G.D.; Fonticoli, L.; Rajan, T.S.; Pierdomenico, S.D.; Trubiani, O.; Pizzicannella, J.; Diomede, F. Epithelial-Mesenchymal Transition (EMT): The Type-2 EMT in Wound Healing, Tissue Regeneration and Organ Fibrosis. Cells 2021, 10, 1587. [Google Scholar] [CrossRef]
  29. Kim, S.J.; Kim, Y.S.; Kim, J.H.; Jang, H.Y.; Ly, D.D.; Das, R.; Park, K.S. Activation of ERK1/2-mTORC1-NOX4 mediates TGF-β1-induced epithelial-mesenchymal transition and fibrosis in retinal pigment epithelial cells. Biochem. Biophys. Res. Commun. 2020, 529, 747–752. [Google Scholar] [CrossRef] [PubMed]
  30. Liu, L.; Sun, Q.; Davis, F.; Mao, J.; Zhao, H.; Ma, D. Epithelial-mesenchymal transition in organ fibrosis development: Current understanding and treatment strategies. Burn. Trauma 2022, 10, tkac011. [Google Scholar] [CrossRef]
  31. Zhang, L.; Xing, R.; Huang, Z.; Ding, L.; Zhang, L.; Li, M.; Li, X.; Wang, P.; Mao, J. Synovial Fibrosis Involvement in Osteoarthritis. Front. Med. 2021, 8, 684389. [Google Scholar] [CrossRef]
  32. Gonzalo-Gil, E.; Galindo-Izquierdo, M. Role of transforming growth factor-beta (TGF) beta in the physiopathology of rheumatoid arthritis. Reumatol. Clin. 2014, 10, 174–179. [Google Scholar] [CrossRef]
  33. Zhu, D.; Zhao, J.; Lou, A.; Huang, Q.; Yang, Q.; Zhu, J.; Fan, M.; He, Y.; Ren, H.; Yang, M. Transforming growth factor β1 promotes fibroblast-like synoviocytes migration and invasion via TGF-β1/Smad signalling in rheumatoid arthritis. Mol. Cell Biochem. 2019, 459, 141–150. [Google Scholar] [CrossRef]
  34. Moulharat, N.; Lesur, C.; Thomas, M.; Rolland-Valognes, G.; Pastoureau, P.; Anract, P.; De Ceuninck, F.; Sabatini, M. Effects of transforming growth factor-beta on aggrecanase production and proteoglycan degradation by human chondrocytes in vitro. Osteoarthr. Cartil. 2004, 12, 296–305. [Google Scholar] [CrossRef]
  35. Schuliga, M. The inflammatory actions of coagulant and fibrinolytic proteases in disease. Mediat. Inflamm. 2015, 2015, 437695. [Google Scholar] [CrossRef] [PubMed]
  36. Zhou, G.; Sun, X.; Qin, Q.; Lv, J.; Cai, Y.; Wang, M.; Mu, R.; Lan, H.Y.; Wang, Q.W. Loss of Smad7 Promotes Inflammation in Rheumatoid Arthritis. Front. Immunol. 2018, 9, 2537. [Google Scholar] [CrossRef] [PubMed]
  37. Fennen, M.; Pap, T.; Dankbar, B. Smad-dependent mechanisms of inflammatory bone destruction. Arthritis Res. Ther. 2016, 18, 279. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, S.Y. Does epithelial-mesenchymal transition happen in rheumatoid joints? Eur. J. Rheumatol. 2014, 1, 86–87. [Google Scholar] [CrossRef]
  39. Benn, M.C.; Weber, W.; Klotzsch, E.; Vogel, V.; Pot, S.A. Tissue transglutaminase in fibrosis- more than an extracellular matrix cross-linker. Curr. Opin. Biomed. Eng. 2019, 10, 156–164. [Google Scholar] [CrossRef]
  40. Wang, Y.; Huang, B.; Jin, T.; Ocansey, D.K.W.; Jiang, J.; Mao, F. Intestinal Fibrosis in Inflammatory Bowel Disease and the Prospects of Mesenchymal Stem Cell Therapy. Front. Immunol. 2022, 13, 835005. [Google Scholar] [CrossRef]
  41. Rieder, F.; Fiocchi, C.; Rogler, G. Mechanisms, Management, and Treatment of Fibrosis in Patients with Inflammatory Bowel Diseases. Gastroenterology 2017, 152, 340–350. [Google Scholar] [CrossRef] [PubMed]
  42. Honzawa, Y.; Yamamoto, S.; Okabe, M.; Seno, H.; Nakase, H. Current Topics of the Mechanism of Intestinal Fibrosis in Crohn’s Disease. Immuno 2021, 1, 574–582. [Google Scholar] [CrossRef]
  43. Sciascia, S.; Cozzi, M.; Barinotti, A.; Radin, M.; Cecchi, I.; Fenoglio, R.; Mancardi, D.; Wilson Jones, G.; Rossi, D.; Roccatello, D. Renal Fibrosis in Lupus Nephritis. Int. J. Mol. Sci. 2022, 23, 14317. [Google Scholar] [CrossRef]
  44. Talakatta, G.; Sarikhani, M.; Muhamed, J.; Dhanya, K.; Somashekar, B.S.; Mahesh, P.A.; Sundaresan, N.; Ravindra, P.V. Diabetes induces fibrotic changes in the lung through the activation of TGF-β signalling pathways. Sci. Rep. 2018, 8, 11920. [Google Scholar] [CrossRef]
