View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Elsevier - Publisher Connector

Biochimica et Biophysica Acta 1793 (2009) 888–892

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

journal homepage: www.elsevier.com/locate/bbamcr

Review The regulation of expression by tissue microenvironments

Richard P. Tucker a,⁎, Ruth Chiquet-Ehrismann b

a Department of Cell Biology and Human Anatomy, University of California at Davis, Davis, California 95616, USA b Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, Maulbeerstrasse 66, CH-4058 Basel, Switzerland

article info abstract

Article history: are a family of four proteins: tenascin-C, X, R and W. The four members of the Received 20 December 2007 family have strikingly diverse patterns of expression during development and in the adult organism Received in revised form 4 December 2008 indicating independent mechanisms of regulation. In this review we illustrate that there are two types of Accepted 19 December 2008 tenascins, those that are significantly regulated by the tissue microenvironment (tenascin-C and tenascin- Available online 31 December 2008 W), and those that have stabile, restricted expression patterns (tenascin-R and tenascin-X). We summarize what is known about the regulation of tenascin expression by transforming growth factor betas, fibroblast Keywords: fl Extracellular matrix growth factors, platelet derived growth factors, as well as pro- and anti-in ammatory cytokines or hormones Growth factor that either induce or inhibit expression of tenascins. Transforming growth factor beta © 2009 Elsevier B.V. All rights reserved. Development Tumor stroma Fibroblast growth factor

1. Introduction become more rounded or for growth cones to collapse (e.g., for tenascin-C see [11]; for tenascin-R see [12]; for tenascin-X see [13]). Tenascins are a family of four multimeric extracellular matrix This is often, but not always, accompanied by an increase in rates of (ECM) : tenascin-C, X, R and W [1]. The best described motility, invasive behavior and proliferation (for reviews see [4,14]). member of the family is tenascin-C, which is found in the embryo near Tenascin-W seems to have similar properties. For example, tenascin- migrating cells, at sites of epithelial–mesenchymal interactions and W added to the medium of primary cultures of osteoblasts on branching morphogenesis, and in developing connective tissue fibronectin leads to cell rounding [15] and the migration of a including bone, cartilage and tendons. Tenascin-C persists in some mammary tumor-derived cell line across a filter in a Boyden chamber connective tissue in the adult, and also appears in the stroma of many assay is stimulated by tenascin-W [8]. An attractive model to explain tumors and at sites of tissue repair and regeneration (for reviews see some of these observations, at least for tenascin-C, comes from studies [2–4]). Tenascin-X is primarily expressed in loose connective tissue of tenascin-C/fibronectin interactions. When cells are cultured on a such as dermis, epimysium and blood vessels both during develop- combination of tenascin-C and fibronectin, as opposed to fibronectin ment and in the adult; mutations in tenascin-X can lead to a type of alone, there is a reduction in the expression or activity of FAK, RhoA Ehlers–Danlos Syndrome (for review see [5]). In contrast, the patterns and tropomyosin I [16–18]. This appears to be mediated by tenascin-C of tenascin-R and tenascin-W expression in normal developing and inhibiting interactions between -4 and fibronectin [19,20], adult tissues are considerably more restricted. Tenascin-R is concen- which in turn inhibits the recruitment of syndecan-4 to the focal trated in the central and peripheral nervous system (e.g. [6]), and adhesion complex. This limits the size of the focal adhesion and alters tenascin-W is found primarily in bone (e.g. [7]). Like tenascin-C, signaling. Associated with this is an increase in the expression tenascin-W is also a prominent component of the stroma of certain of endothelin receptor A, which is correlated with an inhibition of tumors [8–10]. The primary sites of tenascin expression are summar- focal adhesion kinase and a loss of function of RhoA, all of which can ized in Table 1. contribute to tumorigenesis [21]. Diverse functional roles have been assigned to tenascins during Since it was first observed in tumor stroma over two decades ago development and disease. When used as a substratum in tissue [22] considerable research has been directed toward understanding culture, some tenascins inhibit the spreading of cells and the the role of tenascin-C in cancer (for review see [23]). More recently, it formation of large focal adhesions. Similarly, tenascins added to the was observed that tenascin-W is also found in the stroma of murine medium of cells cultured on adhesive ECM can cause the cells to [8] and human [9,10] breast cancers. Given the effects of tenascin-C and tenascin-W on cells in vitro cited above, it is likely that these proteins can contribute directly to the growth and metastasis of at ⁎ Corresponding author. Tel.: +1 530 752 0238. least some tumors. However, care must be taken not to generalize E-mail address: [email protected] (R.P. Tucker). roles for all tenascins in cancer. For example, tenascin-R is expressed

