MATBIO-01044; No of Pages 14 Matrix Biology xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Matrix Biology

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

Invoking the power of thrombospondins: Regulation of thrombospondins expression

Olga Stenina-Adognravi ⁎

Department of Molecular Cardiology, Cleveland Clinic, 9500 Euclid Ave NB50, Cleveland, OH 44195, United States article info abstract

Article history: Increasing evidence suggests critical functions of thrombospondins (TSPs) in a variety of physiological and path- Received 16 December 2013 ological processes. With the growing understanding of the importance of these matricellular , the need to Received in revised form 5 February 2014 understand the mechanisms of regulation of their expression and potential approaches to modulate their levels is Accepted 8 February 2014 also increasing. The regulation of TSP expression is multi-leveled, cell- and tissue-specific, and very precise. Available online xxxx However, the knowledge of mechanisms modulating the levels of TSPs is fragmented and incomplete. This Keywords: review discusses the known mechanisms of regulation of TSP levels and the gaps in our knowledge that prevent Thrombospondin us from developing strategies to modulate the expression of these physiologically important proteins. Regulation of expression © 2014 Elsevier B.V. All rights reserved. Transcription Translation miRNA

1. Introduction important physiological and pathological processes, as well as under- standing that cells do not function in isolation from their environment Thrombospondins (TSPs) are secreted extracellular proteins that be- and ECM. long to a group of matricellular proteins — proteins that are present in The human TSP family consists of five members (TSP-1, TSP-2, the extracellular matrix (ECM), but do not play a primary structural TSP-3, TSP-4, and TSP-5, or cartilage oligomeric matrix protein, COMP), role as other ECM proteins do. Matricellular proteins, including TSPs, which are divided into two subgroups based on their domain structure have diverse functions that integrate ECM and cells: they interact with (Adams, 2001; Adams and Lawler, 2004, 2011) (TSP-1 and TSP-2 belong a number of cell surface receptors and with a variety of ECM proteins to subgroup A, while TSP-3, TSP-4, and TSP-5 compose subgroup B). In (e.g., structural proteins such as collagens, proteases, and growth lower organisms, TSPs are represented by a single protein, as in sponge factors). The revolutionary classification of matricellular proteins pro- (Bentley and Adams, 2010), or by multiple family members, e.g., five posedbylatePaulBornstein(Sage and Bornstein, 1991; Bornstein, proteins in sea anemone (Tucker et al., 2013), that have a structure sim- 1995) has promoted a better appreciation of ECM and its role in ilar to human TSPs of subgroup B. The second subgroup A appears later in the evolutionary development, coinciding with the development of the vascular system (Bentley and Adams, 2010), and develops further as a re- flection of the development of cardiovascular and immune systems wherethisgroupplaysanimportantrole. Abbreviations: AhR, Aryl (aromatic) hydrocarbon receptor; ARE, Adenylate-uridylate- rich element; ATF-1, Activating Transcription Factor-1; CBF, CCAAT-binding factor; COMP, Important physiological functions of TSPs have been discovered in Cartilage oligomeric matrix protein; Cyp1B1, Cytochrome P450 1B1; EBOX, Enhancer box; many organs and systems [reviewed in (Henkin and Volpert, 2011; ECM, Extracellular matrix; Egr-1, Early growth response protein 1; EGRF, Early growth re- Frangogiannis, 2012; Mayer et al., 2013; Stenina-Adognravi, 2013)]. sponse factor 1; eNOS, Endothelial nitric oxide synthase; ER, Endoplasmic reticulum; H3, Regulation of angiogenesis and cancer progression (Lawler and Histone 3; HDAC, Histone Deacetylase; HGF, Hepatocyte growth factor; HuR, Human anti- Lawler, 2012), regulation of inflammation (Frolova et al., 2010; Lopez- gen R; IR, Ischemia/reperfusion; MAPK, Mitogen-activated protein kinases; MI, Myocardial infarction; MYC, Myelocytomatosis oncogene; NO, Nitric oxide; NR4A2, Nuclear receptor Dee et al., 2011; Mustonen et al., 2012; Stenina-Adognravi, 2013; subfamily 4, group A, member 2; PAH, Pulmonary arterial hypertension; PAR-1, Vanhoutte et al., 2013), modulation of immune response (Martin- Protease-activated receptor-1; SPARC, Secreted protein acidic and rich in cysteine; TGFβ, Manso et al., 2012; Miller et al., 2013), formation of myotendinous junc- Transforming growth factor beta; TRPC4, Short transient receptor potential channel 4; tions (Subramanian et al., 2007), maintenance of the myocardium TSP, Thrombospondin; UTR, Untranslated region; WT1, Wilms' tumor suppressor; YY-1, integrity and function (Schroen et al., 2004; Chatila et al., 2007; Yin Yang 1. ⁎ Tel.: +1 216 444 9057 (office); fax: +1 216 445 8204. Swinnen et al., 2009; Cingolani et al., 2011; van Almen et al., 2011a; E-mail address: [email protected]. Frolova et al., 2012; Lynch et al., 2012; Roberts et al., 2012), regulation

