Elsevier

Bone

Volume 44, Issue 5, May 2009, Pages 813-821
Bone

Thrombin-stimulated growth factor and cytokine expression in osteoblasts is mediated by protease-activated receptor-1 and prostanoids

https://doi.org/10.1016/j.bone.2008.12.031Get rights and content

Abstract

Thrombin exerts multiple effects upon osteoblasts including stimulating proliferation, and inhibiting osteoblast differentiation and apoptosis. Some of these effects are believed to be mediated by the synthesis and secretion of autocrine factors such as growth factors and cytokines. Many but not all cellular responses to thrombin are mediated by members of the protease-activated receptor (PAR) family of G protein-coupled receptors. The current study was undertaken to investigate the nature of thrombin's induction of autocrine factors by analysing the expression of twelve candidate genes in thrombin-stimulated primary mouse osteoblasts. Analysis by quantitative reverse transcription polymerase chain reaction (qRT-PCR) demonstrated that thrombin induced transforming growth factor β, cyclooxygenase-2, tenascin C, fibroblast growth factor-1 and -2, connective tissue growth factor and interleukin-6 expression in wild type osteoblasts, but not PAR-1 null mouse osteoblasts. Induction of all the thrombin-responsive genes was blocked by the presence of the non-selective cyclooxygenase inhibitor indomethacin. Further studies were conducted on interleukin-6, which was the gene that showed the greatest increase in expression following stimulation of osteoblast-like cells with thrombin. A PAR-1-specific activating peptide, but neither a PAR-4-activating peptide nor catalytically inactive thrombin induced release of interleukin-6 by osteoblasts. Furthermore, in the presence of the selective cyclooxygenase-1 and -2 inhibitors SC-560 and NS-398 thrombin-induced interleukin-6 release was prevented. Levels of both prostaglandin E2 and interleukin-6 in medium conditioned by thrombin-stimulated osteoblast-like cells were found to be significantly increased compared to medium conditioned by non-stimulated cells, however release of prostaglandin E2 was found to precede release of interleukin-6. Treatment of isolated osteoblast-like cells with a number of synthetic prostanoids stimulated secretion of interleukin-6 with differing potencies. These studies suggest that activation of PAR-1 on osteoblasts by thrombin induces cyclooxygenase activity, which in turn results in the increased expression of multiple secreted factors. The induction of these secreted factors may act in an autocrine fashion to alter osteoblast function, allowing these cells to participate in the earliest stages of bone healing by both autocrine and paracrine mechanisms.

Introduction

As well as converting fibrinogen to fibrin during clot formation, the serine protease thrombin (EC 3.4.21.5) elicits a number of responses from many cell types and is believed to participate in many pathological processes, including platelet activation, inflammation, and tissue repair [1]. Many of the cellular effects of thrombin are achieved through the action of the thrombin-responsive members of the protease-activated receptor (PAR) family of seven transmembrane domain G-protein-coupled receptors, [reviewed by [2]]. Three of the four known members of the PAR family, PARs-1,-3 and -4, are activated by proteolytic cleavage of their N-terminal extracellular domains by thrombin [3], [4], [5], [6], whilst the fourth, PAR-2, is activated by a number of proteases such as trypsin, but not thrombin [7]. Protease activated receptors-1 and -4 can also be activated by synthetic peptides analogous to the new N-terminal “tethered ligands” created by thrombin cleavage [4], [5]. Synthetic activating peptides based upon the naturally occurring tethered ligand sequences are able to activate receptors with high specificity and potency; for example, the peptide TFLLRN (single letter amino acid sequence) specifically activates PAR-1, while the synthetic peptide AYPGKF specifically activates PAR-4. Furthermore, activation of PAR-4 by thrombin or activating peptides is antagonised by trans-cinnamoyl modified peptides [8].

Mice have been derived in which the PAR-1 gene has been targeted by homologous recombination [9]. Although 50% of PAR-1 null mice die in utero due to placental defects, development and haemostasis of the surviving animals are normal [9]. Haemostasis is unaffected in PAR-1 null mice as murine platelet activation is dependent upon the presence of functional PAR-3 and PAR-4 rather than functional PAR-1 [4].

Primary wildtype mouse osteoblast-like cells are known to express PARs-1, -2 and -4 but not PAR-3, whilst PAR-1 null mouse osteoblasts express only PAR-2 and PAR-4 [10]. In vitro, thrombin treatment of osteoblasts has been shown to stimulate proliferation, Cai2+ mobilisation, release of prostaglandin E2 (PGE2), interleukin-6 (IL-6) and plasminogen activator inhibitor, and to inhibit the differentiation and serum deprivation- or glucocorticoid-induced apoptosis of isolated osteoblasts and osteoblast-like cells [10], [11], [12], [13], [14], [15]. The use of peptide agonists of the PARs and cells isolated from PAR-1 null mice indicates that most, but not all of these effects are mediated by the activation of PAR-1 [10], [16], [17].