0167-4889/$ – see front matter © 2009 Elsevier B.V. All rights reserved. doi:10.1016/j.bbamcr.2008.12.012 R.P. Tucker, R. Chiquet-Ehrismann / Biochimica et Biophysica Acta 1793 (2009) 888–892 889

Table 1 Summary of tenascin expression and regulation

Examples of prominent expressiona Examples of regulationb [Ref.] Tenascin-C Organogenesis TGFβ1 [27–31,33,34] Dense connective tissue FGF1 [49] Smooth muscle FGF2 [28,46–50] Inflammation FGF4 [49] Cancer stroma PDGF [55] TNFα [59–61] Notch [77] Downregulation by miRNAs [78] Mechanical strain [76,83] Tenascin-W Bone BMP2 [7,8] Cancer stroma TNFα [8] Tenascin-X Loose connective tissue BDNF [70] Endothelial cells No effect seen with EGF, bFGF, TGFβ1, PDGF, and TNFα [69] Tenascin-R Central and peripheral nervous system NGFc [68]

a See text for references to reviews of expression patterns. b Upregulation unless otherwise noted. c May be related to the differentiation of a cell line. at lower levels in medulloblastomas than in control cerebellum tissue pattern of TGFβ1 in the developing heart [38] and in inflamed joints [24], and melanoma cells are more invasive in the ECM of tenascin-X [39] closely matches that of tenascin-C, tenascin-C is abundant around knockout mice than in controls [25]. TGFβ1-expressing megakaryocytes in bone marrow [40], and levels of Clearly tenascin expression is tightly regulated and is tenascin-C and levels of TGFβ1 expression are correlated in breast dependent on the tissue microenvironment, especially on the growth carcinomas [41] and ovarian tumors [42]. Care must be taken before factor milieux. This is apparent from the distinctive and often making generalizations based on the latter observations, as others complementary patterns of tenascin in the embryo have shown no correlation between the level of tenascin-C expression and from the presence or absence of tenascins in the stroma of certain and TGFβ1 expression in [43], though both are present. This tumors. In this review we will summarize the research on the effects may be explained, in part, by differences in the timing of the first of growth factors on the expression of tenascins in vitro and the appearance of tenascin-C and TGFβ1. signaling pathways used to stimulate tenascin expression. Studies with function-blocking antibodies also link TGFβ1 with the regulation of tenascin-C expression. For example, pre- and post- 2. Regulation of tenascin-C expression by transforming growth operative treatment of rats with anti-TGFβ1 inhibits the induction of factor betas tenascin-C expression in radiation-induced wounds [44]. Others have taken advantage of cells derived from knockout mice. Kalembeyi et al. Transforming growth factor betas (TGFβ) are a superfamily of [45] showed that a matrix metalloprotease, MMP-9, is induced by cytokines that play important roles in regulating cell differentiation TGFβ1 in a tenascin-C knockout-derived tumor cell line, but that this and proliferation, both during development and in pathologies (for induction is increased in the presence of exogenous tenascin-C. Thus, review see [26]). In one of the earliest studies of any tenascin, Pearson tenascin-C and TGFβ1 may act synergistically to increase the levels of et al. [27] showed that tenascin-C expression by chick embryo proteases in tumor stroma that can contribute to metastasis. fibroblasts is upregulated four-fold by the addition of 6–20 ng/ml of TGFβ1 to low-serum medium (see Table 1 for a summary of the 3. Tenascin-C expression and fibroblast growth factors regulation of tenascin expression). Similar induction of tenascin-C expression by low levels of TGFβ1 was reported in Swiss 3T3 cells [28] Fibroblast growth factor 2 (FGF2), also known as basic fibroblast and in various human carcinoma-derived cell lines [29] and growth factor, was first characterized as a potent promoter of transformed mouse mammary epithelial cells [30]. Interestingly, angiogenesis (for a review of the history of its discovery see [46]). It when melanoma cells are transfected with TGFβ1 and seeded into is one of the founding members of a large family of growth factors that collagen gels together with fibroblasts, tenascin-C expression can regulate cell motility, proliferation, differentiation and survival. increases in the fibroblasts [31], suggesting that some of the Like TGFβ1, the pattern of expression of FGF2 is also often correlated tenascin-C found in tumor