  45. You, S.; Thieblemont, N.; Alyanakian, M.A.; Bach, J.F.; Chatenoud, L. Transforming growth factor-beta and T-cell-mediated immunoregulation in the control of autoimmune diabetes. Immunol. Rev. 2006, 212, 185–202. [Google Scholar] [CrossRef] [PubMed]
  46. Derada Troletti, C.; Fontijn, R.D.; Gowing, E.; Charabati, M.; van Het Hof, B.; Didouh, I.; van der Pol, S.M.A.; Geerts, D.; Prat, A.; van Horssen, J.; et al. Inflammation-induced endothelial to mesenchymal transition promotes brain endothelial cell dysfunction and occurs during multiple sclerosis pathophysiology. Cell Death Dis. 2019, 10, 45. [Google Scholar] [CrossRef]
  47. Sisto, M.; Lorusso, L.; Ingravallo, G.; Ribatti, D.; Lisi, S. TGFβ1-Smad canonical and -Erk noncanonical pathways participate in interleukin-17-induced epithelial-mesenchymal transition in Sjögren’s syndrome. Lab. Investig. 2020, 100, 824–836. [Google Scholar] [CrossRef]
  48. Shim, A.H.; Liu, H.; Focia, P.J.; Chen, X.; Lin, P.C.; He, X. Structures of a platelet-derived growth factor/propeptide complex and a platelet-derived growth factor/receptor complex. Proc. Natl. Acad. Sci. USA 2010, 107, 11307–11312. [Google Scholar] [CrossRef]
  49. Pandey, P.; Khan, F.; Upadhyay, T.K.; Seungjoon, M.; Park, M.N.; Kim, B. New insights about the PDGF/PDGFR signalling pathway as a promising target to develop cancer therapeutic strategies. Biomed. Pharmacother. 2023, 161, 114491. [Google Scholar] [CrossRef]
  50. Shimokado, K.; Raines, E.W.; Madtes, D.K.; Barrett, T.B.; Benditt, E.P.; Ross, R. A significant part of macrophage-derived growth factor consists of at least two forms of PDGF. Cell 1985, 43, 277–286. [Google Scholar] [CrossRef]
  51. Daoussis, D.; Tsamandas, A.C.; Liossis, S.N.; Antonopoulos, I.; Karatza, E.; Yiannopoulos, G.; Andonopoulos, A.P. B-cell depletion therapy in patients with diffuse systemic sclerosis associates with a significant decrease in PDGFR expression and activation in spindle-like cells in the skin. Arthritis Res. Ther. 2012, 14, R145. [Google Scholar] [CrossRef] [PubMed]
  52. 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]
  53. Ying, H.Z.; Chen, Q.; Zhang, W.Y.; Zhang, H.H.; Ma, Y.; Zhang, S.Z.; Fang, J.; Yu, C.H. PDGF signalling pathway in hepatic fibrosis pathogenesis and therapeutics. Mol. Med. Rep. 2017, 16, 7879–7889. [Google Scholar] [CrossRef]
  54. Breitkopf, K.; Roeyen, C.V.; Sawitza, I.; Wickert, L.; Floege, J.; Gressner, A.M. Expression patterns of PDGF-A, -B, -C and -D and the PDGF-receptors alpha and beta in activated rat hepatic stellate cells (HSC). Cytokine 2005, 31, 349–357. [Google Scholar] [CrossRef] [PubMed]
  55. Zhao, T.; Zhao, W.; Chen, Y.; Li, V.S.; Meng, W.; Sun, Y. Platelet-derived growth factor-D promotes fibrogenesis of cardiac fibroblasts. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H1719–H1726. [Google Scholar] [CrossRef]
  56. Matsumura, T.; Saito, Y.; Suzuki, T.; Teramoto, A.; Ozasa, Y.; Yamashita, T.; Fujimiya, M.; Saito-Chikenji, T. Phosphorylated Platelet-Derived Growth Factor Receptor-Positive Cells With Anti-apoptotic Properties Accumulate in the Synovium of Patients With Rheumatoid Arthritis. Front. Immunol. 2019, 10, 241. [Google Scholar] [CrossRef]
  57. Iacobaeus, E.; Sugars, R.V.; Tornqvist Andren, A.; Alm, J.J.; Qian, H.; Frantzen, J.; Newcombe, J.; Alkass, K.; Druid, H.; Bottai, M.; et al. Dynamic changes in brain mesenchymal perivascular cells associate with multiple sclerosis disease duration, active inflammation, and demyelination. Stem. Cells Transl. Med. 2017, 6, 1840–1851. [Google Scholar] [CrossRef]
