Research Article
Epithelial cells utilize cortical actin/myosin to activate latent TGF-β through integrin αvβ6-dependent physical force

https://doi.org/10.1016/j.yexcr.2012.01.020Get rights and content

Abstract

Transforming Growth Factor Beta (TGF-β) is involved in regulating many biological processes and disease states. Cells secrete cytokine as a latent complex that must be activated for it to exert its biological functions. We previously discovered that the epithelial-restricted integrin αvβ6 activates TGF-β and that this process is important in a number of in vivo models of disease. Here, we show that agonists of G-protein coupled receptors (Sphingosine-1-Phosphate and Lysophosphatidic Acid) which are ligated under conditions of epithelial injury directly stimulate primary airway epithelial cells to activate latent TGF-β through a pathway that involves Rho Kinase, non-muscle myosin, the αvβ6 integrin, and the generation of mechanical tension. Interestingly, lung epithelial cells appear to exert force on latent TGF-β using sub-cortical actin/myosin rather than the stress fibers utilized by fibroblasts and other traditionally “contractile” cells. These findings extend recent evidence suggesting TGF-β can be activated by integrin-mediated mechanical force and suggest that this mechanism is important for an integrin (αvβ6) and a cell type (epithelial cells) that have important roles in biologically relevant TGF-β activation in vivo.

Highlights

►S1P induces αvβ6-mediated TGF-β activation through Rho Kinase. ►Cell contraction regulates S1P and LPA induced αvβ6-mediated TGF-β activation. ►S1P and LPA induced αvβ6-mediated TGF-β activation requires cellular tension. ►Increased tension in epithelial cells results from cortical actin.

Introduction

Transforming Growth Factor Beta (TGF-β) is a central mediator in multiple in vivo disease models, including fibrosis in the lungs, kidney, and biliary tract [1], [2], [3], [4], [5], acute lung injury [6], [7], and pulmonary emphysema [8]. Cells secrete the pleiotropic cytokine as a large latent complex, which must be activated to exert its biological functions. There are many ways to activate latent TGF-β in vitro, for example, via heat, acidic pH, and matrix-metalloproteinases. However, the relevance of these processes in vivo is poorly understood [9]. Several studies have demonstrated that a subset of integrins that interact with an arginine–glycine–aspartic acid (RGD)-binding site motif also directly bind and activate latent TGF-β [1], [10].

Integrins are a widely expressed family of cell surface receptors that mediate cell adhesion and bidirectional signaling to regulate cellular processes. The importance of integrins in TGF-β activation in vivo is highlighted by transgenic mice that harbor a knock-in mutation of Tgfb1, so that the RGD binding site is mutated to RGE. These mice exhibit multi-organ inflammation and autoimmunity, a phenotype that completely phenocopies that of Tgfb1−/− mice, presumably due to a lack of binding and activation of TGF-β by RGD-binding integrins [11]. However, the only RGD integrins that have been definitively shown to regulate TGF-β activation in vivo are αvβ6 and αvβ8 [1], [12]. In particular, Itgb8−/− mice treated with blocking antibodies against αvβ6 from birth demonstrate all of the developmental phenotypes of mice lacking both TGF-β isoforms 1 and 3 [13].

We previously demonstrated that the epithelial-restricted integrin αvβ6 activates TGF-β and that Itgb6−/− mice exhibit a phenotype mildly similar to Tgfb1−/− mice [1], [14]. These mice are also dramatically protected in models of diseases that are mediated by TGF-β, supporting a role for αvβ6 in regulating TGF-β bioactivity in vivo [1], [4], [5], [9], [14]. Because of the importance of αvβ6-mediated TGF-β activation in the pathogenesis of various disease states, we wanted to determine the molecular signals and the mechanisms that regulate activation of this pathway.