stroma may be deposited by the stromal with the expression of tenascin-C, especially in the central nervous cells under the influence of the tumor-derived microenvironment. system (e.g. [28]). Accordingly, FGF2 can induce the expression of Tenascin-C and TGFβ1 are both found in developing connective tissue tenascin-C in [47,48] and neuroectoderm-derived tumors (e.g., see [32]), so it is not surprising that explants of mouse limb bud [49]. In the latter study FGF1 (aFGF) and FGF4 (K-FGF) also induced mesenchyme also express tenascin-C in response to TGFβs [33]. the expression of tenascin-C. Interestingly, introduction of FGF1 into Though the most robust inductive response was seen with TGFβ1, the lateral ventricles of the rat leads to the specific induction of the significant induction was also reported with TGFβ2 and TGFβ3. large splice variants of tenascin-C [50], and FGF2 induces the Interestingly, TGFβ1 tends to induce most strongly the expression of a expression of large splice variants of tenascin-C in Swiss 3T3 cells low molecular weight alternatively spliced variant of tenascin-C that [28]. Large splice variants of tenascin-C are often associated with lacks the so-called ‘variable domain’. This has been reported in Swiss motile cells and with malignancy and poor outcome (e.g. [51–54]). 3T3 cells [28] and endometrial adenocarcinoma cell lines [34]. The functional significance of this is unclear, but different splice variants 4. Platelet derived growth factors and tenascin-C expression have been shown to be differentially expressed during development (e.g. [35,36]) and in tumors (e.g. [37]) and to have different effects on Platelet derived growth factors (PDGF) are dimers of A, B, C and D cell adhesion in vitro (e.g. [36]). subunits. Best known are PDGF-AA, PDGF-AB and PDGF-BB, which Correlative evidence suggests that TGFβ1 is likely to be a regulator play important rules in influencing cell proliferation and motility in a of tenascin-C expression in vivo as well as in vitro: the expression variety of tissues. Tenascin-C and PDGFs are found together in some 890 R.P. Tucker, R. Chiquet-Ehrismann / Biochimica et Biophysica Acta 1793 (2009) 888–892 blood vessel walls and PDGF-BB has a potent inductive effect on arterial hypertension (see review by [73]), which is characterized in tenascin-C expression in smooth muscle cells derived from the aorta part by the extensive deposition of tenascin-C in the arterial wall (e.g. [55]. As described above for FGF2, PDGF-BB induces primarily large [74]). The receptor mutation leads to a reduction in BMP-induced splice variants of tenascin-C; this variant may play a role in the Smad signaling and an increase in MAPK signaling pathways, which invasion of smooth muscles cells into the vessel wall and the leads to the induction of tenascin-C expression by vascular smooth development of atherosclerotic plaque. Both PDGF receptor alpha muscle cells [75]. The tenascin-C can contribute to the pathobiology of and tenascin-C are expressed in the developing central nervous the disease by increasing vascular smooth muscle cell proliferation system by early oligodendroglia and their precursors (e.g. [56,57]). and survival. Regulation of tenascin-C expression can also depend on PDGF has a potent mitogenic effect on oligodendrocyte precursor the Rho/Rock pathway and actin dynamics ([76]; see accompanying cells; interestingly, this effect requires tenascin-C in the ECM [58], article by M. Chiquet on “Mechanotransduction in Fibroblasts”). implying that synergistic effects between growth factors and Recently, two novel mechanisms regulating tenascin-C expression tenascin-C may account in part for the mitogenic effects of exogenous have been described. On the one hand Notch signaling was shown to tenascin-C. induce tenascin-C expression representing a new oncogenic pathway active in [77]. On the other hand micro RNA (miRNA)- 5. Other growth factors and other tenascins 335 was shown to downregulate tenascin-C expression and the loss of this miRNA in metastatic breast tumors led to increased tenascin-C The induction of tenascin-C in tumor stroma and at other sites of expression. Restoration of miRNA-126 inhibited metastatic cell disease progression is likely to be related to its induction by invasion which depended on the down-regulation of tenascin-C [78]. cytokines associated with inflammation. For example, TNFα expres- sion is linked to tenascin-C expression in bronchial fibroblasts [59] 7. Tenascin promoters and transcription factors and cultured chondrocytes [60], and TNFα and interferon gamma induce tenascin-C expression in bronchial epithelial cells ([61]; see The distinctive differences in the expression and regulation of the also reviews [3,23]). Anti-inflammatories like dexamethasone inhibit tenascins are reflected in features found in their promoters. For the