The current study was conducted to gain a better understanding of how thrombin controls osteoblast function by examining the regulation of expression of a number of secreted factors in response to thrombin treatment of undifferentiated primary mouse osteoblast-like cells. We hypothesised that PAR-1-dependent and PAR-1-independent signalling pathways activated by thrombin would result in an altered pattern of expression of genes encoding factors known to modulate osteoblast function in vivo and in vitro. The twelve candidate genes chosen for study were insulin-like growth factor (IGF)-1 and -2, transforming growth factor β1 (TGFβ), fibroblast growth factor (FGF)-1 and -2, fibronectin (FN), IL-6, connective tissue growth factor (CTGF), tenascin C (TnC), platelet-derived growth factor A (PDGF), cyclooxygenase-2 (COX-2) and vascular endothelial growth factor (VEGF). All of the factors chosen, with the exception of FGF-1 have previously been shown to be expressed by osteoblasts [18], [19], [20], [21], [22], [23], [24], [25], and most of them have been shown to be induced by thrombin in other cell types (FGF-2, PDGF, TGFβ, CTGF, TnC, COX-2, FGF-1, VEGF and FN) or in osteoblast-like cells (IL-6) [14], [26], [27], [28], [29], [30], [31], [32], [33]. Furthermore, FGF-1 and -2, CTGF, TGFβ, TnC and IL-6 function as mitogens and stage-specific modulators of differentiation for osteoblast-like cells [34], [35], [36], [37], [38], while FGF-2 and VEGF stimulate bone repair in animal models [39], [40]; cyclooxygenases (which metabolise arachidonic acid to form prostaglandins) are required for normal bone repair [41], [42].

Having noted that expression by osteoblast-like cells of eight of the factors was thrombin-inducible, we observed that induction of all genes was PAR-1- and cyclooxygenase-dependent. Further experiments were conducted to study the mechanism by which thrombin induces IL-6 and PGE2 expression. The results suggest that activation of PAR-1 on the surface of osteoblasts results in prostanoid release, which is responsible for activating intracellular signalling pathways that result in increased expression of a wide range of secreted factors with important functional effects on both isolated osteoblasts and bone healing generally.

Section snippets

Materials

Reagents were obtained from Sigma-Aldrich Pty Ltd (St. Louis, MO, USA) unless otherwise stated. Synthetic prostaglandin D2 (PGD2), prostaglandin B2 (PGB2), prostaglandin E2 (PGE2), prostaglandin F2 alpha (PGF), prostaglandin I2 (PGI2) and thromboxane B2 (TXB2) were obtained from Cayman Chemicals (Ann Arbour MI, USA). The selective COX-2 inhibitor NS-398 and the selective COX-1 inhibitor SC-560 were obtained from Cayman Chemicals (Ann Arbour, MI, USA) and dissolved in dimethyl sulphoxide. The

Expression of candidate genes in response to thrombin treatment of wild type and PAR-1 null osteoblasts

The expression of twelve genes following thrombin stimulation of undifferentiated primary mouse osteoblast-like cells isolated from wildtype and PAR-1 null animals was studied using qRT-PCR. Values for MNE in cultures of serum-deprived osteoblast-like cells treated with either 0 or 100 nM thrombin are presented in Fig. 1. A final concentration of 100 nM thrombin was used in these experiments, since this concentration of active thrombin has been measured in clotted blood and has previously been

Discussion

We hypothesised that induction of secreted factors such as growth factors and cytokines is one mechanism by which thrombin influences cell function and participates in physiological and pathological processes. As thrombin has previously been shown to stimulate osteoblast proliferation and to inhibit osteoblast differentiation and apoptosis, the current study was undertaken to study the expression of a number of secreted factors by osteoblast-like cells following thrombin treatment.

The current

Acknowledgments

The authors would like to thank Dr S. Coughlin for providing the PAR-1 null mice, Ms S. Toulson for the day-to-day care of the mouse colony, Mr P. Campbell for purification of the human α-thrombin and Mr Kitipong Uaesoontrachoon for technical assistance. Funding was provided by National Health and Medical Research Council (contract grant numbers 251575 and 284233).

References (61)

  • M. Duarte et al.

    Thrombin induces rapid PAR1-mediated non-classical FGF1 release

    Biochem. Biophys. Res. Commun.

    (2006)
  • A. Fakhry et al.