  58. Xie, Y.; Su, N.; Yang, J.; Tan, Q.; Huang, S.; Jin, M.; Ni, Z.; Zhang, B.; Zhang, D.; Luo, F.; et al. FGF/FGFR signalling in health and disease. Sig. Transduct. Target Ther. 2020, 5, 181. [Google Scholar] [CrossRef]
  59. Szymczyk, J.; Sluzalska, K.D.; Materla, I.; Opalinski, L.; Otlewski, J.; Zakrzewska, M. FGF/FGFR-Dependent Molecular Mechanisms Underlying Anti-Cancer Drug Resistance. Cancers 2021, 13, 5796. [Google Scholar] [CrossRef] [PubMed]
  60. Seitz, T.; Hellerbrand, C. Role of fibroblast growth factor signalling in hepatic fibrosis. Liver Int. 2021, 41, 1201–1215. [Google Scholar] [CrossRef]
  61. Guzy, R. Fibroblast Growth Factor Inhibitors in Lung Fibrosis: Friends or Foes? Am. J. Respir. Cell Mol. Biol. 2020, 63, 273–274. [Google Scholar] [CrossRef] [PubMed]
  62. Pan, R.L.; Xiang, L.X.; Wang, P.; Liu, X.Y.; Nie, L.; Huang, W.; Shao, J.Z. Low-molecular-weight fibroblast growth factor 2 attenuates hepatic fibrosis by epigenetic down-regulation of Delta-like1. Hepatology 2015, 61, 1708–1720. [Google Scholar] [CrossRef] [PubMed]
  63. Wollin, L.; Wex, E.; Pautsch, A.; Schnapp, G.; Hostettler, K.E.; Stowasser, S.; Kolb, M. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur. Respir. J. 2015, 45, 1434. [Google Scholar] [CrossRef] [PubMed]
  64. Malemud, C.J. Growth hormone, VEGF and FGF: Involvement in rheumatoid arthritis. Clin. Chim. Acta 2007, 375, 10–19. [Google Scholar] [CrossRef]
  65. Richeldi, L.; Varone, F.; Bergna, M.; de Andrade, J.; Falk, J.; Hallowell, R.; Jouneau, S.; Kondoh, Y.; Morrow, L.; Randerath, W.; et al. Pharmacological management of progressive-fibrosing interstitial lung diseases: A review of the current evidence. Eur. Respir. Rev. 2018, 27, 180074. [Google Scholar] [CrossRef]
  66. Picchianti Diamanti, A.; Markovic, M.; Argento, G.; Giovagnoli, S.; Ricci, A.; Laganà, B.; D’Amelio, R. Therapeutic management of patients with rheumatoid arthritis and associated interstitial lung disease: Case report and literature review. Ther. Adv. Respir. Dis. 2017, 11, 64–72. [Google Scholar] [CrossRef]
  67. Cottin, V.; Brown, K.K. Interstitial lung disease associated with systemic sclerosis (SSc-ILD). Respir. Res. 2019, 20, 13. [Google Scholar] [CrossRef] [PubMed]
  68. Olson, A.L.; Gifford, A.H.; Inase, N.; Fernández Pérez, E.R.; Suda, T. The epidemiology of idiopathic pulmonary fibrosis and interstitial lung diseases at risk of a progressive-fibrosing phenotype. Eur. Respir. Rev. 2018, 27, 180077. [Google Scholar] [CrossRef]
  69. Holmes, D.I.; Zachary, I. The vascular endothelial growth factor (VEGF) family: Angiogenic factors in health and disease. Genome Biol. 2005, 6, 209. [Google Scholar] [CrossRef]
  70. Ye, X.; Gaucher, J.F.; Vidal, M.; Broussy, S. A Structural Overview of Vascular Endothelial Growth Factors Pharmacological Ligands: From Macromolecules to Designed Peptidomimetics. Molecules 2021, 26, 6759. [Google Scholar] [CrossRef]
  71. Ballmer-Hofer, K. Vascular Endothelial Growth Factor, from Basic Research to Clinical Applications. Int. J. Mol. Sci. 2018, 19, 3750. [Google Scholar] [CrossRef] [PubMed]
  72. Peach, C.J.; Mignone, V.W.; Arruda, M.A.; Alcobia, D.C.; Hill, S.J.; Kilpatrick, L.E.; Woolard, J. Molecular Pharmacology of VEGF-A Isoforms: Binding and Signalling at VEGFR2. Int. J. Mol. Sci. 2018, 19, 1264. [Google Scholar] [CrossRef] [PubMed]
  73. Murray, L.A.; Habiel, D.M.; Hohmann, M.; Camelo, A.; Shang, H.; Zhou, Y.; Coelho, A.L.; Peng, X.; Gulati, M.; Crestani, B.; et al. Antifibrotic role of vascular endothelial growth factor in pulmonary fibrosis. J. Clin. Investig. 2017, 2, e92192. [Google Scholar] [CrossRef] [PubMed]
  74. Hamada, N.; Kuwano, K.; Yamada, M.; Hagimoto, N.; Hiasa, K.; Egashira, K.; Nakashima, N.; Maeyama, T.; Yoshimi, M.; Nakanishi, Y. Anti-vascular endothelial growth factor gene therapy attenuates lung injury and fibrosis in mice. J. Immunol. 2005, 175, 1224–1231. [Google Scholar] [CrossRef] [PubMed]
