Elsevier

Autoimmunity Reviews

Volume 10, Issue 5, March 2011, Pages 267-275
Autoimmunity Reviews

Review
Fibrosis in systemic sclerosis: Emerging concepts and implications for targeted therapy

https://doi.org/10.1016/j.autrev.2010.09.015Get rights and content

Abstract

Systemic sclerosis (SSc) is a complex and incompletely understood disease associated with fibrosis in multiple organs. Recent findings identify transforming growth factor-ß (TGF-ß), Wnt ligands, toll-like receptor-mediated signaling, hypoxia, type I interferon, type 2 immune responses and mechanical stress as extracellular cues that modulate fibroblast function and differentiation, and as potential targets for therapy. Moreover, fibrillin-1 has a major role in storing and regulating the bioavailability of TGF-ß and other cytokines, and fibrillin-1 mutations are implicated in a congenital form of scleroderma called stiff skin syndrome. Fibrosis is due not only to the activation of tissue-resident fibroblasts and their transdifferentiation into myofibroblasts, but also the differentiation of bone marrow-derived fibrocytes, and transition of endothelial and epithelial cells, pericytes and adipocytes into activated mesenchymal cells. These responses are modulated by signaling mediators and microRNAs that amplify or inhibit TGF-ß and Wnt signaling. Gain-of-function and loss-of-function abnormalities of these mediators may account for the characteristic activated phenotype of SSc fibroblasts. The nuclear orphan receptor PPAR-γ plays a particularly important role in limiting the duration and intensity of fibroblast activation and differentiation, and impaired PPAR-γ expression or function in SSc may underlie the uncontrolled progression of fibrosis.

Identifying the perturbations in signaling pathways, mediators and differentiation programs that are responsible for SSc tissue damage allows their selective targeting. This in turn opens the door for therapies utilizing novel compounds, or drug repurposing by innovative uses of already-approved drugs. In view of the heterogeneous clinical presentation and unpredictable course of SSc, as well as its complex pathogenesis, only robust clinical trials incorporating the judicious application of biomarkers will be able to clarify the clinical utility of these innovative approaches.

Introduction

Systemic sclerosis (SSc) is a progressive autoimmune disease of unknown cause associated with substantial mortality. To date there are no disease-modifying therapies, and immunomodulatory agents that are highly effective in other rheumatic diseases have shown disappointing results in SSc. The explanation may lie in the complex nature of SSc, characterized by a constellation of vascular injury, inflammation and fibrosis. It is not clear which of these perturbations is primary. Every SSc patient manifests some degree of vascular injury, immune activation and tissue fibrosis, but the relative contributions of these distinct processes to individual disease phenotype and natural history varies greatly from patient to patient. This variability accounts for the strikingly heterogeneous clinical picture of SSc. The three seemingly disparate yet interrelated pathophysiologic processes account for the clinical manifestations of SSc. Evidence of vascular injury and endothelial damage are detected at initial evaluation in a majority of patients. Over time, progressive vascular damage and obliteration of small and medium sized arteries cause tissue ischemia and its myriad complications. Vascular injury also plays a role in activation of the innate and adaptive immune systems, and contribute directly and indirectly to tissue fibrosis. The interrelationship of the three cardinal processes in the pathogenesis of SSc is illustrated in Fig. 1. In this review we focus on recent exciting developments in understanding fibrosis, the final common pathway in SSc that accounts for much of its mortality.

Section snippets

Skin fibrosis

Skin fibrosis is prominent and widespread in diffuse cutaneous SSc (dcSSc), whereas in limited cutaneous SSc (lcSSc) vascular complications rather than fibrosis tend to predominate. Fibrotic skin is characterized by thick dermis and obliteration of appendages such as hair follicles, sweat glands and cutaneous blood vessels. Initially fibrosis is most prominent in the reticular dermis, but with progression, the subjacent adipose layer also becomes affected. Biochemical analysis of affected skin

Fibroblast activation in SSc

Over three decades ago, Carwile LeRoy demonstrated that lesional skin fibroblasts explanted from patients with SSc synthesize increased amounts of collagen in vitro compared to healthy control fibroblasts [1], [2]. Additional features that phenotypically define SSc fibroblasts include increased synthesis of extracellular matrix; constitutive production of cytokines and chemokines; altered expression of cell surface integrins and receptors for transforming growth factor-ß (TGF-ß),