expression of tenascin-C in cultured human macrophages (e.g. example, the promoters of tenascin-X and tenascin-R lack TATA boxes [62]), in the granulation tissue of healing skin wounds [63], and in and in both cases transcription is initiated at multiple sites, whereas the stroma of experimentally induced murine tumors [64]. Gluco- tenascin-C is regulated by a typical TATA-box containing promoter. corticoids can reduce the expression of tenascin-C in bone marrow Nothing is known yet about the tenascin-W promoter, which is [65] and in the respiratory mucosa of patients suffering from pollen- waiting to be identified and characterized. In the case of tenascin-R induced asthma [66]. the information for cell-type specific expression in cells of neuronal Tenascin-W, the most recently discovered member of the tenascin origin is contained within 167 bp of the first exon and contains family [67], is also expressed in tumor stroma and its expression can binding sites for a variety of transcription factors such as p53/p73, NF1 also be upregulated by TNFα in vitro [8]. Nevertheless, tenascin-C and and glucocorticoid receptors [79]. However, it is not known which tenascin-W are clearly regulated independently, since there are factors are relevant for the expression of tenascin-R in the central tumors that express one of these two tenascins but not the other nervous system. In the case of tenascin-X it is thought that several [9,10]. During development, tenascin-W is most abundantly expressed regions harboring binding sites for Sp1 and Sp3 are involved in its in developing bone [7,15]. Accordingly, tenascin-W expression is regulation [80]. In line with the many signaling pathways able to increased in C2C12 cells [7] and HC11 cells [8] by bone morphogenetic induce tenascin-C expression (see above and Table 1) binding sites for protein-2 (BMP-2), but not by TGFβ1. the transcription factors activated by these pathways have been Relatively little is known about the regulation of tenascin-R and identified and are functional in the promoter of tenascin-C. These tenascin-X expression by the tissue microenvironment. Nerve growth include binding sites for paired-related homeobox1 Prx1 shown to be factor (NGF) can stimulate the expression of tenascin-R by PC12 cells activated by focal adhesion kinase signaling [81], Ets factor binding in vitro [68], but NGF stimulates the differentiation of these cells into sites [82] and AP1 elements [83,84] responding to the ERK/MAPK neuron-like cells and the induction may be indirect. The expression of pathway, and Smad2/3 binding sites involved in TGFβ signaling [85]. tenascin-X by fibroblasts in vitro is not upregulated by a host of Interestingly, many of these transcription factors can also collaborate growth factors, including TGFβ1, bFGF or PDGF, but like tenascin-C its in the regulation of tenascin-C expression, since it was shown that expression can be downregulated by glucocorticoids [69]. However, mutations of either the Smad, Ets or SP1/3 binding sites abrogated there is one report that BDNF is able to upregulate tenascin-X tenascin-C promoter induction by TGFβ [85]. Recently, a transcrip- expression by cultured endothelial cells [70]. tional repressor for tenascin-C was found. Overexpression of GATA6 reduced tenascin-C expression and inhibited IL-4 and TGFβ-induced 6. Signaling pathways used to induce tenascin expression tenascin-C promoter activity [86]. This ubiquitously expressed transcriptional inhibitor could be one of the factors keeping TGFβ1 and BMP-2, which are both members of the TGFβ super- tenascin-C expression low under normal conditions in the absence family, induce the expression of tenascin-C and tenascin-W, respec- of microenvironmental stimuli leading to tenascin-C upregulation. tively (see above and especially [8]). Both growth factors signal through ligand-specific transmembrane receptor serine-threonine 8. Conclusions and future directions kinases. Growth factor binding leads to conformational changes, receptor phosphorylation, and the activation of the Smad complex of This review illustrates that there are two types of tenascins: 1) transcription factors (reviewed by [71]). In contrast, osteoblast gene those that are significantly regulated by the tissue microenviron- expression is regulated by BMP-2 via non-canonical MAPK signaling ment (tenascin-C and tenascin-W), and 2) those that have stabile, pathways (e.g. [72]). Scherberich et al. [8] have shown that this latter restricted expression patterns (tenascin-R and tenascin-X). Accord- pathway is important in the induction of tenascin-W: inhibitors of p38 ingly, it is the former group that is frequently encountered in the and JNK reduce the endogenous expression of tenascin-W by mouse ECM at sites of disease or trauma, especially in the tumor stroma. embryo fibroblasts, and p38 inhibitors block