  75. Barratt, S.L.; Blythe, T.; Jarrett, C.; Ourradi, K.; Shelley-Fraser, G.; Day, M.J.; Qiu, Y.; Harper, S.; Maher, T.M.; Oltean, S.; et al. Differential Expression of VEGF-Axxx Isoforms Is Critical for Development of Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2017, 196, 479–493. [Google Scholar] [CrossRef]
  76. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, C.; Tousoulis, D. Inflammatory Mechanisms Contributing to Endothelial Dysfunction. Biomedicines 2021, 9, 781. [Google Scholar] [CrossRef]
  77. Zanin-Silva, D.C.; Santana-Gonçalves, M.; Kawashima-Vasconcelos, M.Y.; Oliveira, M.C. Management of Endothelial Dysfunction in Systemic Sclerosis: Current and Developing Strategies. Front. Med. 2021, 8, 788250. [Google Scholar] [CrossRef]
  78. Silva, I.; Almeida, C.; Teixeira, A.; Oliveira, J.; Vasconcelos, C. Impaired angiogenesis as a feature of digital ulcers in systemic sclerosis. Clin. Rheumatol. 2016, 35, 1743–1751. [Google Scholar] [CrossRef]
  79. Gutsche, M.; Rosen, G.D.; Swigris, J.J. Connective Tissue Disease-associated Interstitial Lung Disease: A review. Curr. Respir. Care Rep. 2012, 1, 224–232. [Google Scholar] [CrossRef]
  80. 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]
  81. Marti, H.H.; Risau, W. Systemic hypoxia changes the organ-specific distribution of vascular endothelial growth factor and its receptors. Proc. Natl. Acad. Sci. USA 1998, 95, 15809–15814. [Google Scholar] [CrossRef] [PubMed]
  82. Manetti, M.; Guiducci, S.; Romano, E.; Bellando-Randone, S.; Lepri, G.; Bruni, C.; Conforti, M.L.; Ibba-Manneschi, L.; Matucci-Cerinic, M. Increased plasma levels of the VEGF165b splice variant are associated with the severity of nailfold capillary loss in systemic sclerosis. Ann. Rheum. Dis. 2013, 72, 1425–1427. [Google Scholar] [CrossRef] [PubMed]
  83. De Santis, M.; Bosello, S.L.; Capoluongo, E.; Inzitari, R.; Peluso, G.; Lulli, P.; Zizzo, G.; Bocci, M.; Tolusso, B.; Zuppi, C.; et al. A vascular endothelial growth factor deficiency characterises scleroderma lung disease. Ann. Rheum. Dis. 2012, 71, 1461–1465. [Google Scholar] [CrossRef] [PubMed]
  84. Hashimoto, N.; Iwasaki, T.; Kitano, M.; Ogata, A.; Hamano, T. Levels of vascular endothelial growth factor and hepatocyte growth factor in sera of patients with rheumatic diseases. Mod. Rheumatol. 2003, 13, 129–134. [Google Scholar] [CrossRef]
  85. Akkız, H. Emerging Role of Cancer-Associated Fibroblasts in Progression and Treatment of Hepatocellular Carcinoma. Int. J. Mol. Sci. 2023, 24, 3941. [Google Scholar] [CrossRef]
  86. Yang, Z.; Sun, Z.; Liu, H.; Ren, Y.; Shao, D.; Zhang, W.; Lin, J.; Wolfram, J.; Wang, F.; Nie, S. Connective tissue growth factor stimulates the proliferation, migration and differentiation of lung fibroblasts during paraquat-induced pulmonary fibrosis. Mol. Med. Rep. 2015, 12, 1091–1097. [Google Scholar] [CrossRef]
  87. Chen, Z.; Zhang, N.; Chu, H.Y.; Yu, Y.; Zhang, Z.K.; Zhang, G.; Zhang, B.T. Connective Tissue Growth Factor: From Molecular Understandings to Drug Discovery. Front. Cell Dev. Biol. 2020, 8, 593269. [Google Scholar] [CrossRef]
  88. Lipson, K.E.; Wong, C.; Teng, Y.; Spong, S. CTGF is a central mediator of tissue remodeling and fibrosis and its inhibition can reverse the process of fibrosis. Fibrogenes. Tissue Repair 2012, 5, S24. [Google Scholar] [CrossRef]
  89. Qi, W.; Chen, X.; Twigg, S.; Zhang, Y.; Gilbert, R.E.; Kelly, D.J.; Pollock, C.A. The differential regulation of Smad7 in kidney tubule cells by connective tissue growth factor and transforming growth factor-beta1. Nephrology 2007, 12, 267–274. [Google Scholar] [CrossRef]
  90. Huang, G.; Brigstock, D.R. Regulation of hepatic stellate cells by connective tissue growth factor. Front. Biosci. 2012, 17, 2495–2507. [Google Scholar] [CrossRef]
