Acessibilidade / Reportar erro

Update on etiopathogenesis of systemic sclerosis

Abstracts

Systemic Sclerosis (SSc) is an autoimmune disease of multifactorial etiology, triggered by a combination of genetic and environmental factors. Its varied clinical expression results from the complex physiopathogenic interaction of three main elements: proliferative vasculopathy, immune dysregulation and abnormal deposition and remodeling of the extracellular matrix (ECM), of which the characteristic disease fibrosis is the result. Early physiopathogenic events appear to be endothelial injury and imbalance in vascular repair with the activation of endothelial cells, the immune system and platelets, with the release of multiple mediators such as TH2 proinflammatory cytokines and growth factors, triggering a sequence of simultaneous or cascading events that involve several intracellular signaling pathways. The most important result of these events is the hyperactivation of fibroblasts, the main effector cells of fibrosis, which will then produce large amounts of ECM constituents and secrete multiple growth factors and cytokines that perpetuate the process. In this article we review the main factors potentially involved in the etiology of SSc and reexamine the current knowledge about the most important mechanisms involved in the development of lesions that are characteristic of the disease. A better understanding of these physiopathogenic mechanisms will help identify potential therapeutic targets, which may result in advances in the management of this complex and debilitating disease.

Systemic sclerosis; Etiopathogenesis; Immune system; Angiopathy; Extracellular matrix


A Esclerose Sistêmica (ES) é uma doença autoimune de etiologia multifatorial, desencadeada pela combinação de fatores genéticos e ambientais. Sua variada expressão clínica resulta da complexa interação fisiopatogênica de três elementos principais: a vasculopatia proliferativa, a desregulação imunológica e a deposição e remodelamento anormais da matriz extracelular (MEC), da qual resulta a fibrose característica da doença. Eventos fisiopatogênicos precoces parecem ser a lesão endotelial e o desequilíbrio no reparo vascular, com a ativação de células endoteliais, do sistema imune e das plaquetas, com a liberação de múltiplos mediadores, como as citocinas proinflamatórias TH2 e os fatores de crescimento, desencadeando uma sequência de eventos simultâneos ou em cascata que envolve diversas vias de sinalização intracelular. O resultado mais importante desses eventos é a hiperativação dos fibroblastos, as principais células efetoras da fibrose, as quais passam a produzir grandes quantidades de constituintes da MEC e a secretar múltiplos fatores de crescimento e citocinas que perpetuam o processo. Neste artigo apresentamos uma revisão dos principais fatores potencialmente implicados na etiologia da ES e revisitamos os conhecimentos atuais sobre os mais importantes mecanismos envolvidos no desenvolvimento das lesões características da doença. O melhor entendimento desses mecanismos fisiopatogênicos possibilita identificar potenciais alvos terapêuticos, o que pode resultar em avanços no manejo dessa complexa e debilitante doença.

Esclerose sistêmica; Etiopatogênese; Sistema imune; Angiopatia; Matriz extracelular


REVIEW ARTICLE

IDivision of Rheumatology, Department of Internal Medicine, Hospital Universitário da Universidade Federal de Santa Catarina, Santa Catarina, SC, Brazil

IIDivision of Pneumology, Department of Internal Medicine, Hospital Universitário da Universidade Federal de Santa Catarina, Santa Cata rina, SC, Brazil

ABSTRACT

Systemic Sclerosis (SSc) is an autoimmune disease of multifactorial etiology, triggered by a combination of genetic and environmental factors. Its varied clinical expression results from the complex physiopathogenic interaction of three main elements: proliferative vasculopathy, immune dysregulation and abnormal deposition and remodeling of the extracellular matrix (ECM), of which the characteristic disease fibrosis is the result. Early physiopathogenic events appear to be endothelial injury and imbalance in vascular repair with the activation of endothelial cells, the immune system and platelets, with the release of multiple mediators such as TH2 proinflammatory cytokines and growth factors, triggering a sequence of simultaneous or cascading events that involve several intracellular signaling pathways.

The most important result of these events is the hyperactivation of fibroblasts, the main effector cells of fibrosis, which will then produce large amounts of ECM constituents and secrete multiple growth factors and cytokines that perpetuate the process. In this article we review the main factors potentially involved in the etiology of SSc and reexamine the current knowledge about the most important mechanisms involved in the development of lesions that are characteristic of the disease. A better understanding of these physiopathogenic mechanisms will help identify potential therapeutic targets, which may result in advances in the management of this complex and debilitating disease.

Keywords: Systemic sclerosis; Etiopathogenesis; Immune system; Angiopathy; Extracellular matrix

Introduction

Systemic sclerosis (SSc) is a connective tissue disease of autoimmune origin, characterized by vascular alterations and progressive skin and visceral fibrosis affecting mainly the lungs, gastrointestinal tract, heart, and kidneys.1 The disease is extremely heterogeneous in its phenotypic expression, and its prognosis is determined by the predominant clinical manifestations, especially regarding visceral involvement.2-4

Despite advances in knowledge about the mechanisms responsible for the disease onset, there is still a lack of understanding regarding the pathogenic process is initiated and undergoes intensification in each subgroup of patients, leading to different clinical expressions. As in other autoimmune diseases, especially those that are not organ specifi c, it is known that the process results from the interaction of multiple factors, both individual and those related to the surrounding environment.5

However, the mechanisms that trigger the disease in genetically susceptible individuals have not yet been elucidated. In this article, the main factors potentially implicated in the etiopathogenesis of SSc are reviewed, as well as the current knowledge on the most important mechanisms involved in the development of SSc characteristic lesions, and the potential treatments directed at molecular and cell targets that have emerged from recent research are listed.