the induction of tenascin- Tenascin-C and tenascin-W are also the types of tenascin with the W expression by BMP-2 [8]. In another system, these pathways can best documented effects on cell proliferation, differentiation and lead to the expression not of tenascin-W, but of tenascin-C. Mutations motility, and these roles may contribute favorably, or not, to the in the BMP type II receptor have been linked to familial pulmonary organism's responses to pathological events. This needs to be taken R.P. Tucker, R. Chiquet-Ehrismann / Biochimica et Biophysica Acta 1793 (2009) 888–892 891 into consideration when designing therapies to combat tumor [22] R. Chiquet-Ehrismann, E.J. Mackie, C.A. Pearson, T. Sakakura, Tenascin: an fl extracellular matrix protein involved in tissue interactions during fetal develop- growth and metastasis or the negative consequences of in amma- ment and oncogenesis, Cell 47 (1986) 131–139. tion. Another exciting potential field for the application of this [23] G. Orend, R. Chiquet-Ehrismann, Tenascin-C induced signaling in cancer, Cancer knowledge is the bioengineering of artificial scaffolds to promote Lett. 244 (2006) 143–163. [24] P.C. Park, M.D. Taylor, T.G. Mainprize, L.E. Becker, M. Ho, W.T. Dura, J. Squire, J.T. successful healing in response to trauma or the integration of Rutka, Transcriptional profiling of medulloblastoma in children, J. Neurosurg. 99 prosthesis into host connective tissues. These can be designed to (2003) 534–541. release the millieau of factors that optimally promotes the [25] T. Minamitani, H. Ariga, K. Matsumoto, Adhesive defect in extracellular matrix fi expression of tenascin-C or tenascin-W, which in turn can lead to tenascin-X-null broblasts: a possible mechanism of tumor invasion, Biol. Pharm. Bull. 25 (2002) 1472–1475. the rapid incorporation of the appropriate host tissue in the scaffold. [26] M. Pennison, B. Pasche, Targeting transforming growth factor-beta signaling, Curr. Future studies should also be directed to better understand the Opin. Oncol. 19 (2007) 579–585. regulation of the other tenascins, which may play important [27] C.A. Pearson, D. Pearson, S. Shibahara, J. Hofsteenge, R. Chiquet-Ehrismann, Tenascin: cDNA cloning and induction by TGF-beta, EMBO J. 7 (1988) 2977–2982. structural or other instructive roles in normal tissues. [28] R.P. Tucker, J.A. Hammarback, D.A. Jenrath, E.J. Mackie, Y. Xu, Tenascin expression in the mouse: in situ localization and induction in vitro by bFGF, J. Cell Sci. 104 (1993) 69–76. References [29] T. Sakai, H. Kawakatsu, M. Ohta, M. Saito, Tenascin induction in tenascin nonproducing carcinoma cell lines in vivo and by TGF-beta 1 in vitro, J. Cell [1] R.P. Tucker, K. Drabikowski, J.F. Hess, J. Ferralli, R. Chiquet-Ehrismann, J.C. Adams, Physiol. 159 (1994) 561–572. Phylogenetic analysis of the tenascin gene family: evidence of origin early in the [30] S. Maschler, S. Grunert, A. Danielopol, H. Beug, G. Wirl, Enhanced tenascin-C chordate lineage, BMC Evol. Biol. 6 (2006) 60. expression and matrix deposition during Ras/TGF-beta-induced progression of [2] R. Chiquet-Ehrismann, Tenascins, Int J Biochem Cell Biol. 36 (2004) 986–990. mammary tumor cells, Oncogene 23 (2004) 3622–3633. [3] R. Chiquet-Ehrismann, M. Chiquet, Tenascins: regulation and putative functions [31] C. Berking, R. Takemoto, H. Schaider, L. Showe, K. Satyamoorthy, P. Robbins, M. during pathological stress, J. Pathol. 200 (2003) 488–499. Herlyn, Transforming growth factor-beta1 increases survival of human melanoma [4] F.S. Jones, P.L. Jones, The tenascin family of ECM glycoproteins: structure, function, through stroma remodeling, Cancer Res. 61 (2001) 8306–8316. and regulation during embryonic development and tissue remodeling, Dev. Dyn. [32] U. Heine, E.F. Munoz, K.C. Flanders, L.R. Ellingsworth, H.Y. Lam, N.L. Thompson, A.B. 218 (2000) 235–259. Roberts, M.B. Sporn, Role of transforming growth factor-beta in the development [5] J. Bristow, W. Carey, D. Egging, J. Schalkwijk, Tenascin-X, collagen, elastin, and of the mouse embryo, J. Cell Biol. 105 (1987) 2861–2876. the Ehlers–Danlos syndrome, Am. J. Med. Genet. C Semin. Med. Genet. 