  91. Kis, K.; Liu, X.; Hagood, J.S. Myofibroblast differentiation and survival in fibrotic disease. Exp. Rev. Mol. Med. 2011, 13, e27. [Google Scholar] [CrossRef] [PubMed]
  92. Effendi, W.I.; Nagano, T. Connective Tissue Growth Factor in Idiopathic Pulmonary Fibrosis: Breaking the Bridge. Int. J. Mol. Sci. 2022, 23, 6064. [Google Scholar] [CrossRef]
  93. Dorn, L.E.; Petrosino, J.M.; Wright, P.; Accornero, F. CTGF/CCN2 is an autocrine regulator of cardiac fibrosis. J. Mol. Cell. Cardiol. 2018, 121, 205–211. [Google Scholar] [CrossRef]
  94. Trampuž, S.R.; van Riet, S.; Nordling, Å.; Ingelman-Sundberg, M. The Role of CTGF in Liver Fibrosis Induced in 3D Human Liver Spheroids. Cells 2023, 12, 302. [Google Scholar] [CrossRef] [PubMed]
  95. Toda, N.; Mukoyama, M.; Yanagita, M.; Yokoi, H. CTGF in kidney fibrosis and glomerulonephritis. Inflamm. Regen. 2018, 38, 14. [Google Scholar] [CrossRef] [PubMed]
  96. Yanagihara, T.; Tsubouchi, K.; Gholiof, M.; Chong, S.G.; Lipson, K.E.; Zhou, Q.; Scallan, C.; Upagupta, C.; Tikkanen, J.; Keshavjee, S.; et al. Connective-Tissue Growth Factor Contributes to TGF-β1-induced Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 66, 260–270. [Google Scholar] [CrossRef]
  97. Sakai, N.; Nakamura, M.; Lipson, K.E.; Miyake, T.; Kamikawa, Y.; Sagara, A.; Shinozaki, Y.; Kitajima, S.; Toyama, T.; Hara, A.; et al. Inhibition of CTGF ameliorates peritoneal fibrosis through suppression of fibroblast and myofibroblast accumulation and angiogenesis. Sci. Rep. 2017, 7, 5392. [Google Scholar] [CrossRef]
  98. Rupérez, M.; Lorenzo, O.; Blanco-Colio, L.M.; Esteban, V.; Egido, J.; Ruiz-Ortega, M. Connective tissue growth factor is a mediator of angiotensin II-induced fibrosis. Circulation 2003, 108, 1499–1505. [Google Scholar] [CrossRef]
  99. Makino, K.; Makino, T.; Stawski, L.; Lipson, K.E.; Leask, A.; Trojanowska, M. Anti-connective tissue growth factor (CTGF/CCN2) monoclonal antibody attenuates skin fibrosis in mice models of systemic sclerosis. Arthritis Res. Ther. 2017, 19, 134. [Google Scholar] [CrossRef] [PubMed]
  100. Lisi, S.; D’Amore, M.; Sisto, M. ADAM17 at the interface between inflammation and autoimmunity. Immunol. Lett. 2014, 162, 159–169. [Google Scholar] [CrossRef] [PubMed]
  101. Schmidt-Arras, D.; Rose-John, S. Regulation of Fibrotic Processes in the Liver by ADAM Proteases. Cells 2019, 8, 1226. [Google Scholar] [CrossRef] [PubMed]
  102. Moss, M.L.; Minond, D. Recent Advances in ADAM17 Research: A Promising Target for Cancer and Inflammation. Mediat. Inflamm. 2017, 2017, 9673537. [Google Scholar] [CrossRef]
  103. Sisto, M.; Ribatti, D.; Lisi, S. ADAM 17 and Epithelial-to-Mesenchymal Transition: The Evolving Story and Its Link to Fibrosis and Cancer. J. Clin. Med. 2021, 10, 3373. [Google Scholar] [CrossRef] [PubMed]
  104. Stolarczyk, M.; Scholte, B.J. The EGFR-ADAM17 Axis in Chronic Obstructive Pulmonary Disease and Cystic Fibrosis Lung Pathology. Mediat. Inflamm. 2018, 2018, 1067134. [Google Scholar] [CrossRef] [PubMed]
  105. Sisto, M.; Lisi, S.; Lofrumento, D.D.; D’Amore, M.; Frassanito, M.A.; Ribatti, D. Sjögren’s syndrome pathological neovascularization is regulated by VEGF-A-stimulated TACE-dependent crosstalk between VEGFR2 and NF-κB. Genes Immun. 2012, 13, 411–420. [Google Scholar] [CrossRef]
  106. Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: Mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 26. [Google Scholar] [CrossRef] [PubMed]
  107. Engelman, J.A.; Luo, J.; Cantley, L.C. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat. Rev. Genet. 2006, 7, 606–619. [Google Scholar] [CrossRef] [PubMed]
  108. Revathidevi, S.; Munirajan, A.K. Akt in cancer: Mediator and more. Semin. Cancer Biol. 2019, 59, 80–91. [Google Scholar] [CrossRef] [PubMed]