http://dx.doi.org/10.1016/j.matbio.2014.02.001 0945-053X/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 2 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx of fibrosis (Sweetwyne and Murphy-Ullrich, 2012), and synaptogenesis (Frolova et al., 2010). Similarly, in the wall of smaller blood vessels (Risher and Eroglu, 2012) are just a few examples of their roles in TSP-3, TSP-4, and TSP-5 not only are produced by different cell types, physiology and pathology. To participate in the critical physiological but also are deposited in ECM in distinct patterns and in distinct locali- processes, TSPs have to be present in the right location at the right zation within the vessel (Frolova, in press). In tendon, both TSP-3 and time. This review summarizes the published information about the TSP-4 are abundant, but are arranged in fibers of different orientations, regulation of the expression of TSPs. stressing the distinctions in their functions (Frolova, in press). When TSPs are expressed in the same structures at the same time [e.g., TSP-4 2. Similarities and distinctions between TSP family members and TSP-5 in tendon (Frolova, in press)], it is still unclear why both are required and what differential properties of tissue they support. All TSPs share a high degree of homology at the protein level, espe- Differences in the expression profiles of individual TSPs in cancers cially in their “signature domain”—the C-terminal half of the human and other cells and tissues [e.g., (Carron et al., 2000; Agah et al., 2002; protein that includes 3–4EGF-likerepeats[1–5 in other species Bornstein et al., 2004)] suggest that TSPs have differential functions in (Adams and Lawler, 2011)], the Ca2+-binding domains with up to 26 pathological processes, despite shared homologous domains and li- metal binding sites per protein subunit, and the globular C-terminal do- gands. Several expression studies demonstrated opposite profiles main [reviewed in (Adams and Lawler, 2011)]. Table 1 summarizes the for TSP-1 and TSP-4 in brain (Bredel et al., 2005; Liang et al., 2005; extent of homology between five TSPs. However, apart from the protein Sun et al., 2006) and breast (Sorlie et al., 2001; Turashvili et al., 2007; coding regions of the TSP (THBS), there is no homology in their Ma et al., 2009) cancers. Examination of the datasets from these studies DNA sequence, and the regulatory parts of TSP genes, either the reveals that TSP-4 upregulation is accompanied by downregulation of untranslated regions of mRNA (UTR) or the gene promoters, are dis- TSP-1 in the tumor samples (www.oncomine.org), clearly indicating tinctly different at the first glance (Adolph et al., 1997). Search for com- distinct, probably even opposite, functions for these two proteins. Unex- mon DNA sequence motifs using SCOPE motif finder (http://genie. pectedly, TSP-2, belonging to subgroup A and sharing high homology dartmouth.edu/scope)(Carlson et al., 2007; Chakravarty et al., 2007) with TSP-1, exhibits a profile similar to the TSP-4 profile — it is upregu- and comparison of the positions of common DNA sequence motifs in lated in both brain and breast cancers, with the exception of one data set the promoters of five TSPs reveal some similarities between TSP pro- (Bredel et al., 2005). In some datasets TSP-2 was in top 1% of upregulat- moters and suggest that in some conditions two or more TSPs may be ed genes, together with TSP-4. These comparisons of the three TSP pro- regulated by the same stimuli simultaneously (Fig. 1). However, there files from the same datasets clearly demonstrate that the three TSPs are no experimental data to support the predicted similarity of the play important distinct roles in cancer growth, and the regulation of DNA motifs. their expression is an efficient mechanism to support these still poorly The five proteins are located on different (human understood functions (with the exception of TSP-1, whose downregula- THBS1 on 15 and mouse Thbs1 on chromosome 2, tion is known to support angiogenesis in tumors). TSP-5 was upregulat- human THBS2 on chromosome 6 and mouse Thbs2 on chromosome ed similar to TSP-4 in multiple datasets from breast cancer studies 17, human THBS3 on and mouse Thbs3 on chromosome (www.oncomine.org), while TSP-3 data were inconsistent, and both 3, human THBS4 on chromosome 5 and mouse Thbs4 on chromosome 13, down-regulation and up-regulation were observed (Ramaswamy human THBS5 on chromosome 19 and mouse Thbs5 on chromosome 8). et al., 2003; Yu et al., 2008). In most tissues, TSPs are expressed at a low level compared to other The expression studies in human samples and in animal tissues non-structural ECM proteins, e.g., SPARC, tenascin C and fibronectin clearly suggest distinct functions and roles in physiological and patho- (Fig. 2)(Su et al., 2002). Similar to TSPs, tenascin C and SPARC are logical processes for five TSPs. However, the molecular mechanisms representatives of the matricellular protein family. SPARC and fibronec- responsible for the differences in TSPs functions still have to be ad- tin regulate collagen deposition and ECM assembly and support the in- dressed and confirmed by examining specific functions of all five TSPs teraction between cells and ECM, similar to TSPs. These proteins have and by studying their interactions with cells and ligands. Precise regula- similar functions in disease regulation: e.g., they support interaction of tion of TSP expression is especially important in a view of TSP homology cancer cells with stromal cells. and a number of shared ligands. Emphasizing the potent effect of TSP presence in the tissue and the need to tightly regulate their expression, there are rapid mechanisms 3. Mechanisms of regulation of TSP expression upregulating TSPs at the transcriptional level (Raugi et al., 1987; Stenina et al., 2003a; Dabir et al., 2008) and mechanisms rapidly Most of the information about the regulation of TSP expression degrading mRNA or blocking its translation into a protein (Janz et al., comes from descriptive studies that either used array type experiments 2000; Dews et al., 2006; Bhattacharyya et al., 2008; Sanghamitra to identify highly upregulated or downregulated genes in tissues of Bhattacharyya et al., 2012). The proteins appear to be unstable after interest or specifically looked at the TSP expression (in most cases they are secreted, stressing the importance of timely elimination of expression of TSP-1), based on known functions of TSPs. However, a TSPs from the ECM and cell environment (Murphy-Ullrich and Mosher, few reports address specific molecular mechanisms of the regulation 1987a, 1987b; Manni et al., 2007). In adult organisms, upregulation of of TSP expression in cells and tissues. TSPs is associated with specific stages of wound healing [e.g., (Raugi et al., 1987; Reed et al., 1993; DiPietro et al., 1996; Kyriakides et al., 3.1. Transcriptional regulation 1999; Streit et al., 2000; Agah et al., 2002)] and tissue remodeling [e.g., (Stenina et al., 2003b; Rysa et al., 2005; Mustonen et al., 2008; 3.1.1. TSP-1 Frolova et al., 2010; Cingolani et al., 2011; Lynch et al., 2012; Pohjolainen et al., 2012)], in which it can be either protective and 3.1.1.1. Positive regulation. The first analysis of TSP-1 promoter and the beneficial or detrimental. identification of the serum response element were reported by Distinct localization of TSPs in tissues suggests differential functions, Framson and Bornstein (1993).Theidentified serum response element despite the high homology between proteins and a number of shared is bi-partite and includes a distal element at the position −1280 and a cell surface receptors and binding partners. Even when two or more proximal element with NF-Y binding site at the position −65 (Fig. 3). TSPs are found in the same tissue, they are localized to the different Interestingly, the proximal element is different in the mouse TSP-1 pro- cell types or structures within the tissue. For example, both TSP-3 moter: it harbors Egr-1 binding site in the position of human NF-Y site and TSP-5 are present in the atherosclerotic lesion of ApoE−/− mice, and does not participate in serum response. The Egr-1 binding site but they are clearly produced by different cell types in the lesion maintained constitutive activity of the promoter acting in concert with