139 [33] J. Chimal-Monroy, L. Díaz de León, Expression of N-cadherin, N-CAM, fibronectin (2005) 24–30. and tenascin is stimulated by TGF-beta1, beta2, beta3 and beta5 during the [6] F.G. Rathjen, J.M. Wolff, R. Chiquet-Ehrismann, Restrictin: a chick neural formation of precartilage condensations, Int. J. Dev. Biol. 43 (1999) 59–67. extracellular matrix protein involved in cell attachment co-purifies with the cell [34] G. Vollmer, M.I. Tan, W. Wünsche, K. Frank, Expression of tenascin-C by human recognition molecule F11, Development 113 (1991) 151–164. endometrial adenocarcinoma and stroma cells: heterogeneity of splice variants [7] A. Scherberich, R.P. Tucker, E. Samandari, M. Brown-Luedi, D. Martin, R. Chiquet- and induction by TGF-beta, Biochem. Cell Biol. 75 (1997) 759–769. Ehrismann, Murine tenascin-W: a novel mammalian tenascin expressed in kidney [35] R.P. Tucker, J. Spring, S. Baumgartner, D. Martin, C. Hagios, P.M. Poss, R. Chiquet- and at sites of bone and smooth muscle development, J. Cell Sci. 117 (2004) Ehrismann, Novel tenascin variants with a distinctive pattern of expression in the 571–581. avian embryo, Development 120 (1994) 637–647. [8] A. Scherberich, R.P. Tucker, M. Degen, M. Brown-Luedi, A.C. Andres, R. [36] D. Fischer, R.P. Tucker, R. Chiquet-Ehrismann, J.C. Adams, Cell-adhesive responses Chiquet-Ehrismann, Tenascin-W is found in malignant mammary tumors, to tenascin-C splice variants involve formation of fascin microspikes, Mol. Biol. promotes alpha8 integrin-dependent motility and requires p38MAPK activity Cell. 8 (1997) 2055–2075. for BMP-2 and TNF-alpha induced expression in vitro, Oncogene 24 (2005) [37] T. Tsunoda, H. Inada, I. Kalembeyi, K. Imanaka-Yoshida, M. Sakakibara, R. Okada, K. 1525–1532. Katsuta, T. Sakakura, Y. Majima, T. Yoshida, Involvement of large tenascin-C splice [9] M. Degen, F. Brellier, R. Kain, C. Ruiz, L. Terracciano, G. Orend, R. Chiquet- variants in breast cancer progression, Am. J. Pathol. 162 (2003) 1857–1867. Ehrismann, Tenascin-W is a novel marker for activated tumor stroma in low-grade [38] R.J. Akhurst, S.A. Lehnert, A. Faissner, E. Duffie, TGF beta in murine morphogenetic human breast cancer and influences cell behavior, Cancer Res. 67 (2007) processes: the early embryo and cardiogenesis, Development 108 (1990) 645–656. 9169–9179. [39] H. Yoshida, T. Yoshida, T. Iizuka, T. Sakakura, S. Fujita, The expression of [10] M. Degen, F. Brellier, S. Schenk, R. Driscoll, K. Zaman, R. Stupp, L. Tornillo, L. transforming growth factor beta (TGF-beta) in the synovial membrane of Terracciano, R. Chiquet-Ehrismann, C. Rüegg, W. Seelentag, Tenascin-W, a new human temporomandibular joint with internal derangement: a comparison marker of cancer stroma, is elevated in sera of colon and breast cancer patients, with tenascin expression, J. Oral. Rehabil. 26 (1999) 814–820. Int. J. Cancer (2008) in press. [40] Y. Soini, D. Kamel, M. Apaja-Sarkkinen, I. Virtanen, V.P. Lehto, Tenascin immunor- [11] J.E. Murphy-Ullrich, V.A. Lightner, I. Aukhil, Y.Z. Yan, H.P. Erickson, M. Höök, Focal eactivity in normal and pathological bone marrow, J. Clin. Pathol. 46 (1993) 218–221. adhesion integrity is downregulated by the alternatively spliced domain of human [41] R.A. Walker, S.J. Dearing, B. Gallacher, Relationship of transforming growth factor tenascin, J. Cell Biol. 115 (1991) 1127–1136. beta 1 to extracellular matrix and stromal infiltrates in invasive breast carcinoma, [12] P. Pesheva, R. Probstmeier, A.P. Skubitz, J.B. McCarthy, L.T. Furcht, M. Schachner, Br. J. Cancer. 69 (1994) 1160–1165. Tenascin-R (J1 160/180) inhibits fibronectin-mediated cell adhesion—functional [42] K.E. Wilson, J.M. Bartlett, E.P. Miller, J.F. Smyth, P. Mullen, W.R. Miller, S.P. Langdon, relatedness to tenascin-C, J. Cell Sci. 107 (1994) 2323–2333. Regulation and function of the extracellular matrix protein tenascin-C in ovarian [13]R.P.Tucker,C.Hagios,A.Santiago,R.Chiquet-Ehrismann,Tenascin-Yis cancer cell lines, Br. J. Cancer. 80 (1999) 685–692. concentrated in adult nerve roots and has barrier properties in vitro, J. Neurosci. [43] P. Hau, L.A. Kunz-Schughart, P. Rümmele, F. Arslan, A. Dörfelt, H. Koch, A. Lohmeier, Res. 66 (2001) 439–447. B. Hirschmann, A. Müller, U. Bogdahn, A.K. Bosserhoff, Tenascin-C protein is [14] A. Trebaul, E.K. Chan, K.S. Midwood, Regulation of fibroblast migration by induced by transforming growth factor-beta1 but does not correlate with time to tenascin-C, Biochem Soc Trans. 35 (2007) 695–697. tumor progression in high-grade gliomas, J. Neurooncol. 77 (2006) 1–7. [15] C.V. Meloty-Kapella, M. Degen, R. Chiquet-Ehrismann, R.P. Tucker, Avian tenascin- [44] F. Wehrhan, F. Rödel, G.G. Grabenbauer, K. Amann, W. Brückl, S. Schultze-Mosgau, W: expression in smooth muscle and bone, and effects on calvarial cell spreading Transforming growth factor beta 1 dependent regulation of Tenascin-C in and adhesion in vitro, Dev. Dyn. 235 (2006) 1532–1542. radiation impaired wound healing, Radiother. Oncol. 