Basic Mechanisms Linking Inflammation and Fibrosis



Fig. 2.1
Myofibroblasts are central to fibrogenesis and are submitted to regulation by pro- and anti-fibrotic cytokines. Schematic and simplified view of the network of cytokines influencing ECM deposition by myofibroblasts. In orange, pro-fibrotic cytokines mostly produced by cells of non-hematopoietic origin. In blue, pro-fibrotic cytokines mostly produced by cells of hematopoietic origin. In red, anti-fibrotic cytokines. In purple, cytokines known to enhance the pro-inflammatory activity of fibroblasts but with no direct pro-fibrotic activity. + denotes activation. − denotes inhibition. CTGF connective tissue growth factor, ECM extracellular matrix, IL interleukin, PAI-1 plasminogen activator inhibitor 1, PDGF platelet-derived growth factor, TGF-β transforming growth factor-β, TNF tumor necrosis factor




Transforming Growth Factor-Beta: TGF-β

TGF-β is believed to play a crucial role in promoting fibrotic responses. Three isoforms of TGF-β exist, and in the context of fibrosis most of our knowledge is centered on TGF-β1 [61, 62]. TGF-β affects the behavior of a number of cellular types either of hematopoietic, mesenchymal, or epithelial origin. TGF-β is a powerful immunosuppressant, regulates inflammation, variably promotes or suppresses tumor growth, and is essential in wound healing [63]. The biological activities of TGF-β are mostly regulated by modifications of the released protein rather than by modifications of its synthesis. Indeed, TGF-β exists in a latent complex bound to the ECM. Following tissue injury, TGF-β is released from ECM and influences the cells in the immediate vicinity. Within the ECM, mature TGF-β exists as a dimer, forming a small latent complex with the latency-associated peptide (LAP), itself bound to a larger complex directly bound to ECM proteins. To become biologically active, the latent TGF-β complex needs to be activated by proteolytic mechanisms or by tractional forces mediated by integrins – most notably, the αVβ6, αVβ5, and αVβ3 integrins [64]. Following receptor binding, TGF-β implicates canonical and noncanonical signaling pathways. The SMAD pathway implicates the phosphorylation of SMAD2 and SMAD3 proteins and their association with the co-SMAD4 for nuclear translocation. This signaling cascade is negatively regulated by SMAD7 [65]. In fibrosis development, the noncanonical TGF-β signaling pathways may play a major role and involve MAPK, PI3K, and NFkB pathways as well as NADPH oxidase 4 (NOX4)/reactive oxygen species (ROS) pathways [66]. Key to avoid excessive ECM deposition, TGF-β signaling needs to be terminated once tissue is repaired and the synthesis of extracellular matrix should return to normal levels. Finely tuned homeostatic mechanisms are thought to be at play, but the molecular mechanisms that underlie the failure to limit TGF-β activity are poorly understood. However, recent work has highlighted the role of the orphan nuclear receptor NR4A1 in downregulating TGF-β signaling and participating to a negative feedback loop that limits excessive TGF signaling and could be exploited to attenuate fibrotic disorders [67].


Platelet-Derived Growth Factor: PDGF

PDGF signals through two different PDGF receptors, alpha and beta, and comprises a family of homo- or heterodimeric growth factors including PDGF-AA, PDGF-AB, PDGF-BB, PDGFCC, and PDGF-DD [68]. After wounding, the expression of PDGF is increased; this growth factor participates in the recruitment of neutrophils, macrophages, fibroblasts, and smooth muscle cells into the wound [69]. PDGF also stimulates the formation of granulation tissue with direct effects on fibroblasts, resulting in collagen matrix contraction and fibroblast differentiation into myofibroblasts in vitro [70]. Tyrosine kinase inhibitors such as imatinib mesylate, dasatinib, and nilotinib exert potent anti-fibrotic effects in experimental models of fibrosis, most likely by inhibiting signaling induced by PDGF-receptor engagement [71].