SSc etiology: environmental factors

In the great majority of cases, SSc is considered to be idiopathic. However, in some situations, environmental factors likely play an important role in its development.These associations have been recognized for many decades, and the most important are those related to exposure to silica and organic solvents.The combined relative risk estimate (CRRE) for silica, calculated in a meta-analysis, was 3.20 (95% CI: 1.89-5.43) for men. Regarding exposure to organic solvents, the CRRE found in a meta-analysis was 2.91 (95% CI: 1.60-5.30),6 and, according to another more recent meta-analysis,7 the latter is a factor associated with increased risk of SSc in men.

The possible association between silicone breast implants and connective tissue disorders, especially scleroderma, has been the subject of great controversy, generated by case reports that suggested this association.8 However, a meta-analysis9 and a recent and comprehensive systematic review10 of the available epidemiological evidence did not confi rm such association.

Other environmental factors that have been implicated in the etiology of SSc are exposures to infectious agents, especially viruses. Among them, parvovirus B19 is an example of a very prevalent infection in patients with SSc.11 Regarding the Epstein-Barr virus, it has been demonstrated that specific adaptive immunity against the virus (the activation of suppressor T lymphocytes) was deficient in scleroderma patients.12 Cytomegalovirus (CMV) infects endothelial cells and monocytes, leading to the production of profi brotic cytokines, causing vascular lesions and activating fibroblasts, and may therefore trigger pathogenic processes associated with SSc development.13

An association between positivity to anti-CMV antibodies and prevalence of specific autoantibodies in patients with SSc has been reported.14 These data suggest an association between infectious and environmental factors and the disease, but more studies are still necessary to determine the possible causal role of external agents in triggering SSc.

SSc etiology: hormonal and genetic factors

Considering the marked predominance of SSc in women,(7:1) a hypothesis was raised that hormonal factors are relevant to the disease development. However, this has not been proven. For instance, an increased incidence of the disease in women who use oral contraceptives has not been observed.15

Among the host factors involved in the etiopathogenesis of autoimmune diseases, genetic susceptibility is perhaps the most relevant, and therefore has been the target of extensive research. In SSc, as in almost all other systemic rheumatic diseases, polymorphisms of the main histocompatibility system (human leukocyte antigen [HLA]) have been associated to the disease development, with the main associations being with class II HLA antigens.

A recent study involving a fairly signifi cant sample showed a positive association of haplotypes HLA-DRB1_1104, DQA1_0501, DQB1_0301 and, in contrast, the protective factor of haplotypes HLA-DRB1_0701, DQA1_0201, DQB1_0202, and DRB1_1501 in whites and Hispanics with SSc; however, in blacks, the association was with HLA-DRB1_0804, DQA1_0501, and DQB1_0301 alleles.16 Moreover, the association of the HLA system with autoantibody specificity in SSc has also been studied. The same abovementioned study demonstrated the association of anti-Scl70 antibodies with HLADPB1_1301 antigens, of anti-centromere antibodies (ACA) with HLADQB1_0501 and DQB1_26, and of anti-RNA polymerase antibodies with HLA-DRB1_0404, DRB1_11, and DQB1_03 alleles(16), which illustrates the multiple influences that this gene system can exert on SSc and its clinical and immunological profile.

Many other genes have been implicated in SSc susceptibility, mostly those encoding proteins responsible for the regulation and transduction of signals that comprise the autoimmunity and inflammation mechanisms involved in the pathogenesis of SSc. One of the main associations found in different populations was with the gene encoding for the signal transducer and activator of transcription 4 (STAT4),17 which promotes the differentiation of type-1 T helper lymphocytes and negatively regulates the Th2 type.

The association of B-cell scaffold protein with ankyrin repeats (BANK1), which binds the B-cell receptor to intracellular signaling proteins such as kinases,18 and with the interferon regulatory factor five (IRF-5), regulator of transcription of type I interferon genes, were also confi rmed.19 Moreover, positive associations have also been reported of SSc with candidate interleukin 23 (IL-23R) receptor genes involved in expression of the IL-23, a cytokine which expands the population of TH17 lymphocytes, but SSc has only been associated with positivity to anti-scleroderma 70 (anti-Scl70) autoantibody, and it acts as a protective factor in relation to pulmonary hypertension.20 Another gene related to SSc susceptibility is the connective tissue growth factor (CTGF), which induces cell proliferation, increasing production of ECM and chemotaxis of mesenchymal cells.21

Recently, it has been observed that interactions between genes are also important, such as the apparent additive effect in SSc exerted by the simultaneous presence of STAT4, IRF-5, and BANK1 on the susceptibility and the development of the diffuse clinical form and pulmonary fi brosis.17,22 Another interesting finding was the sharing of genetic risk factors between different autoimmune diseases, exemplified by the protein tyrosine phosphatase 22 (PTPN22) non-receptor gene, involved in SSc susceptibility23 as well as in diabetes mellitus type I,24 systemic lupus erythematous (SLE),25 and rheumatoid arthritis (RA).(25) Other shared genes, such as BANK1, IRF5, and STAT4 have also been reported as involved in SLE and RA susceptibility.26-28

Another genetic factor of SSc susceptibility was recently demonstrated in white European populations.29 It is the macrophage migration inhibitory factor (MIF)-173, a cytokine with immunoregulatory functions and a mediator of innate and adaptive immunity, which has been implicated in the pathogenesis of vasculopathy in SSc.(30) In addition to the association with susceptibility, the MIF-173C allele was associated with the diffuse form of the disease.29