  109. Wang, J.; Hu, K.; Cai, X.; Yang, B.; He, Q.; Wang, J.; Weng, Q. Targeting PI3K/AKT signalling for treatment of idiopathic pulmonary fibrosis. Acta Pharm. Sin. B 2022, 12, 18–32. [Google Scholar] [CrossRef]
  110. Meltzer, E.B.; Noble, P.W. Idiopathic pulmonary fibrosis. Orphanet J. Rare Dis. 2008, 3, 8. [Google Scholar] [CrossRef]
  111. Sun, Y.; Zhang, Y.; Chi, P. Pirfenidone suppresses TGFbeta1induced human intestinal fibroblasts activities by regulating proliferation and apoptosis via the inhibition of the Smad and PI3K/AKT signalling pathway. Mol. Med. Rep. 2018, 18, 3907–3913. [Google Scholar] [PubMed]
  112. Zhai, C.; Cheng, J.; Mujahid, H.; Wang, H.; Kong, J.; Yin, Y.; Li, J.; Zhang, Y.; Ji, X.; Chen, W. Selective inhibition of PI3K/Akt/mTOR signalling pathway regulates autophagy of macrophage and vulnerability of atherosclerotic plaque. PLoS ONE 2014, 9, e90563. [Google Scholar] [CrossRef] [PubMed]
  113. Liu, Y.M.; Nepali, K.; Liou, J.P. Idiopathic pulmonary fibrosis: Current status, recent progress, and emerging targets. J. Med. Chem. 2017, 60, 527–553. [Google Scholar] [CrossRef] [PubMed]
  114. Hewlett, J.C.; Kropski, J.A.; Blackwell, T.S. Idiopathic pulmonary fibrosis: Epithelial–mesenchymal interactions and emerging therapeutic targets. Matrix Biol. 2018, 71, 112–127. [Google Scholar] [CrossRef]
  115. Winters, N.I.; Burman, A.; Kropski, J.A.; Blackwell, T.S. Epithelial injury and dysfunction in the pathogenesis of idiopathic pulmonary fibrosis. Am. J. Med. Sci. 2019, 357, 374–378. [Google Scholar] [CrossRef]
  116. Pan, L.H.; Yamauchi, K.; Uzuki, M.; Nakanishi, T.; Takigawa, M.; Inoue, H.; Sawai, T. Type II alveolar epithelial cells and interstitial fibroblasts express connective tissue growth factor in IPF. Eur. Respir. J. 2001, 17, 1220–1227. [Google Scholar] [CrossRef]
  117. Khalil, N.; OConnor, R.N.; Flanders, K.C.; Unruh, H. TGF-beta(1), but not TGF-beta(2) or TGF-beta(3), is differentially present in epithelial cells of advanced pulmonary fibrosis: An immunohistochemical study. Am. J. Respir. Cell Mol. Biol. 1996, 14, 131–138. [Google Scholar] [CrossRef]
  118. Webb, G.J.; Hirschfield, G.M.; Krawitt, E.L.; Gershwin, M.E. Cellular and Molecular Mechanisms of Autoimmune Hepatitis. Annu. Rev. Pathol. 2018, 13, 247–292. [Google Scholar] [CrossRef]
  119. Du, Z.; Lin, Z.; Wang, Z.; Liu, D.; Tian, D.; Xia, L. SPOCK1 Overexpression Induced by Platelet-Derived Growth Factor-BB Promotes Hepatic Stellate Cell Activation and Liver Fibrosis through the Integrin α5β1/PI3K/Akt Signalling Pathway. Lab. Investig. 2020, 100, 1042–1056. [Google Scholar] [CrossRef]
  120. Falcon, B.L.; Barr, S.; Gokhale, P.C.; Chou, J.; Fogarty, J.; Depeille, P.; Miglarese, M.; Epstein, D.M.; McDonald, D.M. Reduced VEGF production, angiogenesis, and vascular regrowth contribute to the antitumor properties of dual mTORC1/mTORC2 inhibitors. Cancer Res. 2011, 71, 1573–1583. [Google Scholar] [CrossRef]
  121. Karar, J.; Maity, A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front. Mol. Neurosci. 2011, 4, 51. [Google Scholar] [CrossRef]
  122. Bartalucci, N.; Guglielmelli, P.; Vannucchi, A.M. Rationale for targeting the PI3K/Akt/mTOR pathway in myeloproliferative neoplasms. Clin. Lymphoma Myeloma Leuk. 2013, 13, S307–S309. [Google Scholar] [CrossRef]
  123. Bharadwaj, U.; Kasembeli, M.M.; Robinson, P.; Tweardy, D.J. Targeting Janus Kinases and Signal Transducer and Activator of Transcription 3 to Treat Inflammation, Fibrosis, and Cancer: Rationale, Progress, and Caution. Pharmacol. Rev. 2020, 72, 486–526. [Google Scholar] [CrossRef] [PubMed]
  124. 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]