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx 3

Table 1 For example, in response to high glucose, TSP-1 mRNA is upregulated Homology of human TSP proteins [calculated using algorithm described in (Huang and in major cell types of the vascular wall — endothelial cells, smooth Miller, 1991)]. muscle cells and fibroblasts (Stenina et al., 2003a). However, the time TSP-1 TSP-2 TSP-3 TSP-4 of upregulation is distinctly different — in endothelial cells upregulation TSP-1 can be detected earlier than in smooth muscle cells. The analysis of the TSP-2 62.6 promoter revealed that in two cell types the promoter activity is regu- TSP-3 52.2 52.9 lated by different transcriptional protein complexes. In endothelial TSP-453.952.661 cells, a proximal regulatory region of the promoter binding aryl hydro- TSP-5 53.6 54.4 65.3 69.9 carbon receptor is sufficient to drive the transcription (Dabir et al., 2008; Raman et al., 2011), while in smooth muscle cells the same promoter region is interacting with a distal part of the promoter the adjacent GC-rich region binding SP1 (Shingu and Bornstein, 1994). through the formation of a single protein complex between transcrip- The importance of the first intron for the efficient transcription of the tion factors binding to the proximal and to the distal regions (Raman gene was noted (Laherty et al., 1989): the deletion of this region results et al., 2007; Raman et al., 2011). The regulation is determined by a in 4-fold decrease in the reporter production, suggesting a cis-acting cell-type-specificprofile of the expression and the activation of positive element in the first intron. transcription factors. AP-1 binding to TSP-1 gene promoter mediates the activation of Transcriptional upregulation of TSP-1 in response to high glucose is the promoter in response to protein kinase C (PKC) activation in probably the most dissected pathway among all the pathways regulat- human hepatoma cells (Kim et al., 2001). However, a similar study ing TSP-1 expression. TSP-1 has been implicated in the development of the effects of PKC activation in porcine aortic endothelial cells re- of vascular complications of diabetes, and its mRNA was dramatically sulted in opposite conclusions: a specific region of the promoter was upregulated in hyperglycemia in various tissues and organs (Poczatek responsible for the downregulation of the promoter activity (Kim et al., 2000; Stenina et al., 2003a; Wang et al., 2003; Daniel et al., and Hong, 2000). The cell-specificity is a very common theme in 2004; Wang et al., 2004; Wang et al., 2006; Zhou et al., 2006; the studies of TSP expression. The cell-and tissue-specific differences Belmadani et al., 2007; Bhattacharyya et al., 2008; Raman et al., in regulation reflect the multi-functional nature of these proteins 2011; Chavez et al., 2012; Sanghamitra Bhattacharyya et al., 2012; and the importance of a very strictly localized and timely production Sweetwyne and Murphy-Ullrich, 2012). The initial observation of and elimination of TSPs. upregulation of TSP-1 in response to high glucose in mesangial cells The cell-specificity of TSP-1 gene promoter regulation was reported (Poczatek et al., 2000) was later confirmed in multiple cell types and tis- in several studies. The activity of a promoter is determined not only by sues. The precise transcriptional mechanisms appear to vary depending its sequence, but also by the availability of target binding sites in a spe- on the cell type, with several transcriptional factors and regulatory re- cific cell type, presence of transcription factors capable of binding the gions being constant independently of the cell type — e.g., upstream promoter, and, finally, cell-specific signals to activate the transcription stimulatory factors (USFs) (Wang et al., 2004) that are common media- factors or protein co-activators. The regulatory complex assembled on tors of glucose effects. Another transcription factor activated by high TSP-1 gene promoter is often cell-specific, and the promoter regions glucose and essential for TSP-1 gene promoter activation is aryl hydro- involved in regulation are also different depending on the cell type. carbon receptor, or AhR (Dabir et al., 2008; Raman et al., 2011). The

Fig. 1. Common DNA sequence motifs in the promoters of TSPs. The analysis of the promoters of TSP-1, TSP-2, TSP-3, TSP-4 and COMP was performed using SCOPE motif finder (http://genie.dartmouth.edu/scope)(Carlson et al., 2007; Chakravarty et al., 2007). Motifs positioned similarly in the promoters of two or more TSPs are shown in the figure (6 out of 20 most significant matches).

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 4 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx

Fig. 2. Expression of TSPs in human tissues and organs, comparison to tenascin C, SPARC and fibronectin expression. The figure was prepared using the data from the database http://expression.gnf.org (Su et al., 2002). Organs from left to right: cerebellum, whole brain, cortex, caudate nucleus, amygdala, thalamus, corpus collosum, spinal cord, whole blood, testis, pancreas, placenta, pituitary gland, thyroid, prostate, ovary, uterus, DRG, salivary gland, trachea, lung, thymus, spleen, adrenal gland, kidney, liver, and heart. activation of the AhR pathway may provide a mechanistic connection factor that mediates many effects of hypoxia in complexes with Arnt). between glucose and hypoxia regulatory pathways (AhR is a ligand for AhR is a member of the Clock family of proteins and can form complexes HIF1β or Arnt, whose other well studied ligand is HIF1α, a transcription with several other proteins from this family that regulate circadian rhythms. The activation of AhR by glucose may provide insights into the well-known connection between food intake and maintenance of the circadian rhythms. TSP-1, as a major and highly upregulated target of AhR, may play a role in this regulation. Increasingly appreciated significance of TSP-1 function in metabolic disorders (Moura et al., 2008; Li et al., 2011) prompted an investigation of the transcriptional regulation of TSP-1 gene by leptin, a hormone im- plicated in the development of obesity and diabetes. Leptin induced TSP-1 at the transcriptional level through the JAK2/ERK/JNK-dependent mechanism (Chavez et al., 2012). The promoter of TSP-1 gene has an active Egr-1 binding site. The promoter activity is upregulated by Egr-1-inducing stimuli (Moon et al., 2005). TSP-1 mRNA can be very rapidly (within 10–15 min) upregulated in response to injury, both in vivo and in vitro (Stenina Fig. 3. Promoter of TSP-1: regulatory regions and experimentally confirmed binding sites et al., 2003a), and, although not investigated, this upregulation most for transcription factors. probably requires Egr-1, one of the first-response transcription factors.