Physiopathogenic mechanisms of systemic sclerosis

The current accumulated knowledge regarding the physiopathogenesis of SSc derives from extensive research in different areas, and indicates that the pathological process arises from the complex interrelation between three main components: vascular dysfunction, innate and adaptive immunity dysregulation, and excess activation of fi broblasts and related cells, which culminates in the development of fibrosis (Fig. 1).31

Proliferative vasculopathy

The occurrence of Raynaud's phenomenon in almost all patients with SSc, with high severity in many of them, demonstrates the importance of vasculopathy in the pathogenic context of the disease. It is believed that the vasculopathy may originate after injury and activation of endothelial cells by unknown factors.32 The endothelial activation has been demonstrated in several studies that detected high plasma levels and increased expression of von Willebrand factor, intercellular adhesion molecules, endothelin-1, and thrombomodulin, which also represent evidence of excessive apoptosis of endothelial cells.33-35

Chronic endothelial injury results in platelet adhesion and activation of the fibrinolytic system, as well as in increased vascular permeability and leukocyte adhesion to the vascular wall.34 Increased levels of endothelin-1, a potent vasoconstrictor, have been detected in plasma and bronchoalveolar lavage fluid of patients with SSc.36 This protein is responsible for increasing leukocyte adhesion to the endothelium, promoting migration and proliferation of smooth muscle cells into the intima layer of vessels and activating fi broblasts.

This process promotes the synthesis and deposition of ECM molecules, leading to fibrosis with loss of elasticity and progressive reduction of the vascular lumen, causing progressive tissue necrosis and hypoxia.37

Tissue hypoxia promotes angiogenesis (formation of new vessels from remaining functional vessels) and vasculogenesis (formation of new vessels from endothelial progenitor cells),38 in order to restore cell oxygen supply. Due to multiple causes not yet understood, these processes are defi cient in SSc, and there is an imbalance between angiogenic and angiostatic factors and ineffective vascular repair.39-40

The result is the formation of morphologically aberrant and dysfunctional capillaries, which can be viewed in the nailfold capillaroscopy examination. It is now known that tissue hypoxia present in SSc plays an important pathogenic role in the disease through different factors, such as the production of hypoxia-induced factor (HIF-1), whose production and deficient regulation is ineffective in restoring normal blood pressure levels of oxygen. This defi ciency creates a vicious circle, with increased activation of immune cells, increased activation of fibroblasts, increased levels of transforming growth factor β (TGFβ), and ECM deposition, which in turn aggravate the hypoxic environment. Furthermore, the increase in oxidative stress caused by hypoxia has a proinflammatory and profibrotic effect, exerted by reactive oxygen species (ROS).41

An additional factor that contributes to microvasculopathy in SSc is the pericytes, cells very close to the endothelium that normally inhibit cell migration and vascular proliferation. These cells are hyperplastic and hyperactivated in SSc, inhibiting the process of angiogenesis and transdifferentiating into myofibroblasts, producing excess ECM, which accumulates in the perivascular area and aggravates the proliferative vasculopathy of the disease.38 Table 1 summarizes the most relevant physiopathological events in relation to SSc vasculopathy.

Autoimmunity and infl ammation

Another important factor implicated in the pathogenesis of SSc is the involvement of both humoral and cellular immune system, whose mediators are the connection between vascular disease and tissue fibrosis, as they are involved in both processes.32 The involvement of the humoral immune system is demonstrated by the presence of B lymphocyte infiltrates with expression of chronic activation markers (CD19, CD85), which have been detected in patients with SSc skin using DNA microarray techniques.42 The homeostasis of circulating B lymphocytes is altered, expanding the population of non-activated cells (naïve) and reducing the number, but increasing the activation of memory B cells.43 This knowledge has led to the use of therapies directed against B cells (monoclonal antibodies belimumab and rituximab) in the treatment of patients with SSc, as well as to the blocking of the costimulation between the latter and T lymphocytes.44

The specific autoantibodies, which represent one of the attributes of the disease, characterize its clinical forms and have important prognostic associations; they are the most obvious expression of the humoral immune system involvement in the etiopathogenesis of SSc. The main autoantibodies whose pathogenic potential has been established, in addition Scl70 and ACA, are those directed against endothelial cells and against the platelet-derived growth factor (PDGF) receptor.32

ACA are characteristic of limited SSc, which is generally more benign than the diffuse form, but is associated with later development of pulmonary hypertension, one of the most severe complications of the disease, as well as digital ulcers and important gastrointestinal involvement. Patients with limited SSc may have anti-Th/To antibodies (more frequent in men, with interstitial pulmonary involvement and earlier mortality),45 anti-PM/Scl antibodies (associated with myositis, calcinosis, acro-osteolysis and interstitial lung disease),46 and the anti-U1RNP antibodies (that characterize overlapping forms between SSc, SLE, and myositis).47

Conversely, the presence of anti-Scl-70 is associated with the diffuse form of SSc, with interstitial pulmonary involvement and increased risk of renal crisis, especially in the early years of the disease. Musculoskeletal complications, such as flexion contractures of the fingers, are common and very disabling.47 More rarely, the anti-RNA polymerase III (POL3) antibodies occur in the diffuse form, whose patients have a high frequency of renal crisis, rapidly progressive skin fibrosis, and little pulmonary and gastrointestinal involvement.48 The diffuse form can also show the presence of anti-U3RNP (fi brillarin), which is more common in black individuals and has worse prognosis, with severe pulmonary fibrosis, arterial pulmonary hypertension, pigmentation alterations, and joint contractures.49