72 (2004) 297–303. [16] C. Ruiz, W. Huang, M.E. Hegi, K. Lange, M.F. Hamou, E. Fluri, E.J. Oakeley, R. [45] I. Kalembeyi, H. Inada, R. Nishiura, K. Imanaka-Yoshida, T. Sakakura, T. Yoshida, Chiquet-Ehrismann, G. Orend, Growth promoting signaling by tenascin-C, Cancer Tenascin-C upregulates -9 in breast cancer cells: direct Res. 64 (2004) 7377–7385. and synergistic effects with transforming growth factor beta1, Int. J. Cancer 105 [17] K.S. Midwood, J.E. Schwarzbauer, Tenascin-C modulates matrix contraction via (2003) 53–60. focal adhesion kinase- and Rho-mediated signaling pathways, Mol. Biol. Cell. 13 [46] D. Ribatti, A. Vacca, M. Rusnati, M. Presta, The discovery of basic fibroblast growth (2002) 3601–3613. factor/fibroblast growth factor-2 and its role in haematological malignancies, [18] W. Huang, R. Chiquet-Ehrismann, J.V. Moyano, A. Garcia-Pardo, G. Orend, Cytokine Growth Factor Rev. 18 (2007) 327–334. Interference of tenascin-C with syndecan-4 binding to fibronectin blocks cell [47] S. Meiners, M. Marone, J.L. Rittenhouse, H.M. Geller, Regulation of astrocytic adhesion and stimulates tumor cell proliferation, Cancer Res. 61 (2001) tenascin by basic fibroblast growth factor, Dev. Biol. 160 (1993) 480–493. 8586–8594. [48] M. Mahler, L. Ferhat, A. Gillian, Y. Ben-Ari, A. Represa, Tenascin-C mRNA and [19] K.S. Midwood, L.V. Valenick, H.C. Hsia, J.E. Schwarzbauer, Coregulation of tenascin-C protein immunoreactivity increase in astrocytes after activation by fibronectin signaling and matrix contraction by tenascin-C and syndecan-4, bFGF, Cell Adhes. Commun. 4 (1996) 175–186. Mol. Biol. Cell. 15 (2004) 5670–5677. [49] W.J. Rettig, H.P. Erickson, A.P. Albino, P. Garin-Chesa, Induction of human tenascin [20] G. Orend, W. Huang, M.A. Olayioye, N.E. Hynes, R. Chiquet-Ehrismann, Tenascin-C (neuronectin) by growth factors and cytokines: cell type-specific signals and blocks cell-cycle progression of anchorage-dependent fibroblasts on fibronectin signalling pathways, J. Cell Sci. 107 (1994) 487–497. through inhibition of syndecan-4, Oncogene 22 (2003) 3917–3926. [50] S. Suzuki, A.J. Li, M. Ikemoto, T. Imamura, Expression of tenascin-C long isoforms is [21] K. Lange, M. Kammerer, M.E. Hegi, S. Grotegut, A. Dittmann, W. Huang, E. Fluri, G. induced in the hypothalamus by FGF-1, Neuroreport 13 (2002) 1041–1045. W. Yip, M. Götte, C. Ruiz, G. Orend, Endothelin receptor type B counteracts [51] K.E. Wilson, S.P. Langdon, A.M. Lessells, W.R. Miller, Expression of the extracellular tenascin-C-induced endothelin receptor type A-dependent focal adhesion and matrix protein tenascin in malignant and benign ovarian tumours, Br. J. Cancer 74 actin stress fiber disorganization, Cancer Res. 67 (2007) 6163–6173. (1996) 999–1004. 892 R.P. Tucker, R. Chiquet-Ehrismann / Biochimica et Biophysica Acta 1793 (2009) 888–892

[52] L.B. Derr, R. Chiquet-Ehrismann, R. Gandour-Edwards, J. Spence, R.P. Tucker, The [70] K. Takeda, H. Shiba, N. Mizuno, N. Hasegawa, Y. Mouri, A. Hirachi, H. Yoshino, H. expression of tenascin-C with the AD1 variable repeat in embryonic tissues, cell Kawaguchi, H. Kurihara, Brain-derived neurotrophic factor enhances periodontal lines and tumors in various vertebrate species, Differentiation 62 (1997) 71–82. tissue regeneration, Tissue Eng. 11 (2005) 1618–1629. [53] M.A. Ghert, S.T. Jung, W. Qi, J.M. Harrelson, H.P. Erickson, J.A. Block, S.P. Scully, The [71] L. Mishra, K. Shetty, Y. Tang, A. Stuart, S.W. Byers, The role of TGF-beta and clinical significance of tenascin-C splice variant expression in chondrosarcoma, Wnt signaling in gastrointestinal stem cells and cancer, Oncogene 24 (2005) Oncology 61 (2001) 306–314. 5775–5789. [54] C. Dang, M. Gottschling, J. Roewert, T. Forschner, E. Stockfleth, I. Nindl, Tenascin-C [72] S. Gallea, F. Lallemand, A. Atfi, G. Rawadi, V. Ramez, S. Spinella-Jaegle, S. Kawai, C. patterns and splice variants in actinic keratosis and cutaneous squamous cell Faucheu, L. Huet, R. Baron, S. Roman-Roman, Activation of mitogen-activated carcinoma, Br. J. Dermatol. 155 (2006) 763–770. protein kinase cascades is involved in regulation of bone morphogenetic protein- [55] D.W. LaFleur, J.A. Fagin, J.S. Forrester, S.A. Rubin, B.G. Sharifi, Cloning and 2-induced osteoblast differentiation in pluripotent C2C12 cells, Bone 28 (2001) characterization of alternatively spliced isoforms of rat tenascin. Platelet-derived 491–498. growth factor-BB markedly stimulates expression of spliced variants of tenascin [73] J.R. Runo, J.E. Loyd, Primary pulmonary hypertension, Lancet 361 (2003) mRNA in arterial smooth muscle cells, J. Biol. Chem. 269 (1994) 20757–20763. 1533–1544. [56] N.P. Pringle, W.D. Richardson, A singularity of PDGF alpha-receptor expression in [74] P.L. Jones, M. Rabinovitch, Tenascin-C is induced with progressive pulmonary the dorsoventral axis of the neural tube may define the origin of the vascular disease in rats and is functionally related to increased smooth muscle cell oligodendrocyte lineage, Development 117 (1993) 525–533. proliferation, Circ. Res. 79 (1996) 1131–1142. [57] R.P. Tucker, S.E. McKay, The expression of tenascin by cells and , [75] K. Ihida-Stansbury, D.M. McKean, K.B. Lane, J.E. Loyd, L.A. Wheeler, N.W. Morrell, P. Development 112 (1991) 1031–1039. L. Jones, Tenascin-C is induced by mutated BMP type II receptors in familial forms [58] E. Garcion, A. Faissner, C. ffrench-Constant, Knockout mice reveal a contribution of of pulmonary arterial hypertension, Am. J. Physiol. Lung Cell Mol. Physiol. 