  125. Czepiel, M.; Diviani, D.; Jaźwa-Kusior, A.; Tkacz, K.; Rolski, F.; Smolenski, R.T.; Siedlar, M.; Eriksson, U.; Kania, G.; Błyszczuk, P. Angiotensin II receptor 1 controls profibrotic Wnt/β-catenin signalling in experimental autoimmune myocarditis. Cardiovasc. Res. 2022, 118, 573–584. [Google Scholar] [CrossRef]
  126. Weintraub, R.G.; Semsarian, C.; Macdonald, P. Dilated cardiomyopathy. Lancet 2017, 390, 400–414. [Google Scholar] [CrossRef] [PubMed]
  127. Lu, H.; Zong, G.; Zhou, S.; Jiang, Y.; Chen, R.; Su, Z.; Wu, Y. Angiotensin II-C-C chemokine receptor2/5 axis-dependent monocyte/macrophage recruitment contributes to progression of experimental autoimmune myocarditis. Microbiol. Immunol. 2017, 61, 539–546. [Google Scholar] [CrossRef]
  128. Tomita, H.; Egashira, K.; Ohara, Y.; Takemoto, M.; Koyanagi, M.; Katoh, M.; Yamamoto, H.; Tamaki, K.; Shimokawa, H.; Takeshita, A. Early induction of transforming growth factor-beta via angiotensin II type 1 receptors contributes to cardiac fibrosis induced by long-term blockade of nitric oxide synthesis in rats. Hypertension 1998, 32, 273–279. [Google Scholar] [CrossRef]
  129. Zhao, Y.; Wang, C.; Wang, C.; Hong, X.; Miao, J.; Liao, Y.; Zhou, L.; Liu, Y. An essential role for Wnt/β-catenin signalling in mediating hypertensive heart disease. Sci. Rep. 2018, 8, 8996. [Google Scholar] [CrossRef] [PubMed]
  130. Działo, E.; Czepiel, M.; Tkacz, K.; Siedlar, M.; Kania, G.; Błyszczuk, P. WNT/β-Catenin Signalling Promotes TGF-β-Mediated Activation of Human Cardiac Fibroblasts by Enhancing IL-11 Production. Int. J. Mol. Sci. 2021, 22, 10072. [Google Scholar] [CrossRef]
  131. Shi, J.; Li, F.; Luo, M.; Wei, J.; Liu, X. Distinct Roles of Wnt/β-Catenin Signalling in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis. Mediat. Inflamm. 2017, 2017, 3520581. [Google Scholar] [CrossRef] [PubMed]
  132. 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. Signalling Pathway in Liver Fibrosis for Drug Options: An Update. J. Clin. Transl. Hepatol. 2021, 9, 960–971. [Google Scholar]
  133. Tan, K.; Xie, X.; Shi, W.; Miao, L.; Dong, X.; Yang, W.; Shao, C.; Zhao, H.; Wang, Y.; Wang, G.; et al. Deficiency of canonical Wnt/β-catenin signalling in hepatic dendritic cells triggers autoimmune hepatitis. Liver Int. 2020, 40, 131–140. [Google Scholar] [CrossRef]
  134. Liu, Y.; Wang, J.; Luo, S.; Zhan, Y.; Lu, Q. The roles of PPARγ and its agonists in autoimmune diseases: A comprehensive review. J. Autoimmun. 2020, 113, 102510. [Google Scholar] [CrossRef] [PubMed]
  135. Kersten, S.; Stienstra, R. The role and regulation of the peroxisome proliferator activated receptor alpha in human liver. Biochimie 2017, 136, 75–84. [Google Scholar] [CrossRef]
  136. Derosa, G.; Sahebkar, A.; Maffioli, P. The role of various peroxisome proliferator-activated receptors and their ligands in clinical practice. J. Cell. Physiol. 2018, 233, 153–161. [Google Scholar] [CrossRef] [PubMed]
  137. Dubois, V.; Eeckhoute, J.; Lefebvre, P.; Staels, B. Distinct but complementary contributions of PPAR isotypes to energy homeostasis. J. Clin. Investig. 2017, 127, 1202–1214. [Google Scholar] [CrossRef]
  138. Iglarz, M.; Touyz, R.M.; Viel, E.C.; Paradis, P.; Amiri, F.; Diep, Q.N.; Schiffrin, E.L. Peroxisome proliferator-activated receptor-alpha and receptor-gamma activators prevent cardiac fibrosis in mineralocorticoid-dependent hypertension. Hypertension 2003, 42, 737–743. [Google Scholar] [CrossRef] [PubMed]
  139. Kawai, T.; Masaki, T.; Doi, S.; Arakawa, T.; Yokoyama, Y.; Doi, T.; Kohno, N.; Yorioka, N. PPAR-gamma agonist attenuates renal interstitial fibrosis and inflammation through reduction of TGF-beta. Lab. Investig. 2009, 89, 47–58. [Google Scholar] [CrossRef]
  140. Kulkarni, A.A.; Thatcher, T.H.; Olsen, K.C.; Maggirwar, S.B.; Phipps, R.P.; Sime, P.J. PPAR-γ ligands repress TGFβ-induced myofibroblast differentiation by targeting the PI3K/Akt pathway: Implications for therapy of fibrosis. PLoS ONE 2011, 6, e15909. [Google Scholar] [CrossRef]