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx 5

Egr-1 is responsible for transcriptional upregulation of TSP-1 by transcriptional factor Id1 that is known to regulate tumor angiogenesis thrombin, together with MYC, in a thrombin receptor PAR-1, G(i/o), (Volpert et al., 2002). G(q), EBOX/EGRF-signaling cascade (McLaughlin et al., 2005). Importantly, transcriptional repression is a common mechanism TSP-1 is a potent anti-angiogenic protein, and most of the reports on regulating the expression of TSP-1 — it results in a rapid effect, because its expression and transcriptional regulation are related to the regula- both the protein and mRNA of TSP-1 are unstable (Raman et al., 2007; El tion of angiogenesis in various tissues and pathological conditions. It is Btaouri et al., 2011; McGray et al., 2011a; Raman et al., 2011; Chavez difficult to judge whether the focus on its anti-angiogenic function is et al., 2012). due to the fact that this is truly the main function of TSP-1, whether MicroRNA regulation is usually associated with the modulation of this is due to the current popularity of the angiogenesis topics, or mRNA translation or stability. However, miR-182 was reported to whether this is a result of a lack of sufficient understanding and knowl- regulate the transcription of TSP-1 gene in colon cancer by modulating edge about other functions of TSP-1. TSP-1 inhibits angiogenesis even in the function of transcription factors Egr-1 and Sp-1 function and their the presence of pro-angiogenic factors (Almog et al., 2006; Zaslavsky binding to THBS1 promoter (Amodeo et al., 2013). et al., 2010; Roudier et al., 2013). Both TSP-1 and TSP-2 were downregulated in Akt−/− mice, and this CCAAT box binding CCAAT-binding factor (CBF) mediates the effect downregulation was responsible for the increased angiogenesis in these of histone deacetylase (HDAC) inhibitors in the regulation of angiogen- mice. The TSP-2 promoter inactivation by dominant negative Akt sug- esis and tumor growth: acetylated CBF specifically binds to TSP-1 gested that the regulation is transcriptional, but the exact mechanism promoter, and its activity is increased by the binding of acetylated H3 has not been addressed (Chen et al., 2005). Although the studies identi- (Kang et al., 2008). fying the signals that lead to the transcriptional upregulation are infor- Recently, the role of TSP-1 in inflammation has been actively studied mative in defining the stimuli regulating the expression of TSPs, without and confirmed in several models (McMorrow et al., 2013). Transcrip- addressing the molecular mechanism and delineating the promoter tional regulation of TSP-1 in chronic inflammation was addressed in in- elements involved in the regulation, it is impossible to know whether flammatory joint disease (McMorrow et al., 2013): the orphan receptor the observed effects are direct or mediated by an autocrine mediator 4A2 (NR4A2) was responsible for the downregulation of the promoter and whether the mechanism of the regulation in these examples is activity, and the anti-tumor necrosis factor treatment, which decreases truly transcriptional. NR4A2, increased TSP-1 expression and suggested that TSP-1 plays a role in resolution of inflammation. 3.1.2. TSP-2 Many studies reported the stimuli regulating transcription without The promoters of human and mouse TSP-2 genes were analyzed addressing specific transcriptional mechanisms and transcription and compared in (Adolph et al., 1997). Interestingly, tissue-specific factors involved. differences in transcription sites were found, resulting in transcripts For example, TGFβ1 and TGFβ2stimulatedTSP-1andTSP-2produc- of different lengths depending on the tissue. This was the only report tion in bovine adrenocortical cells, and the effect was blocked by the considering the features of THBS2 promoter. Putative transcription transcription inhibitors (Negoescu et al., 1995). factor binding sites were analyzed, and comparison to THBS1 promoter did not reveal any similarities between two genes in the promoter regions. 3.1.1.2. Negative regulation. TSP-1 gene promoter appears to have an in- hibitory element between −300 and −750 bp positions, as was report- 3.1.3. Other TSPs ed in two independent studies (Kang et al., 2004; Dabir et al., 2008). Dr. Bornstein's group reported on the promoter of THBS3 as well. Shorter promoter deletion constructs are significantly more active, as Interaction between THBS3 gene and adjacent metaxin gene was are the longer constructs, both constitutively and in response to stimuli. revealed (Collins et al., 1998), and the active SP1-binding elements The promoter was found to be sensitive to treatment with nickel were identified in the common regulatory region of the two genes (Salnikow et al., 1994): nickel downregulated the activity of the pro- (Collins and Bornstein, 1996). Analysis of the transcription initiation moter and decreased the expression of endogenous TSP-1 in cultured sites revealed alternative transcripts (Adolph and Bornstein, 1999). hamster embryo cells. Activating transcription factor-1 (ATF-1) was Since the last publication in 1999, there have been no additional reports identified as a component of the protein complex that bound to a on the regulation of THBS3 transcription. negative regulatory site in the mouse TSP-1 gene promoter (Salnikow There is very little information about the transcriptional regulation et al., 1997). ATF-1 binding to the region between positions −1210 of TSP-4 and TSP-5 genes, despite their reported importance in several and −1123 of the promoter harboring a cAMP-responsive element physiological and pathological processes (Hecht et al., 1995; Mustonen (CRE) induced the repression of the promoter in response to hepatocyte et al., 2008; Eroglu et al., 2009; Frolova et al., 2010; Cingolani et al., growth factor (HGF) and played a key role in tumor progression 2011; Frolova et al., 2012; Lynch et al., 2012; Risher and Eroglu, 2012). triggered by HGF (Ghoneim et al., 2007). The promoter of TSP-5 (COMP) has been analyzed, and a region required Further analyses of THBS1 promoter identified many active binding for the activity in chondrocytes was identified within 375 bp of the sites for transcription factors that tightly regulate TSP-1 expression at translational start site, as well as enhancer elements between −1.0 kb the transcriptional level. YY-1 binds to a site at the position −440 of and −1.7 kb (Posey et al., 2005). the promoter, which results in an interaction that can be weakened by the increased c-Jun levels, suggesting that the interaction of YY- and 3.2. Alternative splicing c-Jun decreases the promoter activity (Kang et al., 2004). The well-documented suppression of TSP-1 production in tumors With a large number of exons in TSP genes (N20 in most TSPs), the (upregulating angiogenesis to promote tumor growth) includes tran- possibility of alternative splicing and regulation of expression and func- scriptional downregulation: e.g., the product of Wilms' tumor suppres- tions by this mechanism is plausible. However, alternative splicing and sor gene, WT1, binds to the −210 region of the TSP-1 gene promoter its potential significance in regulation of TSPs have not been seriously and represses THBS1 transcription (Dejong et al., 1999), while tumor addressed to date. Several publications reported alternative forms of suppressor protein p53 activates TSP-1 promoter (Su et al., 2010). TSP-2 and TSP-3 (Adolph, 1999; Adolph and Bornstein, 1999). A produc- Another mechanism used by cancers to prevent TSP-1 promoter ac- tion of a Lumbar-disk-herniation-associated form of TSP-2 is due to a tivation is hypermethylation of the promoter that has been detected in polymorphism and the alternative splicing of TSP-2 that results in a variety of cancers [e.g., (Kanai et al., 2001; Yang et al., 2003; Liu et al., skipping exon 11 (Hirose et al., 2008). Lack of exon 11 causes decreased 2005; Lindner et al., 2013)]. TSP-1 promoter is also a target for a interaction of TSP-2 with metalloproteinases. Alternative splicing of

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 6 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx

TSP-4 gene associated with change in cell motility was reported in production of a protein may not always reflect the increased levels cancer cells (Lee et al., 2008). of mRNA [e.g., (Manni et al., 2007; Bhattacharyya et al., 2008; Multiple TSP forms of different sizes are routinely observed Sanghamitra Bhattacharyya et al., 2012)]. As more examples of such (e.g.,Stenina et al., 2003a, 2005), but the origin of these forms has discordant levels of protein and mRNA accumulate, we will be forced not been thoroughly explored, and the possibility of the alternative to address the translational regulation of TSPs. splicing has not been excluded. TSP-1 mRNA is associated with rich in adenines and uracils ARE binding protein HuR. HuR facilitates translation, and it has been co- 3.3. Regulation of mRNA stability precipitated with TSP-1 mRNA from the cultured MCF7 breast cancer cells (Caux et al., 1991). Another example of translational regulation of As has been mentioned above, both TSP-1 mRNA and protein have a TSP-1 production is translational silencing of TSP-1 mRNA in response short half-life. The short half-life of TSP-1 mRNA is due, at least in part, to high glucose (Bhattacharyya et al., 2008; Sanghamitra Bhattacharyya to AU-rich elements in the 3′ untranslated regions (3′UTR) of the TSP-1 et al., 2012) that is tissue-specific and regulated by a cell-specific mRNA (McGray et al., 2011a, 2011b). Unusually large regulatory 3′UTR induction of miR-467 and its binding to TSP-1 3′UTR. of TSP-1 and TSP-2 mRNAs suggests extensive post-transcriptional reg- ulation at the level of mRNA (Table 2). The rest of TSPs have an average 3.5. Post-translational regulation — secretion sized UTR; however, this does not exclude regulation of mRNA stability or translation. UTR of all TSPs has putative target binding sites for TSPs are secreted extracellular matrix proteins. Although some of miRNAs. There is no information on the regulation of TSP-3, TSP-4, them, e.g., TSP-1, TSP-4 and TSP-5, perform important functions inside and TSP-5 expression at the level of mRNA, but several reports de- the secretory pathways (Hecht et al., 1998, 2001; Hecht and Sage, scribed the regulation of TSP-1 and TSP-2 through the modulation of 2006; Lynch et al., 2012), most of the widely known functions rely on mRNA stability or translation. the presence of TSPs outside the cell. TSP-1 mRNA was destabilized by the activation of myc oncoprotein The regulation of secretion of pre-made protein is a very fast and a (Janz et al., 2000). Further analysis of this mechanism revealed that de- very efficient way to regulate the protein availability, and this type of stabilization is a result of increased expression of miR-17-92 family that regulation is used by the cells to control the extracellular TSP levels. binds TSP-1 mRNA (Dews et al., 2010). The decrease in the levels of the TSP-1 can be retained inside the cells, and the secretion depends on members of this family of miRNA, miR-18/19, was associated with the Ca2+ regulation: TSP-1 retention inside the cells can be induced increased TSP-1 level in cardiomyocytes of aging hearts, identifying by a calcium chelator and is regulated in renal cell carcinoma cells by the aging process (van Almen et al., 2011b). p53-responsive miR-194 TRPC4 calcium channel expression (Veliceasa et al., 2007). TSP-1 is a decreased the levels of mature TSP-1 mRNA despite the increased levels major component of platelet alpha granules: pre-synthesized protein of the primary transcript, prevented the production of TSP-1 protein, is released upon platelets activation (Lawler and Slayter, 1981; Dawes and promoted angiogenesis in colon cancers (Sundaram et al., 2011), et al., 1983). Macrophages also regulate TSP-1 secretion: the secretion presumably, due to destabilization of TSP-1 mRNA. is induced by LPS, and the threshold for the level of secretion is regulated Increased TSP-1 mRNA stability was detected in response to by the environmental signals, including the mediators of the adaptive heat shock (Kang et al., 2006), and a region of TSP-1 3′UTR respon- immune system, Th1 and Th2 cytokines (Fordham et al., 2012). TSP-1 sible for the increased stability was identified as 968–1258 from secretion depends on cell density in culture: lower density results in the stop codon. TGFβ also increased TSP-1 expression in cultured higher secretion of TSP-1 by fibroblasts, endothelial and smooth muscle osteosarcoma cells by stabilizing mRNA via p38 MAPK signaling cells without change in protein synthesis (Mumby et al., 1984). (Okamoto et al., 2002). Both TSP-4 and TSP-5 appear to have additional intracellular There are no mechanistic reports describing the effects on TSP-2 functions in the secretory pathways: TSP-5 can associate with its mRNA stability. Although there are examples of post-transcriptional co-secreted extracellular ligands and ER chaperones (Hecht et al., regulation of TSP-2 [e.g., in (Bein et al., 1998) describing the effect of 1998, 2001; Hecht and Sage, 2006), and TSP-4 bind a transcriptional c-myb on TSP-2 expression via a post-transcriptional mechanism], the factor Aft6α promoting its translocation to the nucleus (Lynch et al., details of this regulation remain unknown. 2012). The latter function is shared with TSP-1 and is thought to be a common feature of TSPs in protecting and augmenting ER function 3.4. Regulation of mRNA translation (Lynch et al., 2012).

Due to the complexity of translational mechanisms and lack of 4. Descriptive studies of TSP expression detailed knowledge of these mechanisms, as well as to the requirement of specific skills and experience for the experimental manipulations More than 2400 publications address the expression of TSPs in dif- with RNA, investigation of translational regulation of TSPs has not ferent tissues and various conditions. The findings clearly indicate the been the most popular of research topics. The large size of TSP mRNA importance of the altered expression in physiology and pathology and and, especially, the long regulatory UTR of TSP-1 and TSP-2 may have stress the need to develop understanding of the mechanisms that con- discouraged this type of study. However, studies of translational regula- trol the expression. Most of the information about the expression of tion of TSPs may prove very rewarding: there are multiple independent TSPs comes from descriptive studies, in which the mechanisms of regu- observations that increased levels of mRNA do not guarantee increased lation have not been addressed. This information is valuable in identify- production of the corresponding protein and that the increased ing physiological and pathological processes dependent on TSPs and in providing basis for future studies of precise mechanisms. Among all the descriptive studies, the most valuable are the ones where protein Table 2 levels have been assessed. Whenever only the mRNA levels were evalu- Size of untranslated regions of mRNA of TSPs. ated, there is always a possibility that they do not translate into protein 5′UTR 3′UTR production or that the produced protein is not secreted, as seems to be a

TSP-1 179 2130 frequent occurrence with TSPs. TSP-2 320 2061 Although an overview of conditions resulting in altered expression TSP-3 21 234 of TSPs is not the goal of this article, several major themes in the regula- TSP-4 191 156 tion of TSPs are very prominent and must be mentioned: growth and TSP-5 36 161 remodeling of tissues, altered angiogenesis and cancer, ischemia and