Connective Tissue Growth Factor: CTGF or CCN2

CTGF is a member of the CCN proteins that are key signaling and regulatory molecules involved in cell proliferation, angiogenesis, tumorigenesis, wound healing, and fibrosis [72]. CTGF is a modular matricellular component produced by fibroblasts (among other cells) and induced in response to various stimuli including TGF-β, endothelin-1, and angiotensin-II and is thought to act mainly by favoring an environment which promotes fibrogenesis [73]. It acts mostly in an autocrine manner interacting with other matrix components and adhesion molecules [74]. It has been implicated in several fibrotic conditions including scleroderma, chronic heart failure, and proliferative diabetic retinopathy.


Plasminogen Activator Inhibitor 1 (PAI-1)

PAI-1 is a member of the serine protease inhibitor (serpin) gene family and the major physiologic inhibitor of the serine proteases urokinase plasminogen activator (uPA) and tissue plasminogen activator (tPA). Inhibition of uPA/tPA results in the inhibition of plasminogen-to-plasmin conversion as well as plasmin-dependent MMP activation. Thus, PAI-1 protects ECM proteins from proteolytic degradation and helps accelerate wound healing. However, sustained PAI-1 activation may contribute to excessive collagen accumulation [75]. Fibroblasts obtained from fibrotic tissues spontaneously produce higher amounts of PAI-1 compared to control fibroblasts, and inhibition of PAI-1 protects against fibrosis in the lung [76], heart, and kidney but not the skin [77] in animal models of fibrosis. Factors that may induce high production of PAI-1 are VEGF and TGF-β1. Thus, PAI-1 participation to fibrosis development exemplifies the role of an excessive brake directed against events aimed at ECM reabsorption.


Interleukin-4 (IL-4) and IL-13

Classical Th2 cell products, such as IL-4 and particularly IL-13, have been shown to have direct and indirect roles in fibrosis development. IL-4 and IL-13 enhance type I collagen production in dermal fibroblasts [78, 79]. IL-13 in vivo acts by inducing TGF-β production by macrophages and directly stimulating myofibroblast and fibroblast synthetic activities. IL-13 function is regulated by the relative expression of the two IL-13 receptor subunits, IL-13Rα1 (signaling component) and IL-13Rα2 (decoy component). When IL-13Rα2 levels are low, fibrotic responses are enhanced [80]. Consistent with the importance of Th2-like response in fibrotic pathologies, the report shows increased eotaxin/CXCL13 serum levels in idiopathic retroperitoneal fibrosis [81].


Interleukin-6 (IL-6)

IL-6 is rather a newcomer in the fibrosis field, whose importance has been highlighted by the availability of targeted biological therapies. IL-6 is a pleiotropic cytokine with direct pro-angiogenic and pro-fibrotic activities [82]. Its levels are increased in the serum and skin of scleroderma patients [83], and its inhibition attenuates bleomycin-induced experimental fibrosis in the murine lung and skin [84, 85]. Of importance, IL-6 blockade has shown beneficial effects in humans affected by idiopathic retroperitoneal fibrosis [86].


Tumor Necrosis Factor (TNF)

Historically, TNF has been associated with the development of fibrosis in experimental models dominated by inflammation. In a pioneering work, Piguet and coworkers demonstrated that TNF blockade strongly attenuated the fibrotic response in the mouse with bleomycin-induced lung fibrosis [87]. However, when TNF inhibitors have become available in the clinics, their use in humans was not always associated with fibrosis halting; conversely, in some cases worsening of the fibrotic process was observed, particularly in individuals suffering from systemic sclerosis [88]. On the one hand, TNF is particularly important among inflammatory cytokines because it enhances the recruitment of inflammatory cells which mandatorily precede fibrosis; on the other hand, TNF inhibits the transcription of type I and type III procollagen mRNA [89, 90] and is a strong inducer, in fibroblasts and macrophages, of MMP production, in particular of the interstitial collagenase MMP1, which participates in ECM degradation [91] [92]. Thus, the net effect on fibrosis development of TNF may very well depend on the context and timing in which it operates, variably resulting in enhanced or decreased ECM deposition.