  141. Bobeica, C.; Niculet, E.; Tatu, A.L.; Craescu, M.; Vata, D.; Statescu, L.; Iancu, A.V.; Musat, C.L.; Draganescu, M.L.; Onisor, C.; et al. Old and new therapeutic strategies in systemic sclerosis (Review). Exp. Ther. Med. 2022, 23, 134. [Google Scholar] [CrossRef]
  142. Truchetet, M.E.; Brembilla, N.C.; Chizzolini, C. Current Concepts on the Pathogenesis of Systemic Sclerosis. Clin. Rev. Allergy Immunol. 2021, in press. [Google Scholar] [CrossRef]
  143. Bhattacharyya, S.; Wei, J.; Varga, J. Understanding fibrosis in systemic sclerosis: Shifting paradigms, emerging opportunities. Nat. Rev. Rheumatol. 2011, 8, 42–54. [Google Scholar] [CrossRef]
  144. Kohno, S.; Endo, H.; Hashimoto, A.; Hayashi, I.; Murakami, Y.; Kitasato, H.; Kojima, F.; Kawai, S.; Kondo, H. Inhibition of skin sclerosis by 15deoxy delta12,14-prostaglandin J2 and retrovirally transfected prostaglandin D synthase in a mouse model of bleomycin-induced scleroderma. Biomed. Pharmacother. 2006, 60, 18–25. [Google Scholar] [CrossRef]
  145. Wu, M.; Melichian, D.S.; Chang, E.; Warner-Blankenship, M.; Ghosh, A.K.; Varga, J. Rosiglitazone abrogates bleomycin-induced scleroderma and blocks profibrotic responses through peroxisome proliferator-activated receptor-gamma. Am. J. Pathol. 2009, 174, 519–533. [Google Scholar] [CrossRef]
  146. Kapoor, M.; McCann, M.; Liu, S.; Huh, K.; Denton, C.P.; Abraham, D.J.; Leask, A. Loss of peroxisome proliferator-activated receptor gamma in mouse fibroblasts results in increased susceptibility to bleomycin-induced skin fibrosis. Arthritis Rheum. 2009, 60, 2822–2829. [Google Scholar] [CrossRef]
  147. Kisseleva, T.; Brenner, D.A. Anti-fibrogenic strategies and the regression of fibrosis. Best Pract. Res. Clin. Gastroenterol. 2011, 25, 305–317. [Google Scholar] [CrossRef]
  148. Yu, Q.; Cheng, P.; Wu, J.; Guo, C. PPARγ/NF-κB and TGF-β1/Smad pathway are involved in the anti-fibrotic effects of levo-tetrahydropalmatine on liver fibrosis. J. Cell. Mol. Med. 2021, 25, 1645–1660. [Google Scholar] [CrossRef]
Figure 1. A schematic view of the main factors and signalling pathways involved in organs failure- dependent fibrosis.
Figure 1. A schematic view of the main factors and signalling pathways involved in organs failure- dependent fibrosis.
Ijms 24 09060 g001
Figure 2. TGF-β is the key modulator in the pathogenesis of fibrosis. Activation of latent TGF-β triggers fibrotic cascades that influence the differentiation of resident fibroblasts to myofibroblasts.
Figure 2. TGF-β is the key modulator in the pathogenesis of fibrosis. Activation of latent TGF-β triggers fibrotic cascades that influence the differentiation of resident fibroblasts to myofibroblasts.
Ijms 24 09060 g002
Figure 3. A schematic overview of TGF-β signalling pathways triggering the EMT process that contributes to fibrotic evolution in several autoimmune diseases.
Figure 3. A schematic overview of TGF-β signalling pathways triggering the EMT process that contributes to fibrotic evolution in several autoimmune diseases.
Ijms 24 09060 g003
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

Sisto, M.; Lisi, S. Towards a Unified Approach in Autoimmune Fibrotic Signalling Pathways. Int. J. Mol. Sci. 2023, 24, 9060. https://doi.org/10.3390/ijms24109060

AMA Style

Sisto M, Lisi S. Towards a Unified Approach in Autoimmune Fibrotic Signalling Pathways. International Journal of Molecular Sciences. 2023; 24(10):9060. https://doi.org/10.3390/ijms24109060

Chicago/Turabian Style

Sisto, Margherita, and Sabrina Lisi. 2023. "Towards a Unified Approach in Autoimmune Fibrotic Signalling Pathways" International Journal of Molecular Sciences 24, no. 10: 9060. https://doi.org/10.3390/ijms24109060

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