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx 7 reperfusion, activation of TGFβ,inflammation and immune response, (Scott-Burden et al., 1990). TSP-2 expression is downregulated by aging, and embryonic development (Table 3). Any of these pathological Cyp1B1, and this regulation is important for a proper capillary mor- or physiological situations consistently result in or are caused by the phogenesis in a model of retinopathy of prematurity (a neovascular change in the levels of one or several TSPs. As usual, TSP-1 expression response during oxygen-induced ischemic retinopathy)(Tang et al., has been better studied than the expression of the other four TSPs, 2009). The activity of endothelial nitric oxide synthase (eNOS) neg- although many recent reports repeatedly find the associations of atively correlated with TSP-2 levels: deficiency in eNOS resulted in in- TSP-4 expression and pathological changes. creased levels of TSP-2 and decreased angiogenesis in a mouse model, Altered expression of TSPs during the growth and remodeling of tis- and NO repressed the promoter of TSP-2 (MacLauchlan et al., 2011). sues is associated either with the need for angiogenesis, matrix remod- However, the precise transcriptional mechanisms were not dissected eling, or the direct stimulation of cell growth. TSP-4 is upregulated in in these studies. the mouse heart in response to pressure overload and in human The extent of angiogenesis after ischemia and reperfusion (IR) corre- hypertrophied hearts. It has a protective function regulating cardiomyo- lates with the successful repair process in tissues and with the patient cyte function and production of extracellular matrix (Gabrielsen et al., survival. Elevation of TSP-1 levels was detected after IR of the brain, 2007; Mustonen et al., 2008; Mustonen et al., 2010; Cingolani et al., myocardium, lung, and kidney (Lin et al., 2003; Frangogiannis et al., 2011; Melenovsky et al., 2011; Frolova et al., 2012; Lynch et al., 2012). 2005; Sezaki et al., 2005; Lario et al., 2007; Sage et al., 2008). While TSP-4 is expressed in atherosclerotic lesions and promotes atheroscle- the expression of TSP-2 was elevated at the peak of angiogenesis rotic process by attracting and retaining macrophages in the lesion (2 weeks after reperfusion), the expression of TSP-1 was dramatically (Frolova et al., 2010). Thbs4 is responsive to the ischemic injury in the elevated early after the event and did not seem to represent a response brain cortex and plays an important role in post-injury astrogenesis to the angiogenic process. Although it is unclear what the role of TSP-1 is (Benner et al., 2013). THBS4 expression increases with age in the brain in tissues just hours after IR, it appears to have a protective effect in the gray matter and may be associated with Alzheimer's disease and altered heart: e.g., myocardial infarction (MI) patients with higher levels of TSP- inflammatory response (Cagliani et al., 2013). Increased TSP-1 expres- 1 in the platelet-poor plasma had a better outcome and fewer adverse sion is also often found in tissue growth and remodeling, e.g., in embryo cardiac events within 14 months after MI (Kaiser et al., 2013). In animal fibroblasts in response to c-Jun stimulation (Mettouchi et al., 1994). In- models of MI, TSP-1 mRNA was induced only 1 h of ischemia and was creased levels of TSP-1 and TSP-2 have been reported in remodeling elevated 3–7 days after reperfusion. TSP-1 was localized to the infarc- large arteries in atherosclerotic lesions and after injury (Stenina et al., tion border zone and reduced inflammation and granulation tissue 2003a; Moura et al., 2008; Frolova et al., 2010; Pohjolainen et al., 2012). formation protecting non-infarcted myocardium from fibrosis (Sezaki Inflammation and immunity is an emerging field in TSP studies et al., 2005; Kaiser et al., 2013). Interestingly, in kidneys increased (Yabkowitz et al., 1993; Riessen et al., 1998; Vallejo et al., 2000; Zhao expression of TSP-1 early after IR (only 3–12 h, returning to the base et al., 2001; Lange-Asschenfeldt et al., 2002; Manna and Frazier, 2003; line by 48 h) mediated the injury, and Thbs1−/− mice showed signifi- Frangogiannis et al., 2005; Pluskota et al., 2005; Grimbert et al., 2006; cant protection from the damage after IR injury and from the renal fail- Lopez-Dee et al., 2011; Papageorgiou et al., 2012). Although functions ure (Thakar et al., 2005). Clearly, the function and the effect of the of TSPs in immune response and inflammation are not completely un- altered expression of TSPs depend on the injury mechanism and the derstood, these proteins are clearly important for the adequate immune specific cellular event characteristic for a tissue. response and the resolution of the inflammation. TSP-1 is a major activator of TGFβ1, and there is a reciprocal feed- Multiple studies documented changed expression of TSP-1 and TSP- back mechanism maintaining and amplifying the circuit: TGFβ was 2 associated with altered angiogenesis and cancer growth. Decreased identified as TSP-1 expression regulator increasing the production of TSP-1 and/or TSP-2 expression correlates with increased vascularity TSP-1 in multiple studies (Bein et al., 2004; Flugel-Koch et al., 2004). in non-small cell lung cancers (Oshika et al., 1998), colon cancer As a major activator of TGFβ1(Crawford et al., 1998), TSP-1 is often (Tokunaga et al., 1999; Kawakami et al., 2001), invasive cervical cancer expressed in tissues producing TGFβ1, e.g., in cancers with high TGFβ1 (Kodama et al., 2001; Wu et al., 2004), glioma cells (Harada et al., 2003), levels (Kawataki et al., 2000) and in embryonic development (Melnick in cytomegalovirus (CMV) infection (Cinatl et al., 1999), and many et al., 2000). TSP-1 gene mutation was identified in a family with the other situations where angiogenesis is increased. familial pulmonary arterial hypertension (PAH) (Maloney et al., 2012). Interestingly, the expression of group B thrombospodins (TSP-3, The ability of the mutant Asp362Asn TSP-1 to activate TGFβ1was TSP-4 and TSP-5) increases in cancers. Increased levels of TSP-3 were as- reduced to 1/2 of the activity of the wild-type TSP-1. TSP-1 contributes sociated with poor prognosis in osteosarcoma patients (Dalla-Torre to the development of PAH in more than one way: the levels of et al., 2006). TSP-4 expression was increased in invasive breast cancer TSP-1 are increased in PAH patients and in animal models of PAH, and was suggested to facilitate the invasion of tumor cells (Amy et al., and the interaction of TSP-1 with its receptor CD47 disrupts the 2013). It was differentially expressed in lobular versus ductal breast constitutive interaction between CD47 and Caveolin-1, causing tumors (Korkola et al., 2003). THBS4 was found to be a powerful marker increased eNOS-dependent superoxide production and oxidative of diffuse-type gastric adenocarcinomas: its expression in fibroblasts stress (Bauer et al., 2012). was stimulated by tumor cells (Forster et al., 2011). Based on the Levels of TSPs increase in multiple tissues with age (Swinnen et al., existing data, THBS4 expression is a selective marker for specificcancers, 2009; Frazier et al., 2011; van Almen et al., 2011b; Cai et al., 2012; although the regulation and the significance of the increased levels have Rogers et al., 2012; Starr et al., 2013). The significance of this increase not been explored. in TSP production is still unclear, although in some tissues it appears Many stimuli able to reduce TSP-1 levels have been identified: to be beneficial (Swinnen et al., 2009; Frazier et al., 2011; Frolova e.g., all-trans-retinoid acid in smooth muscle cells (Axel et al., 2001); et al., 2012; Rogers et al., 2012). In others, TSPs are associated with hepatocyte growth factor/scatter factor that mediates angiogenesis aging-related pathological changes (Agah et al., 2004; van Almen through positive VEGF and negative TSP-1 regulation (Zhang et al., et al., 2011b; Cai et al., 2012; Roberts et al., 2012). 2003); shear stress (Bongrazio et al., 2006), and hypoxia (Bienes- While the significance of the altered expression of TSP-1 and TSP-2 Martinez et al., 2012). Increase in TSP-1 levels is often detected in asso- is well understood in most situations, and specific consequences are ciation with decreased angiogenesis and ischemia, e.g., in chronic leg expected (e.g., altered angiogenesis, matrix remodeling, TGFβ activa- ischemia (Favier et al., 2005). Increased levels of TSP-1 mRNA were tion), we don't have a clear understanding of the significance of altered detected upon stimulation with dexamethasone (Flugel-Koch et al., expression of group B TSPs in the majority of cases. Studies of the signals 2004); progesterone (Mirkin and Archer, 2004); high glucose (Poczatek and molecular pathways regulating their expression would greatly in- et al., 2000; Stenina et al., 2003a), thrombin and angiotensin II crease our understanding of TSP associations with specific pathological

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 8 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx

Table 3 Summary of examples of pathological and physiological conditions associated with altered expression of TSPs (references to published reports). Green — upregulated expression, red — downregulated expression, blue — both the upregulated expression and downregulated expression have been reported.