291 the extracellular matrix molecule tenascin-C to neural precursor proliferation and (2006) L694–702. migration, Development 128 (2001) 2485–2496. [76] A. Sarasa-Renedo, V. Tunç-Civelek, M. Chiquet, Role of RhoA/ROCK-dependent [59] Y. Nakamura, S. Esnault, T. Maeda, E.A. Kelly, J.S. Malter, N.N. Jarjour, Ets-1 actin contractility in the induction of tenascin-C by cyclic tensile strain, Exp. Cell regulates TNF-alpha-induced matrix metalloproteinase-9 and tenascin expression Res. 312 (2006) 1361–1370. in primary bronchial fibroblasts, J. Immunol. 172 (2004) 1945–1952. [77] B. Sivasankaran, M. Degen, A. Ghaffari, M.E. Hegi, M.-F. Hamou, M.-C.S. Ionescu, C. [60] Y. Nakoshi, M. Hasegawa, A. Sudo, T. Yoshida, A. Uchida, Regulation of tenascin-C Zweifel, M. Tolnay, M. Wasner, S. Mergenthaler, A.R. Miserez, R. Kiss, M.M. Lino, A. expression by -alpha in cultured human osteoarthritis Merlo, R. Chiquet-Ehrismann, J.L. Boulay, Tenascin-C is a novel RBPJκ-induced chondrocytes, J. Rheumatol. (2007) (e-publication). target gene for Notch signaling in gliomas, Cancer Res. (in press). [61] E. Härkönen, I. Virtanen, A. Linnala, L.L. Laitinen, V.L. Kinnula, Modulation of [78] S.F. Tavazoie, C. Alarcón, T. Oskarsson, D. Padua, Q. Wang, P.D. Bos, W.L. Gerald, J. fibronectin and tenascin production in human bronchial epithelial cells by Massagué, Endogenous human microRNAs that suppress breast cancer metastasis, inflammatory cytokines in vitro, Am. J. Respir. Cell. Mol. Biol. 13 (1995) 109–115. Nature 451 (2008) 147–152. [62] A. Gratchev, J. Kzhyshkowska, J. Utikal, S. Goerdt, Interleukin-4 and dexametha- [79] P. Putthoff, N. Akyüz, M. Kutsche, L. Zardi, U. Borgmeyer, M. Schachner, Structure of sone counterregulate extracellular matrix remodelling and phagocytosis in type-2 the murine tenascin-R gene and functional characterisation of the promoter, macrophages, Scand. J. Immunol. 61 (2005) 10–17. Biochem. Biophys. Res. Commun. 308 (2003) 940–949. [63] R. Fässler, T. Sasaki, R. Timpl, M.L. Chu, S. Werner, Differential regulation of fibulin, [80] S.D. Wijesuriya, J. Bristow, W.L. Miller, Localization and analysis of the principal tenascin-C, and nidogen expression during wound healing of normal and promoter for human tenascin-X, Genomics 80 (2002) 443–452. glucocorticoid-treated mice, Exp. Cell Res. 222 (1996) 111–116. [81] D.M. McKean, L. Sisbarro, D. Ilic, N. Kaplan-Alburquerque, R. Nemenoff, M. Weiser- [64] J.F. Talts, A. Weller, R. Timpl, M. Ekblom, P. Ekblom, Regulation of mesenchymal Evans, M.J. Kern, P.L. Jones, FAK induces expression of Prx1 to promote tenascin-C- extracellular matrix protein synthesis by transforming growth factor-beta and dependent fibroblast migration, J. Cell Biol. 161 (2003) 393–402. glucocorticoids in tumor stroma, J. Cell Sci. 108 (1995) 2153–2162. [82] G. Watanabe, H. Nishimori, H. Irifune, Y. Sasaki, S. Ishida, H. Zembutsu, T. Tanaka, S. [65] M. Ekblom, R. Fässler, B. Tomasini-Johansson, K. Nilsson, P. Ekblom, Down- Kawaguchi, T. Wada, J. Hata, M. Kusakabe, K. Yoshida, Y. Nakamura, T. Tokino, regulation of tenascin expression by glucocorticoids in bone marrow stromal cells Induction of tenascin-C by tumor-specificEWS–ETS fusion , Genes and in fibroblasts, J. Cell Biol. 123 (1993) 1037–1045. Cancer 36 (2003) 224–232. [66] A. Laitinen, A. Altraja, M. Kämpe, M. Linden, I. Virtanen, L.A. Laitine, Tenascin is [83] R. Chiquet-Ehrismann, M. Tannheimer, M. Koch, A. Brunner, J. Spring, D. Martin, S. increased in airway basement membrane of asthmatics and decreased by an Baumgartner, M. Chiquet, Tenascin-C expression by fibroblasts is elevated in inhaled steroid, Am. J. Respir. Crit. Care Med. 156 (1997) 951–958. stressed collagen gels, J. Cell Biol. 127 (1994) 2093–2101. [67] P. Weber, D. Montag, M. Schachner, R.R. Bernhardt, Zebrafish tenascin-W, a new [84] R. Chiquet-Ehrismann, C. Hagios, S. Schenk, The complexity in regulating the member of the tenascin family, J. Neurobiol. 35 (1998) 1–16. expression of tenascins, Bioessays 17 (1995) 873–878. [68] L.B. Derr, L.A. McKae, R.P. Tucker, The distribution of tenascin-R in the developing [85] M. Jinnin, H. Ihn, Y. Asano, K. Yamane, M. Trojanowska, K. Tamaki, Tenascin-C avian nervous system, J. Exp. Zool. 280 (1998) 152–164. upregulation by transforming growth factor-beta in human dermal fibroblasts [69] T. Sakai, Y. Furukawa, R. Chiquet-Ehrismann, M. Nakamura, S. Kitagawa, T. involves Smad3, Sp1, and Ets1, Oncogene 23 (2004) 1656–1667. Ikemura, K. Matsumoto, Tenascin-X expression in tumor cells and fibroblasts: [86] A. Ghatnekar, M. Trojanowska, GATA-6 is a novel transcriptional repressor of the glucocorticoids as negative regulators in fibroblasts, J. Cell Sci. 109 (1996) human Tenascin-C gene expression in fibroblasts, Biochim. Biophys. Acta 1779 2069–2077. (2008) 145–151.