2.6 Fibrosis in IgG4-Related Disorder and Unresolved Questions


The pathological features currently used to identify IgG4-RD include a lymphoplasmacytic infiltrate rich in IgG4+ plasma cells, storiform fibrosis, obliterative phlebitis, and mild-to-moderate tissue eosinophilia. Minor features include the presence of germinal centers, lymphoid follicles, non-obliterative phlebitis, and obliterative arteritis (usually in the lung) [9396]. From the fibro-inflammatory point of view, specific and peculiar pathological characteristics of IgG4-RD are the presence of storiform fibrosis and a very rich inflammatory infiltrate composed largely by CD4+ T cells, macrophages, and plasma cells. The presence of eosinophils is also remarkable. Storiform fibrosis is rare if ever encountered in connective tissue diseases or other diseases characterized by organ fibrosis but sometimes described in association with solid tumors [97]. Thus, one of the unanswered questions is why collagen and ECM deposition in florid IgG4-RD assumes this typical disposition. It is known that myofibroblasts and spindle-shaped fibroblasts are tightly associated with freshly deposed collagen fibers in IgG4-RD, therefore conforming to the paradigm that α-smooth muscle actin (SMA)+ cells are central to fibrosis development [98]. It is also remarkable that under certain circumstances fibrosis associated to IgG4-RD can regress [99], or at least, the size of lesions, as assessed by imaging studies, definitely decreases; this leads to the hypothesis that collagen bundles are not extensively cross-linked and could undergo degradation, perhaps under the influence of MMPs. This notwithstanding, in several cases thick, acellular matrix results in permanent tumorlike masses.

When searched for, classical mediators involved in fibrosis including LAP (TGF-β1) are mostly expressed in macrophages, TGF-RII and PDGF-B are mostly expressed in myofibroblasts and epithelial cells, and PDGF-Rα and PDGR-Rβ have been documented in type I autoimmune pancreatitis [98]. In these settings, the strongest positivity for such mediators was observed in grade 3 histological pattern [100], when the inflammatory component is maximal.

As mentioned in previous paragraphs, Th2 cells and their products, particularly IL-4 and IL-13, are associated with fibrotic responses in many distinct pathological situations. Thus, the documentation of expanded Th2 cell responses in IgG4-RD may provide a pathogenically interesting link between inflammation and fibrosis [101104]. However, Th2 cell responses have been documented only in a fraction of patients with IgG4-RD [105, 106]. Th2 cells produce also IL-5, which enhances eosinophil maturation, recruitment, and activation, and eosinophils are frequently present in fibroblastic tissues in IgG4-RD [107]. Eosinophils may release IL-13, TGF-β1, and PDGF, thus participating in the activation of fibroblasts [108]. The inflammatory infiltrate in IgG4-RD is also rich in macrophages, which may also release TGF-β1 and PDGF. Given the presence of B cells as well as of plasma cells particularly those producing the eponym IgG4, it is clearly tempting to speculate that these cells may produce factors that enhance the production of ECM by fibroblasts. B-cell contribution to fibrosis is an area of relatively scarce research. However, B cells were shown to induce contact-dependent human dermal fibroblast activation with upregulation of, among other mediators, type I collagen. This B-cell activity was enhanced by B-cell activation in the presence of anti-IgM and B-cell-activating factor (BAFF) [109]. Interestingly, BAFF levels are increased in IgG4-RD, positively correlating with IgG4 serum levels and decreasing after glucocorticoid therapy [110]. Thus, this remains an area of specific interest in IgG4-RD fibrosis.

Additional inflammatory cells identified in tissues undergoing IgG4-RD pathological transformation are Foxp3+ T regulatory cells [111]. These cells may produce TGF-β, and therefore they may be mechanistically associated with the development of fibrosis. However, no direct evidence has been gathered to back this hypothesis. Furthermore, in humans Foxp3 can be acquired in fully differentiated effector cells with no regulatory activity. This is the case in systemic sclerosis skin where Foxp3+ cells have been proved to produce high levels of IL-4, which has direct pro-fibrotic activities [112].