TSP-1 TSP-2 TSP-3 TSP-4 TSP-5 (COMP)

Cingolani et al., (2011); Frolova et al., (2012); Schroen et al., (2004); Gabrielsen et al., (2007); Swinnen et al., (2009); Lynch et al., (2012); Heart hypertrophy Wang et al., (2003a) van Almen et al., (2011a) Melenovsky et al., (2011); Mustonen et al., (2008); Mustonen et al., (2010); Rysa et al., (2005)

Frolova et al., (2010); Moura et al., (2008); Frolova et al., (2012); Atherosclerosis Frolova et al., (2010) Frolova et al., (2010) Frolova et al., (2010) Reed et al., (1995); Pohjolainen et al., (2012) Riessen et al., (1998)

Sweetwyne and Murphy- Pohjolainen et al., (2012); Fibrosis Ullrich, (2012) Reinecke et al., (2013)

Chen et al., (1999); Miano et al., (1993); Raugi et al., (1990); Arterial injury Roth et al., (1998); Sajid et al., (2001); Stenina et al., (2003)

Favier et al., (2005); Frangogiannis et al., (2005); Kaiser et al., (2013); Ischemia and reperfusion Lario et al., (2007); Benner et al., (2013) Lin et al., (2003); Sage et al., (2008); Sezaki et al., (2005); Thakar et al., (2005)

Alzheimer’s disease Cagliani et al., (2013)

Frangogiannis et al., Lange-Asschenfeldt et al., Inflammation ad (2005); (2002); Frolova et al., (2010); immune response Grimbert et al., (2006); Papageorgiou et al., Mustonen et al., (2012); Lopez-Dee et al., (2011); (2012); Pluskota et al., (2005) Manna and Frazier, (2003); Vanhoutte et al., (2013)

Martin-Manso et al., (2012); Miller et al., (2013); Mumby et al., (1984); Negoescu et al., (1995); Riessen et al., (1998); Streit et al., (2000); Vallejo et al., (2000); Vanhoutte et al., (2013); abkowitz etY al., (1993); Zhao et al., (2001)

Harada et al., (2003); Amy et al., (2013); Kang et al., (2006); Hawighorst et al., (2001); Bredel et al., (2005); Kawakami et al., (2001); Kodama et al., (2001); Forster et al., (2011); Kawataki et al., (2000); Oshika et al., (1998); Korkola et al., (2003); Kodama et al., (2001); Dalla-Torre et al., (2006); Cancers Streit et al., (1999); Liang et al., (2005); Lindner et al., (1992); Ramaswamy et al., (2003) Sun et al., (2006); Ma et al., (2009); Liu et al., (2005); Tokunaga et al., (1999) Sorlie et al., (2001); Volpert et al., (2002); Sun et al., (2006); Wu et al., (2004); Turashvili et al., (2007) Yang et al., (2003)

CMV infection Cinatl et al., (1999)

Belmadani et al., (2007); Bhattacharyya et al., (2008); Dabir et al., (2008); Daniel et al., (2004); Poczatek et al., (2000); Raman et al., (2011); Diabetes and Raman et al., (2007); metabolic disorders Stenina, (2005); Stenina et al., (2003); Sweetwyne and Murphy- Ullrich, (2012); Wang et al., (2006); Wang et al., (2004); Wang et al., (2003b); Zhou et al., (2006) Chavez et al., (2012)

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx 9

conditions and would suggest the functions performed by TSPs in Urry et al., 1998). TSP-2 may be important in palate development: in physiological and pathological processes. the earlier stages of palatogenesis, it was found throughout the extracel- lular matrix of shelf mesenchyme (Melnick et al., 2000), but later it was 5. Expression of TSPs in developing embryo restricted to the EM and bone of the maxilla. TSP-1 was also abundantly expressed in developing head mesenchyme, including the palate. TSPs are highly expressed in tissues of developing embryos of Developmental stage-dependent co-expression of TSP-1 and TSP-2 β various species. In some cases, the significance of high expression has was proposed to regulate TGF 1 activity. TSP-4 expression pattern sug- been examined and understood. For example, in drosophila, single TSP gests its role in the regulation of neurite outgrowth and differentiation of the fly is expressed in tendon during the formation of myotendinous of cartilage, tendons, and bone (Arber and Caroni, 1995; Tucker et al., junctions (Subramanian et al., 2007; Gilsohn and Volk, 2010). Its ex- 1995). In mouse embryo, TSP-5 is expressed in mesenchyme at day pression is induced by the tendon-specific transcription factor Stripe. E10, and by E19 it is clearly expressed in osteogenic and chondrogenic TSP accumulates in the myotendinous junction, and flies deficient in tissues (Di Cesare et al., 2000; Fang et al., 2000; Kipnes et al., 2003). Its TSP fail to form functional somatic musculature. expression in both the embryonic and adult tissues indicates its impor- Pluripotent human embryonic stem cells expressed and secreted tance in skeletal and cartilage development (Chen et al., 2004; Lin et al., TSP-1, and it was the highest expressed protein detected in expression 2005; Kong et al., 2010; Roman-Blas et al., 2010). fi profiling of these cells (LaFramboise et al., 2010) and potentiating cell Most of the studies that documented the expression pro les of TSPs division in cardiomyocytes. This observation suggested that TSP-1 in embryonic development are descriptive [in addition to the studies might participate in cardiogenesis and cardiac repair by stem cells. mentioned above (Corless et al., 1992; Laherty et al., 1992)andothers]. Similarly to the adult tissues, TSP expression is tightly regulated in em- Most molecular mechanisms of the regulation of the expression in the fi bryos and is associated with specific stages of embryonic development. developing embryos have never been addressed, and the signi cance Increased TSP-1 expression at mid-gestation was responsible for em- of the tightly timed distinct expression of each TSP is unclear in most fi bryonic lethality with pronounced heart defects and vascular abnormal- cases. However, similar to the investigation into expression pro les fi ities (Fouladkou et al., 2010). The expression of TSP-1 in mouse embryo and regulation of expression in adults, these studies suggest speci c fi brain was also time dependent with a peak at days 11 and 12 and down- distinct functions for ve TSPs in development. regulation at the later stages (Iruela-Arispe et al., 1993). The expression patterns of TSP-1 suggested a role in early CNS development (Adams 6. Therapeutic regulation of expression and Tucker, 2000). Differential functions in developing embryo are sup- ported by the distinct profile of expression of TSP-1, TSP-2 and TSP-3: Traditionally, the regulation of expression was not the first choice in TSP-1 was observed transiently in the neural tube, head mesenchyme, seeking to target specific proteins for therapeutic purpose. Difficulties of and cardiac cushions and constitutively in the resident megakaryocytes delivery of the molecular regulators into the nucleus and lack of speci- of the liver and in circulating megakaryocytes. In contrast, high expres- ficity in the action of the expression activators and inhibitors prevented sion of TSP-2 was detected in the connective tissue: e.g., in pericardium, the practical use of expression regulation in most cases. In the case of pleura, perichondrium, periosteum, meninges, ligaments, reticular TSPs, the regulation of expression is not only poorly understood, but is dermis, and cartilage and bone precursors (Iruela-Arispe et al., 1993; also multi-staged and complex, in part due to large regulatory regions Tooney et al., 1998). TSP-2 was also expressed in blood vessels and of mRNA (TSP-1 and TSP-2) and numerous introns (all TSPs). skeletal myoblasts. mRNA of TSP-3 was restricted to brain, cartilage In an attempt to control the foreign body response and to prevent and lung. The overlapping expression of TSP-1 and TSP-2 was found the granuloma formation in response to implantation and to increase only in the kidney and the gut (Iruela-Arispe et al., 1993). The dynamic angiogenesis, plasmids for the expression of a sense and an antisense pattern of TSP expression in Xenopus and avian embryos also support cDNA for TSP-2 were delivered in a collagen solution, which was applied the distinct non-overlapping functions for TSPs (Tucker et al., 1997; to biomaterials implanted subcutaneously (Kyriakides et al., 2001).