The pathogenic role of cell immunity, represented mainly by T lymphocytes, is evidenced in SSc by the increase in endothelial transmigration of T CD4 + cells originating from perivascular infl ammatory infiltrates and oligoclonal activation of these cells, resulting in profibrotic cytokine production.50 The oligoclonal activation of T-lymphocytes was demonstrated by the increased levels of soluble IL-2 receptor in the serum of SSc patients, which showed a strong association with skin involvement (Rodnan skin score) in the disease.51

The predominant production of these cytokines (IL-4, IL-5, IL-6, IL-10, IL-13, and monocyte chemoattractant protein-1 [MCP-1]) results from an imbalance between the profiles of T helper type 1 (TH1) and type 2 (TH2) cells, derived from TH2- cells, which is the predominant profile in SSc and promotes worsening of tissue fi brosis.52 Current knowledge allows for the inference that the processes triggered by profibrotic cytokines derived from the interaction between T lymphocytes and fibroblasts play a key role in the development of fibrosis (Table 2).

Development of fi brosis: fi broblast activation

The third key pathogenic mechanism involved in SSc, and the most representative of the disease, is fi brosis, which lends its name to the disease and affects the skin, lungs, heart, digestive tract, kidneys, and musculoskeletal system. The extent of skin fibrosis is associated with mortality by SSc, as recently demonstrated both in the European and Brazilian populations.3-4

Fibrosis results from complex interactions between multiple concurrent or cascading processes, in which dozens of cell mediators, cytokines and their receptors, chemokines, growth factors, and intracellular signaling molecules are present (Table 3). The result of this interaction is the excessive production and accumulation of insoluble material in the tissues that constitutes the ECM. Fibrillar collagens types I and II, type VII collagen, and elastin and fi brillin fi brils are mainly found in ECM, as well as enzymes that promote the production of collagen crosslinks.53

Regarding fibrosis cell mediators, the fibroblast is the cell primarily responsible for the production and remodeling of the ECM, and its main physiological importance lies in the healing process of connective tissue injuries, having a self-limited action, depending on the injury extent.53 In SSc, as in other fibroproliferative diseases, there is a state in which the fibroblast is permanently hyperactivated, with altered expression of genes that determine ECM overproduction.

In SSc, one of the mechanisms responsible for this hyperactivation is the mechanotransduction abnormality, in which fibroblasts and myofibroblasts would be able to perceive tensional forces and change their intracellular signaling in response, leading to altered homeostasis and remodeling of the ECM.54 In addition to the fi broblasts, myofibroblasts also participate in the process of fi brosis formation in SSc. These contractile and ECM-producing cells originate from the transdifferentiation of fi broblasts, epithelial cells, and pericytes.55 Unlike the normal healing process, in which myofibroblasts occur only transiently in granulation tissue, in SSc these cells become permanent, producing areas of fibrosis with contracture of the ECM.56

The bone marrow also contributes to the increase of the amount of profibrotic cells, with the release of pluripotent mesenchymal cells and fibroblast progenitors (fi brocytes). The latter physiologically replenish the population of fi broblasts maintaining tissue homeostasis, but they also exert a pathogenic role in SSc. Fibrocyte precursors migrate and accumulate in tissues by gradient of chemokine receptors (CRs), such as CCR3, CCR5, and CXCR4.32 These receptors and their ligands have been found at high levels in the skin of patients with SSc.57 Furthermore, tissue-infi ltrating fi brocytes have also been implicated in the pathogenesis of pulmonary fi brosis disease.58

The main effector proteins involved in the fi brosis mechanisms in SSc are the cytokines, chemokines, and the family of ECM growth factors, which induce fibrogenic cell response by increasing the expression of genes encoding ECM constituents.59 Profibrotic cytokines play a pathogenic role in SSc. The best-known and most often studied cytokine regarding its mechanisms in fibrosis is IL-4, which stimulates the synthesis of TGFβ and CTGF; it also promotes the migration, proliferation, and synthesis of collagen by fi broblasts. High levels of IL-4 were found in serum and tissues of patients with SSc, and a larger number of T cells that produce this cytokine were detected in these patients.60-61 It is also known that IL-13 induces the expression of type I collagen gene and that its levels are elevated in patients with SSc, but its possible role in the induction of fibrosis is not well established.60 Very recently, a study found increased levels of IL-6 in serum and tissues of patients with diffuse SSc, which was associated with more severe skin involvement and increased mortality after three years,62 suggesting that IL-6 antagonism would be a promising therapeutic target in this situation.

Perhaps the most important of all effector proteins of fi brosis is TGFβ, considered to be the orchestrator of the physiological process of tissue repair and known to be involved in the development of pathological fibrosis, as in SSc.59 TGFβ is produced by fi broblasts, T cells, monocytes, and platelets, and it is released as a latent complex that is activated by tissue injury to its biologically active form within the ECM, exerting its function by binding to a specific receptor (TGFβRII) on the surface of different cell types.37

After this binding, an intracellular signaling cascade starts, which results in the expression of target genes such as type I collagen, CTGF, and plasminogen activator inhibitor-1, among others.63 The main control of TGFβ intracellular signaling system is exercised by the SMAD system (SMAD2 and SMAD3 activators, as well as endogenous inhibitor SMAD7 and its cofactors), whose dysregulation results in increased fibrogenesis, a phenomenon already demonstrated in SSc.64 Recently, other regulatory mechanisms of TGFβ were studied and their importance was demonstrated in the pathogenesis of SSc, such as c-Abl kinase (c-Abelson, a tyrosine kinase implicated in the pathogenesis of chronic myeloid leukemia), a potent regulator of the TGFβ-induced profi brotic response in fi broblasts.65