Binding of αvβ6 to latent TGF-β1 and 3 in vitro is insufficient for activation of the cytokine, and cytoplasmic interactions between the integrin and the actin cytoskeleton are required for this process [1]. However, the mechanism and significance of these interactions in modulating αvβ6-mediated TGF-β activation are unknown. Furthermore, tethering of the latent complex to the extracellular matrix (ECM) by latent TGF-β binding protein-1 (LTBP-1) is required [15]. These findings suggest that TGF-β activation by αvβ6 potentially involves a mechanical mechanism. In vitro evidence supporting a role for mechanical force in integrin-dependent TGF-β activation has been provided by studies of αvβ3 and αvβ5-mediated TGF-β activation in fibroblasts [16], but thus far, there are no convincing data demonstrating roles for either of these integrins in activating TGF-β in vivo. The recently solved crystal structure of the small latent complex of TGF-β1 provides a model for how an RGD-binding integrin could exert mechanical force on a tethered latent complex and activate it. Electron microscopic analysis of purified integrin αvβ6 and the small latent complex provided further evidence that this integrin binds to the latent complex in an appropriate fashion to exert activating mechanical force [17]. However, the relevance of mechanical force in TGF-β activation in vivo remains unknown.

The purpose of this study was to identify signals that regulate αvβ6-mediated TGF-β activation and to determine what role, if any, mechanical force plays in the activation process. Here, we describe a novel activator, Sphingosine 1-Phosphate (S1P) that regulates αvβ6-mediated TGF-β activation in primary lung epithelial cells. We also demonstrate that both S1P and Lysophosphatidic Acid (LPA) induced αvβ6-mediated TGF-β activation require cell contraction and the generation of cellular tension. Finally, we show that cell tension generated by these primary epithelial cells is associated with organization of sub-cortical actin rings without evidence of actin stress fibers. Our findings highlight the potential for the development of additional therapeutic strategies for diseases that involve aberrant αvβ6-mediated TGF-β activation signaling and demonstrate a role for mechanical force in mediating TGF-β activation in a cell type and by an integrin that have been clearly shown to be relevant in vivo.

Section snippets

Cell lines, antibodies, and reagents

NHBE cells were cultured at 37 °C, 5% CO2 (Lonza, Walkersville, MD, USA). The β6-blocking antibody, 3G9, was provided by Paul Weinreb and Shelia Violette (Biogen Idec and Stromedix, Cambridge, MA, USA), and the pan-TGF-β blocking antibody, 1D11, was purchased (R&D Systems, Minneapolis, MN, USA). Agonists and inhibitors used were S1P, LPA, Y-27632, and Blebbistatin (Sigma-Aldrich, St. Louis, MO, USA), and active human recombinant TGF-β1 (R&D Systems, Minneapolis, MN, USA).

Western Blot

NHBE cells were cultured

S1P induces αvβ6-mediated TGF-β activation by acting through ROCK

S1P is a G-protein coupled receptor (GPCR) agonist that is abundantly stored in platelets and released at sites of increased TGF-β signaling, such as inflamed or injured tissues. The phospholipid induces contraction in airway smooth muscle cells and epithelial cells and has been implicated to play a role in tissue fibrosis [20], [21]. To determine whether S1P could induce αvβ6-mediated TGF-β activation in epithelial cells, primary normal human bronchial epithelial (NHBE) cells were treated with

Conflict of interest

The authors declare that no conflict of interest exists.

Acknowledgments

This work was supported by grant R37HL53949 from the NHLBI to D.S., U19AI077439 from the NIAID to D.S., and by a Pre-doctoral Fellowship in Pharmacology/Toxicology from the PhRMA Foundation to M.M.G.

References (24)

  • R.G. Jenkins et al.

    Ligation of protease-activated receptor 1 enhances alpha(v)beta6 integrin-dependent TGF-beta activation and promotes acute lung injury

    J. Clin. Invest.

    (2006)
  • D.G. Morris et al.

    Loss of integrin alpha(v)beta6-mediated TGF-beta activation causes Mmp12-dependent emphysema

    Nature

    (2003)
  • Cited by (0)

    View full text