Cell types and cell fate switching in fibrosis

It is not surprising that a multiplicity of cell types are activated or deregulated in SSc, and play a role in tissue damage. Effector cells prominent at various stages of the disease include endothelial cells and vascular pericytes, cellular components of both the adaptive and innate arms of the immune system (Th2 cells and B lymphocytes, dendritic cells, NK cells, monocytes, alternatively-activated macrophages, mast cells and eosinophils) and possibly plasmacytoid dendritic cells. In the

Persistent fibrotic responses: innate immune recognition signaling via toll-like receptors

Recent studies indicate a potential important role for innate immune signaling via toll-like receptors (TLRs) in fibrosis in SSc. A critical aspect of normal host defence against invading microorganisms is the ability to recognize pathogen-associated molecular patterns. Pattern recognition receptors (PRRs) are the critical cellular sensors for exogenous danger. The family of PRRs includes, in addition to TLRs, lectin receptors, scavenger receptors CD36 and MARCO and complement receptors [7].

Soluble factors and extracellular cues regulating fibroblast activation and differentiation

Transforming growth factor-ß is the pre-eminent cue for initiating connective tissue remodeling during both normal wound healing and pathological fibrosis. The expression of receptors for TGF-ß as well as surface integrins that activate latent TGF-ß are elevated on SSc fibroblasts, suggesting that the SSc phenotype reflects autocrine TGF-ß stimulation of sensitized cells [10]. While recent studies provide support for the “autocrine TGF-ß hypothesis” [11], [12], [13], [14], pharmacological

Peroxisome proliferator-activated receptor-γ: negative regulation of fibroblast activation and differentiation

Peroxisome proliferator-activated receptor gamma (PPAR-γ) directly modulates TGF-ß signaling and mesenchymal cell plasticity, and is increasingly associated with regulation of matrix remodeling and fibrosis. Originally identified in adipocytes, PPAR-γ is a nuclear hormone receptor and ligand-inducible transcription factor with fundamental roles in adipogenesis and lipid metabolism. In the absence of a recognized true physiologic ligand, PPAR-γ is considered to be an orphan receptor. Multiple

Impaired PPAR-γ in fibrosis and SSc: role in pathogenesis

Fibroblast-specific gene targeting of PPAR-γ resulted in markedly exaggerated skin fibrosis when mice were injected with bleomycin [62]. Similarly, PPAR-γ deletion in follicular stem cells in mice causes localized fibrosis that resembles scarring alopecia [63]. On the other hand, we found that treatment with rosiglitazone attenuated bleomycin-induced dermal fibrosis in mice, suggesting a critical physiologic role for PPAR-γ in preventing excessive fibrotic responses. Consistent with this notion

Conclusion

The intractable problem of fibrosis is finally beginning to yield its secrets. Unbiased genome-wide expression analysis using DNA microarrays, population-based genetic association studies and transgenic animal models are contributing to a substantially enhanced understanding of the cellular and molecular basis of fibrosis in SSc. Cell-intrinsic alterations in SSc fibroblasts contribute to their activated phenotype. These include aberrant expression of TGF-ß receptors, activation of downstream

Take-home messages

  • SSc is highly heterogeneous in its clinical and autoimmune manifestations; individualized therapy required

  • SSc is distinct from other rheumatic diseases; the unique combination of fibrosis and vascular disease is prominent

  • because its manifestations and clinical course of SSc are highly heterogeneous, there is an urgent need for the discovery and validation of biomarkers to help classify patients, identify high risk groups, predict drug responders

  • There are no effective therapies for reversing or

Acknowledgements

Supported by grants from the National Institutes of Health (AR 42309) and the Scleroderma Research Foundation. We are grateful to our colleagues Gabriella Lakos, Carol Feghali-Bostwick, Monique Hinchcliff, Robert Lafyatis, Michael Whitfield, Guofei Zhou, Cara Gottardi, Anna Lam, Carol Artlett and Maria Trojanowska for helpful comments, and members of the Varga lab for valuable insights.

References (68)

  • A. Vlad et al.

    The first five years of the Wnt targetome

    Cell Signal

    (May 2008)
  • J. Bayle et al.

    Increased expression of Wnt2 and SFRP4 in Tsk mouse skin: role of Wnt signaling in altered dermal fibrillin deposition and systemic sclerosis

    J Invest Dermatol

    (Apr 2008)
  • M. Chilosi et al.

    Aberrant Wnt/beta-catenin pathway activation in idiopathic pulmonary fibrosis

    Am J Pathol

    (May 2003)
  • B. Eckes et al.

    Fibroblast-matrix interactions in wound healing and fibrosis

    Matrix Biol

    (2000)
  • Y. Asano et al.