Imatinib, an inhibitor of c-Abl, reduces fi brotic responses in animal models and have shown to be promising in the treatment of patients with SSc.44,66 Also relevant as mediators of profibrotic cell response to TGFβ are genes from the early growth response (EGR) family, particularly EGR-1, whose expression is induced by chemical or mechanical tissue injury and whose levels are elevated in skin lesions of SSc, as demonstrated in a murine model of bleomycininduced scleroderma.67

Another relevant mechanism in the process of fi brosis that has recently been demonstrated, but whose role still requires elucidation, is the Wnt-β-catenin, an important intracellular signaling pathway in the embryonic period during organogenesis whose posterior and aberrant reactivation is associated with disease (cancer, for instance) by increasing fibroblast activity and promoting transdifferentiation. In biopsies of scleroderma patients, this pathway was activated and found responsible for significant production of fi brosis and lipoatrophy.68

In addition to TGFβ, other growth factors are important in the pathogenesis of SSc, such as CTGF, whose effects are similar to those of TGFβ and whose levels are increased in lesions of patients with SSc. Its expression is stimulated by TGFβ itself, by hypoxia, and by ET-1.37 PDGF is produced by fibroblasts, platelets, macrophages, and endothelial cells, with important mitogen and chemoattractant activity for fi broblasts, inducing their production of collagen, fi bronectin, and proteoglycans, and the release of profi brotic mediators IL-6 and MCP-1. This is another growth factor implicated in the interaction with TGFβ and CTGF, which was detected at high levels in the lungs of patients with SSc.69

In addition to all the aforementioned mechanisms, which help to promote excessive fibrogenesis, in SSc there are also abnormalities in the physiological counterregulation systems that normally prevent the occurrence of fi brosis. This was demonstrated in the case of SMAD7, signaling molecules that exert an antifibrotic role through negative regulation of TGFβ action.64

Searching for antifibrotic mechanisms that could be explored for therapeutic purposes in SSc, the peroxisome proliferator activated receptor-gamma (PPAR-ϒ) was identifi ed. This molecule is a key hormonal nuclear receptor in lipid and glucose metabolism, which has been increasingly associated with ECM remodeling and fibrosis, capable of modulating TGFβ signaling and mesenchymal cell plasticity. It was found that this receptor is involved in an important antifibrotic mechanism, and its expression and activity were found to be reduced in patients with SSc.70 Natural or synthetic PPAR-ϒ agonists, such as the antidiabetic drugs pioglitazone or rosiglitazone, or tissue stimulation of its expression could, therefore, play a role in the treatment of fi brosis. Table 3 summarizes the elements involved in the physiopathological process of establishment fibrosis in SSc.

In spite of the fact that SSc is considered to be incurable, in recent years intensive research has led to the identifi cation of antifibrotic cellular and molecular targets and the development of therapeutic options directed against them. Table 4 lists some treatments aimed at targets (mostly discussed above) that have emerged from in vitro studies, animal models, and early trials in humans.44 A better understanding of the physiopathological mechanisms of SSc has shed light on the factors that most likely initiate the process and lead to its perpetuation.

Considering that the vascular lesion and its defective repair are known early events, it is possible that the clinical expression of the disease depends more on the magnitude of vascular changes and its chronicity than on the triggering event itself.41 Therefore, efforts are needed in order to diagnose the early establishment of vasculopathy and to institute treatment in order to halt the progression of the process. In addition, the elucidation of the molecular mechanisms involved in SSc may contribute to the identifi cation of other potential targets to be assessed in the search for more specific and effective treatments for this severe and debilitating disease.

Conflicts of interest

The authors declare no conflicts of interest.