It should however be noted that no specific studies have been devoted in IgG4-RD to investigate the contribution of each cell type to the activation of fibroblasts.


Conclusions

An exuberant inflammatory infiltrate rich in CD4+ T cells, macrophages, and IgG4+ plasma cells in addition to storiform fibrosis participates to the pathological definition of IgG4-RD. It is reasonable to speculate that the inflammatory infiltrate drives fibroblast activation, myofibroblast recruitment, and subsequent ECM deposition in this disorder. The fundamental processes, mediators, and cells associated to fibrosis in general seem to be at play in IgG4-RD fibrosis. However, the mechanisms specifically involved in its generation remain elusive. This is particularly true taking into consideration the very heterogeneous clinical presentation of this condition and the difficulties we experience in understanding the relationship between the fundamental aspects of the disease and the variety of organs affected. Thus, we should try to answer the question whether indeed a single etiopathogenic event is at the base of all forms of IgG4-RD or whether IgG4-RD is a default reaction to a variety of different etiopathogenic events. The functional characterization of inflammatory cells present in early lesions, the hunting for an eventual antigen – autoantigen recognized by infiltrating T cells or B cells – and their interplay with cells of the innate immune system should bring new light and improve our knowledge of IgG4-RD in the next few years. This should allow to implement more targeted therapies or even more optimistically to adopt preventive strategies.


References



1.

Gabbiani G, Majno G (1972) Dupuytren’s contracture: fibroblast contraction? An ultrastructural study. Am J Pathol 66:131–146PubMedPubMedCentral


2.

Hinz B et al (2007) The myofibroblast: one function, multiple origins. Am J Pathol 170:1807–1816PubMedPubMedCentral


3.

Lamouille SJX, Derynck R (2014) Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 15:178–196PubMedPubMedCentral


4.

Jimenez SA (2013) Role of endothelial to mesenchymal transition in the pathogenesis of the vascular alterations in systemic sclerosis. ISRN Rheumatol 2013:835948PubMedPubMedCentral


5.

Lin SL et al (2008) Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am J Pathol 173:1617–1627PubMedPubMedCentral


6.

Humphreys BD et al (2010) Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol 176:85–97PubMedPubMedCentral


7.

Rock JR et al (2011) Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc Natl Acad Sci U S A 108:E1475–E1483PubMedPubMedCentral


8.

Dulauroy S et al (2012) Lineage tracing and genetic ablation of ADAM12(+) perivascular cells identify a major source of profibrotic cells during acute tissue injury. Nat Med 18:1262–1270PubMed


9.

Duffield JS et al (2013) Host responses in tissue repair and fibrosis. Annu Rev Pathol 8:241–276PubMed


10.

Hung C et al (2013) Role of lung pericytes and resident fibroblasts in the pathogenesis of pulmonary fibrosis. Am J Respir Crit Care Med 188:820–830PubMedPubMedCentral


11.

Xia H et al (2014) Identification of a cell-of-origin for fibroblasts comprising the fibrotic reticulum in idiopathic pulmonary fibrosis. Am J Pathol 184:1369–1383PubMedPubMedCentral


12.

Yoshizaki A et al (2008) CD19 regulates skin and lung fibrosis via Toll-like receptor signaling in a model of bleomycin-induced scleroderma. Am J Pathol 172:1650–1663PubMedPubMedCentral


13.

Hinz B (2009) Tissue stiffness, latent TGF-beta1 activation, and mechanical signal transduction: implications for the pathogenesis and treatment of fibrosis. Curr Rheumatol Rep 11:120–126PubMed


14.

Mih JD et al (2011) A multiwell platform for studying stiffness-dependent cell biology. PLoS One 6:e19929PubMedPubMedCentral

Sep 2, 2017 | Posted by in NEPHROLOGY | Comments Off on Basic Mechanisms Linking Inflammation and Fibrosis

Full access? Get Clinical Tree

Get Clinical Tree app for offline access