    Increased expression levels of integrin alphavbeta5 on scleroderma fibroblasts

    Am J Pathol

    (2004)
  • S.K. Huang et al.

    Eicosanoid lipid mediators in fibrotic lung diseases: ready for prime time?

    Chest

    (Jun 2008)
  • M.Y. Xu et al.

    Lysophosphatidic acid induces alphavbeta6 integrin-mediated TGF-beta activation via the LPA2 receptor and the small G protein G alpha(q)

    Am J Pathol

    (Apr 2009)
  • S.J. Chen et al.

    The early-immediate gene EGR-1 is induced by transforming growth factor-beta and mediates stimulation of collagen gene expression

    J Biol Chem

    (2006)
  • P. Karnik et al.

    Hair follicle stem cell-specific PPARgamma deletion causes scarring alopecia

    J Invest Dermatol

    (May 2009)
  • M. Kapoor et al.

    Sequential induction of pro- and anti-inflammatory prostaglandins and peroxisome proliferators-activated receptor-gamma during normal wound healing: a time course study

    Prostaglandins Leukot Essent Fatty Acids

    (2007)
  • T. Miyahara et al.

    Peroxisome proliferator-activated receptors and hepatic stellate cell activation

    J Biol Chem

    (Nov 17 2000)
  • E.C. Leroy

    Connective tissue synthesis by scleroderma skin fibroblasts in cell culture

    J Exp Med

    (1972)
  • E.C. LeRoy

    Increased collagen synthesis by scleroderma skin fibroblasts in vitro: a possible defect in the regulation or activation of the scleroderma fibroblast

    J Clin Invest

    (1974)
  • J. Varga et al.

    Systemic sclerosis: a prototypic multisystem fibrotic disorder

    J Clin Invest

    (2007)
  • C. Chizzolini

    Update on pathophysiology of scleroderma with special reference to immunoinflammatory events

    Ann Med

    (2007)
  • J. Pannu et al.

    Recent advances in fibroblast signaling and biology in scleroderma

    Curr Opin Rheumatol

    (2004)
  • E. Seki et al.

    TLR4 enhances TGF-beta signaling and hepatic fibrosis

    Nat Med

    (Nov 2007)
  • H. Ihn

    Scleroderma, fibroblasts, signaling, and excessive extracellular matrix

    Curr Rheumatol Rep

    (2005)
  • H. Ihn et al.

    Blockade of endogenous transforming growth factor beta signaling prevents up-regulated collagen synthesis in scleroderma fibroblasts: association with increased expression of transforming growth factor beta receptors

    Arthritis Rheum

    (2001)
  • Castelino F, Varga J. Interstitial lung disease in the connective tissue diseases: evolving concepts of pathogenesis...
  • J. Pannu et al.

    Increased levels of transforming growth factor beta receptor type I and up-regulation of matrix gene program: a model of scleroderma

    Arthritis Rheum

    (2006)
  • Y. Chen et al.

    Contribution of activin receptor-like kinase 5 (transforming growth factor beta receptor type I) signaling to the fibrotic phenotype of scleroderma fibroblasts

    Arthritis Rheum

    (2006)
  • H. Gardner et al.

    Gene profiling of scleroderma skin reveals robust signatures of disease that are imperfectly reflected in the transcript profiles of explanted fibroblasts

    Arthritis Rheum

    (2006)
  • M.L. Whitfield et al.

    Systemic and cell type-specific gene expression patterns in scleroderma skin

    Proc Natl Acad Sci USA

    (2003)
  • Cited by (149)

    • MicroRNAs in Skin Fibrosis

      2023, MicroRNA in Regenerative Medicine, Second Edition
    • Mesenchymal stromal cells for systemic sclerosis treatment

      2021, Autoimmunity Reviews
      Citation Excerpt :

      These profibrotic cytokines, as well as TGF-β, stimulate differentiation and activation of myofibroblasts, with increased ECM production leading to fibrosis. Disruptions in the ECM vary with disease progression; changes in different collagens, proteoglycans, and fibrillins [94] occur typically earlier in the disease, with type I collagen accumulating in later disease stages. In vitro expanded MSC are defined a minima since 2006 by the International Society for Cellular Therapy (ISCT) MSc committee, as : 1) a plastic-adherent polyclonal population with fibroblast-like morphology, 2) expressing CD73, CD90 and CD105 (in > 95% MSC), 3) in the absence of hematopoietic and endothelial markers, and 4) able to differentiate in vitro into osteoblasts, adipocytes, and chondroblasts [21].

    View all citing articles on Scopus
    View full text