REFERENCES

  • 1
    LeRoy EC, Black C, Fleischmajer R, Jablonska S, Krieg T, Medsger TA, Jr., et al. Scleroderma (systemic sclerosis): classification, subsets and pathogenesis. J Rheumatol. 1988 Feb;15(2):202-5.
  • 2
    Steen VD, Medsger TA. Changes in causes of death in systemic sclerosis, 1972-2002. Ann Rheum Dis. 2007 Jul;66(7):940-4.
  • 3
    Sampaio-Barros PD, Bortoluzzo AB, Marangoni RG, Rocha LF, Del Rio AP, Samara AM, et al. Survival, causes of death, and prognostic factors in systemic sclerosis: analysis of 947 Brazilian patients. J Rheumatol. 2012 Oct;39(10):1971-8.
  • 4
    Tyndall AJ, Bannert B, Vonk M, Airo P, Cozzi F, Carreira PE, et al. Causes and risk factors for death in systemic sclerosis: a study from the EULAR Scleroderma Trials and Research (EUSTAR) database. Ann Rheum Dis. 2010 Oct;69(10):1809-15.
  • 5
    Tan FK. Systemic sclerosis: the susceptible host (genetics and environment). Rheum Dis Clin North Am. 2003 May;29(2):211-37.
  • 6
    Aryal BK, Khuder SA, Schaub EA. Meta-analysis of systemic sclerosis and exposure to solvents. Am J Ind Med. 2001 Sep;40(3):271-4.
  • 7
    Kettaneh A, Al Moufti O, Tiev KP, Chayet C, Toledano C, Fabre B, et al. Occupational exposure to solvents and genderrelated risk of systemic sclerosis: a metaanalysis of casecontrol studies. J Rheumatol. 2007 Jan;34(1):97-103.
  • 8
    Levy Y, Rotman-Pikielny P, Ehrenfeld M, Shoenfeld Y. Silicone breast implantation-induced scleroderma: description of four patients and a critical review of the literature. Lupus. 2009 Nov;18(13):1226-32.
  • 9
    Janowsky EC, Kupper LL, Hulka BS. Meta-analyses of the relation between silicone breast implants and the risk of connective-tissue diseases. N Engl J Med. 2000 Mar 16;342(11):781-90.
  • 10
    Lipworth L, Holmich LR, McLaughlin JK. Silicone breast implants and connective tissue disease: no association. Semin Immunopathol. 2011 May;33(3):287-94.
  • 11
    Zakrzewska K, Corcioli F, Carlsen KM, Giuggioli D, Fanci R, Rinieri A, et al. Human parvovirus B19 (B19V) infection in systemic sclerosis patients. Intervirology. 2009;52(5):279-82.
  • 12
    Kahan A, Menkes CJ, Amor B. Defective Epstein-Barr virus specific suppressor T cell function in progressive systemic sclerosis. Ann Rheum Dis. 1986 Jul;45(7):553-60.
  • 13
    Radic M, Martinovic Kaliterna D, Radic J. Infectious disease as aetiological factor in the pathogenesis of systemic sclerosis. Neth J Med. 2010 Nov;68(11):348-53.
  • 14
    Neidhart M, Kuchen S, Distler O, Bruhlmann P, Michel BA, Gay RE, et al. Increased serum levels of antibodies against human cytomegalovirus and prevalence of autoantibodies in systemic sclerosis. Arthritis Rheum. 1999 Feb;42(2):389-92.
  • 15
    Mayes MD. Epidemiologic studies of environmental agents and systemic autoimmune diseases. Environ Health Perspect. 1999 Oct;107 Suppl 5:743-8.
  • 16
    Arnett FC, Gourh P, Shete S, Ahn CW, Honey RE, Agarwal SK, et al. Major histocompatibility complex (MHC) class II alleles, haplotypes and epitopes which confer susceptibility or protection in systemic sclerosis: analyses in 1300 Caucasian, African-American and Hispanic cases and 1000 controls. Ann Rheum Dis. 2010 May;69(5):822-7.
  • 17
    Dieude P, Guedj M, Wipff J, Ruiz B, Hachulla E, Diot E, et al. STAT4 is a genetic risk factor for systemic sclerosis having additive effects with IRF5 on disease susceptibility and related pulmonary fibrosis. Arthritis Rheum. 2009 Aug;60(8):2472-9.
  • 18
    Rueda B, Gourh P, Broen J, Agarwal SK, Simeon C, Ortego-Centeno N, et al. BANK1 functional variants are associated with susceptibility to diffuse systemic sclerosis in Caucasians. Ann Rheum Dis. 2010 Apr;69(4):700-5.
  • 19
    Dieude P, Guedj M, Wipff J, Avouac J, Fajardy I, Diot E, et al. Association between the IRF5 rs2004640 functional polymorphism and systemic sclerosis: a new perspective for pulmonary fibrosis. Arthritis Rheum. 2009 Jan;60(1):225-33.
  • 20
    Agarwal SK, Gourh P, Shete S, Paz G, Divecha D, Reveille JD, et al. Association of interleukin 23 receptor polymorphisms with anti-topoisomerase-I positivity and pulmonary hypertension in systemic sclerosis. J Rheumatol. 2009 Dec;36(12):2715-23.
  • 21
    Fonseca C, Lindahl GE, Ponticos M, Sestini P, Renzoni EA, Holmes AM, et al. A polymorphism in the CTGF promoter region associated with systemic sclerosis. N Engl J Med. 2007 Sep 20;357(12):1210-20.
  • 22
    Dieude P, Wipff J, Guedj M, Ruiz B, Melchers I, Hachulla E, et al. BANK1 is a genetic risk factor for diffuse cutaneous systemic sclerosis and has additive effects with IRF5 and STAT4. Arthritis Rheum. 2009 Nov;60(11):3447-54.
  • 23
    Dieude P, Guedj M, Wipff J, Avouac J, Hachulla E, Diot E, et al. The PTPN22 620W allele confers susceptibility to systemic sclerosis: findings of a large case-control study of European Caucasians and a meta-analysis. Arthritis Rheum. 2008 Jul;58(7):2183-8.
  • 24