    Effects of FGF-2/-9 in calvarial bone cell cultures: differentiation stage-dependent mitogenic effect, inverse regulation of BMP-2 and noggin, and enhancement of osteogenic potential

    Bone

    (2005)
  • Q. Luo et al.

    Connective tissue growth factor (CTGF) is regulated by Wnt and bone morphogenetic proteins signaling in osteoblast differentiation of mesenchymal stem cells

    J. Biol. Chem.

    (2004)
  • C. Xie et al.

    COX-2 from the injury milieu is critical for the initiation of periosteal progenitor cell mediated bone healing

    Bone

    (2008)
  • T.R. Faruqi et al.

    Structure-function analysis of protease-activated receptor 4 tethered ligand peptides. Determinants of specificity and utility in assays of receptor function

    J. Biol. Chem.

    (2000)
  • M.D. Rand et al.

    Blood clotting in minimally altered whole blood

    Blood

    (1996)
  • J. Shinoda et al.

    Effect of ceramide on interleukin-6 synthesis in osteoblast-like cells

    Cell Signal

    (1999)
  • N. Futaki et al.

    NS-398, a new anti-inflammatory agent, selectively inhibits prostaglandin G/H synthase/cyclooxygenase (COX-2) activity in vitro

    Prostaglandins

    (1994)
  • G. Bluteau et al.

    The modulation of gene expression in osteoblasts by thrombin coated on biphasic calcium phosphate ceramic

    Biomaterials

    (2006)
  • C.C. Chua et al.

    TGF-beta1 inhibits multiple caspases induced by TNF-alpha in murine osteoblastic MC3T3-E1 cells

    Biochim. Biophys. Acta

    (2002)
  • S.J. Song et al.

    The role of protease-activated receptor-1 in bone healing

    Am. J. Pathol.

    (2005)
  • E. Nakata et al.

    Expression of connective tissue growth factor/hypertrophic chondrocyte-specific gene product 24 (CTGF/Hcs24) during fracture healing

    Bone

    (2002)
  • S.M. Strukova

    Thrombin as a regulator of inflammation and reparative processes in tissues

    Biochemistry (Mosc)

    (2001)
  • S.R. Macfarlane et al.

    Proteinase-activated receptors

    Pharmacol. Rev.

    (2001)
  • H. Ishihara et al.

    Protease-activated receptor 3 is a second thrombin receptor in humans

    Nature

    (1997)
  • M.L. Kahn et al.

    A dual thrombin receptor system for platelet activation

    Nature

    (1998)
  • W.F. Xu et al.

    Cloning and characterization of human protease-activated receptor 4

    Proc. Natl. Acad. Sci. U. S. A.

    (1998)
  • S. Nystedt et al.

    Molecular cloning of a potential proteinase activated receptor

    Proc. Natl. Acad. Sci. U. S. A.

    (1994)
  • M.D. Hollenberg et al.

    Proteinase-activated receptor-4: evaluation of tethered ligand-derived peptides as probes for receptor function and as inflammatory agonists in vivo

    Br. J. Pharmacol.

    (2004)
  • A.J. Connolly et al.

    Role of the thrombin receptor in development and evidence for a second receptor

    Nature

    (1996)
  • Cited by (37)

    • The effect of oral dabigatran etexilate on bone density, strength, and microstructure in healthy mice

      2018, Bone Reports
      Citation Excerpt :

      The effect of thrombin and the thrombin receptor in relation to bone have been investigated in both in vitro and in vivo experiments (Pagel et al., 2006; Sivagurunathan et al., 2013; Wienen et al., 2007). In vitro studies of cell cultures have shown that thrombin can increase the expression of RANKL relatively to OPG mediated by IL6 and PGE2, thereby enhancing osteoclast activation and bone degeneration (Kozawa et al., 1997; Pagel et al., 2009). Therefore, it is reasonable to assume that inhibition of thrombin will result in a higher bone mass.

    • Relationship between heterotopic ossification and traumatic brain injury: Why severe traumatic brain injury increases the risk of heterotopic ossification

      2018, Journal of Orthopaedic Translation
      Citation Excerpt :

      Cyclooxygenase is a key enzyme in the conversion of arachidonic acid to prostaglandins, playing an important role in the inflammatory response. Pagel et al. [64] proposed that fibroblast growth factor-1 and fibroblast growth factor-2, connective tissue growth factor, TGF-β1, tenascin C, VEGF and interleukin-6 (IL-6) induced by local prostaglandin are important factors in fracture healing. In addition, the activation of PAR-1 by thrombin after the injury could be a key adjustment for cyclooxygenase-2 expression.

    View all citing articles on Scopus
    View full text