    Onengut-Gumuscu S, Ewens KG, Spielman RS, Concannon P. A functional polymorphism (1858C/T) in the PTPN22 gene is linked and associated with type I diabetes in multiplex families. Genes Immun. 2004 Dec;5(8):678-80.
  • 25
    Orozco G, Sanchez E, Gonzalez-Gay MA, Lopez-Nevot MA, Torres B, Caliz R, et al. Association of a functional singlenucleotide polymorphism of PTPN22, encoding lymphoid protein phosphatase, with rheumatoid arthritis and systemic lupus erythematosus. Arthritis Rheum. 2005 Jan;52(1):219-24.
  • 26
    Remmers EF, Plenge RM, Lee AT, Graham RR, Hom G, Behrens TW, et al. STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med. 2007 Sep 6;357(10):977-86.
  • 27
    Graham RR, Kyogoku C, Sigurdsson S, Vlasova IA, Davies LR, Baechler EC, et al. Three functional variants of IFN regulatory factor 5 (IRF5) define risk and protective haplotypes for human lupus. Proc Natl Acad Sci U S A. 2007 Apr 17;104(16):6758-63.
  • 28
    Orozco G, Abelson AK, Gonzalez-Gay MA, Balsa A, Pascual-Salcedo D, Garcia A, et al. Study of functional variants of the BANK1 gene in rheumatoid arthritis. Arthritis Rheum. 2009 Feb;60(2):372-9.
  • 29
    Bossini-Castillo L, Simeon CP, Beretta L, Vonk MC, Callejas-Rubio JL, Espinosa G, et al. Confirmation of association of the macrophage migration inhibitory factor gene with systemic sclerosis in a large European population. Rheumatology (Oxford). 2011 Nov;50(11):1976-81.
  • 30
    Selvi E, Tripodi SA, Catenaccio M, Lorenzini S, Chindamo D, Manganelli S, et al. Expression of macrophage migration inhibitory factor in diffuse systemic sclerosis. Ann Rheum Dis. 2003 May;62(5):460-4.
  • 31
    Denton CP, Black CM, Abraham DJ. Mechanisms and consequences of fibrosis in systemic sclerosis. Nat Clin Pract Rheumatol. 2006 Mar;2(3):134-44.
  • 32
    Kahaleh MB. Raynaud phenomenon and the vascular disease in scleroderma. Curr Opin Rheumatol. 2004 Nov;16(6):718-22.
  • 33
    Scheja A, Akesson A, Geborek P, Wildt M, Wollheim CB, Wollheim FA, et al. Von Willebrand factor propeptide as a marker of disease activity in systemic sclerosis (scleroderma). Arthritis Res. 2001;3(3):178-82.
  • 34
    Cerinic MM, Valentini G, Sorano GG, D'Angelo S, Cuomo G, Fenu L, et al. Blood coagulation, fibrinolysis, and markers of endothelial dysfunction in systemic sclerosis. Semin Arthritis Rheum. 2003 Apr;32(5):285-95.
  • 35
    McHugh NJ, Distler O, Giacomelli R, Riemekasten G. Non organ based laboratory markers in systemic sclerosis. Clin Exp Rheumatol. 2003;21(3 Suppl 29):S32-8.
  • 36
    Cambrey AD, Harrison NK, Dawes KE, Southcott AM, Black CM, du Bois RM, et al. Increased levels of endothelin-1 in bronchoalveolar lavage fluid from patients with systemic sclerosis contribute to fibroblast mitogenic activity in vitro. Am J Respir Cell Mol Biol. 1994 Oct;11(4):439-45.
  • 37
    Wei J, Bhattacharyya S, Tourtellotte WG, Varga J. Fibrosis in systemic sclerosis: Emerging concepts and implications for targeted therapy. Autoimmunity Reviews. [doi: DOI: 10.1016/j.autrev.2010.09.015]. 2011;10(5):267-75.
  • 38
    Muller-Ladner U, Distler O, Ibba-Manneschi L, Neumann E, Gay S. Mechanisms of vascular damage in systemic sclerosis. Autoimmunity. 2009 Nov;42(7):587-95.
  • 39
    Koch AE, Distler O. Vasculopathy and disordered angiogenesis in selected rheumatic diseases: rheumatoid arthritis and systemic sclerosis. Arthritis Res Ther. 2007;9 Suppl 2:S3.
  • 40
    van Hal TW, van Bon L, Radstake TR. A system out of breath: how hypoxia possibly contributes to the pathogenesis of systemic sclerosis. Int J Rheumatol. 2011;2011:824972.
  • 41
    Geyer M, Muller-Ladner U. The pathogenesis of systemic sclerosis revisited. Clin Rev Allergy Immunol. 2011 Apr;40(2):92-103.
  • 42
    Whitfield ML, Finlay DR, Murray JI, Troyanskaya OG, Chi JT, Pergamenschikov A, et al. Systemic and cell type-specific gene expression patterns in scleroderma skin. Proc Natl Acad Sci U S A. 2003 Oct 14;100(21):12319-24.
  • 43
    Sato S, Fujimoto M, Hasegawa M, Takehara K. Altered blood B lymphocyte homeostasis in systemic sclerosis: expanded naive B cells and diminished but activated memory B cells. Arthritis Rheum. 2004 Jun;50(6):1918-27.
  • 44
    Maurer B, Distler O. Emerging targeted therapies in scleroderma lung and skin fibrosis. Best Pract Res Clin Rheumatol. 2011 Dec;25(6):843-58.
  • 45
    Mitri GM, Lucas M, Fertig N, Steen VD, Medsger TA, Jr. A comparison between anti-Th/To- and anticentromere antibody-positive systemic sclerosis patients with limited cutaneous involvement. Arthritis Rheum. 2003 Jan;48(1):203-9.
  • 46
    Oddis CV, Okano Y, Rudert WA, Trucco M, Duquesnoy RJ, Medsger TA, Jr. Serum autoantibody to the nucleolar antigen PM-Scl. Clinical and immunogenetic associations. Arthritis Rheum. 1992 Oct;35(10):1211-7.
  • 47
    Steen VD, Powell DL, Medsger TA, Jr. Clinical correlations and prognosis based on serum autoantibodies in patients with systemic sclerosis. Arthritis Rheum. 1988 Feb;31(2):196-203.
  • 48
    Santiago M, Baron M, Hudson M, Burlingame RW, Fritzler MJ. Antibodies to RNA polymerase III in systemic sclerosis detected by ELISA. J Rheumatol. 2007 Jul;34(7):1528-34.
  • 49
    Okano Y, Steen VD, Medsger TA, Jr. Autoantibody to U3 nucleolar ribonucleoprotein (fibrillarin) in patients with systemic sclerosis. Arthritis Rheum. 1992 Jan;35(1):95-100.
  • 50
    Sakkas LI, Xu B, Artlett CM, Lu S, Jimenez SA, Platsoucas CD. Oligoclonal T cell expansion in the skin of patients with systemic sclerosis. J Immunol. 2002 Apr 1;168(7):3649-59.
  • 51
    Steen VD, Engel EE, Charley MR, Medsger TA, Jr. Soluble serum interleukin 2 receptors in patients with systemic sclerosis. J Rheumatol. 1996 Apr;23(4):646-9.
  • 52
    Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol. 2004 Aug;4(8):583-94.
  • 53
    Cox TR, Erler JT. Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. Dis Model Mech. 2011 Mar;4(2):165-78.
  • 54
    Huang C, Ogawa R. Fibroproliferative disorders and their mechanobiology. Connect Tissue Res. 2012;53(3):187-96.
  • 55
    Abraham DJ, Eckes B, Rajkumar V, Krieg T. New developments in fibroblast and myofibroblast biology: implications for fibrosis and scleroderma. Curr Rheumatol Rep. 2007 May;9(2):136-43.
  • 56
    Desmouliere A, Chaponnier C, Gabbiani G. Tissue repair, contraction, and the myofibroblast. Wound Repair Regen. 2005 Jan-Feb;13(1):7-12.
  • 57
    Cipriani P, Franca Milia A, Liakouli V, Pacini A, Manetti M, Marrelli A, et al. Differential expression of stromal cellderived factor 1 and its receptor CXCR4 in the skin and endothelial cells of systemic sclerosis patients: Pathogenetic implications. Arthritis Rheum. 2006 Sep;54(9):3022-33.
  • 58
    Phillips RJ, Burdick MD, Hong K, Lutz MA, Murray LA, Xue YY, et al. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J Clin Invest. 2004 Aug;114(3):438-46.
  • 59
    Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008 Jan;214(2):199-210.
  • 60
    Hasegawa M, Fujimoto M, Kikuchi K, Takehara K. Elevated serum levels of interleukin 4 (IL-4), IL-10, and IL-13 in patients with systemic sclerosis. J Rheumatol. 1997 Feb;24(2):328-32.
  • 61
    Tsuji-Yamada J, Nakazawa M, Minami M, Sasaki T. Increased frequency of interleukin 4 producing CD4+ and CD8+ cells in peripheral blood from patients with systemic sclerosis. J Rheumatol. 2001 Jun;28(6):1252-8.
  • 62
    Khan K, Xu S, Nihtyanova S, Derrett-Smith E, Abraham D, Denton CP, et al. Clinical and pathological significance of interleukin 6 overexpression in systemic sclerosis. Ann Rheum Dis. 2012 Jul;71(7):1235-42.
  • 63
    Chizzolini C, Brembilla NC, Montanari E, Truchetet M-E. Fibrosis and immune dysregulation in systemic sclerosis. Autoimmunity Reviews. [doi: DOI: 10.1016/j. autrev.2010.09.016]. 2011;10(5):276-81.
  • 64
    Varga J. Scleroderma and Smads: dysfunctional Smad family dynamics culminating in fibrosis. Arthritis Rheum. 2002 Jul;46(7):1703-13.
  • 65
    Bhattacharyya S, Ishida W, Wu M, Wilkes M, Mori Y, Hinchcliff M, et al. A non-Smad mechanism of fibroblast activation by transforming growth factor-beta via c-Abl and Egr-1: selective modulation by imatinib mesylate. Oncogene. 2009 Mar 12;28(10):1285-97.
  • 66
    Gordon JK, Spiera RF. Targeting tyrosine kinases: a novel therapeutic strategy for systemic sclerosis. Curr Opin Rheumatol. 2010 Nov;22(6):690-5.
  • 67
    Wu M, Melichian DS, de la Garza M, Gruner K, Bhattacharyya S, Barr L, et al. Essential roles for early growth response transcription factor Egr-1 in tissue fibrosis and wound healing. Am J Pathol. 2009 Sep;175(3):1041-55.
  • 68
    Wei J, Fang F, Lam AP, Sargent JL, Hamburg E, Hinchcliff ME, et al. Wnt/beta-catenin signaling is hyperactivated in systemic sclerosis and induces Smad-dependent fibrotic responses in mesenchymal cells. Arthritis Rheum. 2012 Feb 10.
  • 69
    Ludwicka A, Ohba T, Trojanowska M, Yamakage A, Strange C, Smith EA, et al. Elevated levels of platelet derived growth factor and transforming growth factor-beta 1 in bronchoalveolar lavage fl uid from patients with scleroderma. J Rheumatol. 1995 Oct;22(10):1876-83.
  • 70
    Wei J, Ghosh AK, Sargent JL, Komura K, Wu M, Huang QQ, et al. PPARgamma downregulation by TGFss in fi broblast and impaired expression and function in systemic sclerosis: a novel mechanism for progressive fi brogenesis. PLoS One. 2010;5(11):e13778.
  • Update on the etiopathogenesis of systemic sclerosis

    Adriana Fontes ZimmermannI,*; Marcia Margaret Menezes PizzichiniII
  • Publication Dates

    • Publication in this collection
      27 Jan 2014
    • Date of issue
      Dec 2013

    History

    • Received
      24 June 2012
    • Accepted
      28 Feb 2013
    Sociedade Brasileira de Reumatologia Av Brigadeiro Luiz Antonio, 2466 - Cj 93., 01402-000 São Paulo - SP, Tel./Fax: 55 11 3289 7165 - São Paulo - SP - Brazil
    E-mail: sbre@terra.com.br