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 10 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx

TSP-2 sense cDNA reversed the foreign body response in Thbs2−/− References mice, and TSP-2 anti-sense cDNA reversed the granuloma formation in wild-type mice, demonstrating that manipulations with the ex- Adams, J.C., 2001. Thrombospondins: multifunctional regulators of cell interactions. Annu. Rev. Cell Dev. Biol. 17, 25–51. pression of TSPs may be a valuable approach to regulate angiogenesis Adams, J.C., Lawler, J., 2004. The thrombospondins. Int. J. Biochem. Cell Biol. 36, 961–968. and matrix deposition. This approach was successfully used to alter Adams, J.C., Lawler, J., 2011. The thrombospondins. Cold Spring Harb. Perspect. Biol. 3, the wound healing response in wild-type and Thbs2−/− mice using a009712. Adams, J.C., Tucker, R.P., 2000. The thrombospondin type 1 repeat (TSR) superfamily: an in vivo murine excisional wound healing model with the im- diverse proteins with related roles in neuronal development. Dev. Dyn. 218, proved polymer facilitating the delivery and release of the plasmid 280–299. and increasing the efficiency of cell transfection in the healing Adolph, K.W., 1999. Detection of exon skipping and retained introns in transcription of the human thrombospondin 2 gene. Biochem. Biophys. Res. Commun. 259, 527–532. wound (Kyriakides et al., 2002). Adolph, K.W., Bornstein, P., 1999. The human thrombospondin 3 gene: analysis of The discovery and the rapid progression of our knowledge of miRNA transcription initiation and an alternatively spliced transcript. Mol. Cell Biol. Res. rekindled the interest in therapeutic regulation of expression. Synthetic Commun. 2, 47–52. miRNA and their sequence-specific antisense antagonists can be rela- Adolph, K.W., Liska, D.J., Bornstein, P., 1997. Analysis of the promoter and transcription start sites of the human thrombospondin 2 gene (THBS2). Gene 193, 5–11. tively easily designed, produced and modified to stabilize the oligonu- Agah, A., Kyriakides, T.R., Lawler, J., Bornstein, P., 2002. The lack of thrombospondin-1 cleotide and to facilitate its better delivery into cells (Krutzfeldt et al., (TSP1) dictates the course of wound healing in double-TSP1/TSP2-null mice. Am. – 2005; Davis et al., 2006; Krutzfeldt et al., 2007; Scherr et al., 2007; J. Pathol. 161, 831 839. Agah, A., Kyriakides, T.R., Letrondo, N., Bjorkblom, B., Bornstein, P., 2004. Thrombospondin Horwich and Zamore, 2008; Stenvang and Kauppinen, 2008; Stenvang 2 levels are increased in aged mice: consequences for cutaneous wound healing and et al., 2008a, 2008b; van Rooij et al., 2008; Wu and Belasco, 2008; angiogenesis. Matrix Biol. 22, 539–547. Wang and Olson, 2009). The synthetic oligonucleotides are stable, and Almog, N., Henke, V., Flores, L., Hlatky, L., Kung, A.L., Wright, R.D., Berger, R., Hutchinson, L., Naumov, G.N., Bender, E., Akslen, L.A., Achilles, E.G., Folkman, J., 2006. Prolonged they are retained in tissues for weeks. Despite a large number of predict- dormancy of human liposarcoma is associated with impaired tumor angiogenesis. ed targets for each miRNA that can be found in public databases, the FASEB J. 20, 947–949. predictions based solely on the sequence match not necessarily become Amodeo, V., Bazan, V., Fanale, D., Insalaco, L., Caruso, S., Cicero, G., Bronte, G., Rolfo, C., Santini, D., Russo, A., 2013. Effects of anti-miR-182 on TSP-1 expression in human experimentally proven targets. The regulatory mechanisms mediated colon cancer cells: there is a sense in antisense? Expert Opin. Ther. Targets 17, by miRNA now appear to be more complex than simple no-nonsense 1249–1261. binding of miRNA to the target and degradation of the sequence- Amy, E.M., Song, S., Kutasovic, J.R., Reid, L.E., Valle, J.M., Vargas, A.C., Smart, C.E., Simpson, P.T., 2013. Thrombospondin-4 expression is activated during the stromal response to matched mRNA. Thus, if a mechanism operating through miRNA is invasive breast cancer. Virchows Arch. 463, 535–545. dissected, targeting of this mechanism may be specificandefficient. Arber, S., Caroni, P., 1995. Thrombospondin-4, an extracellular matrix protein expressed We have demonstrated that the antagonist of miR-467 efficient- in the developing and adult nervous system promotes neurite outgrowth. J. Cell – ly prevents hyperglycemia-induced cancer angiogenesis and tumor Biol. 131, 1083 1094. Axel, D.I., Frigge, A., Dittmann, J., Runge, H., Spyridopoulos, I., Riessen, R., Viebahn, R., growth in mouse models by targeting miR-467 interaction with 3′UTR Karsch, K.R., 2001. All-trans retinoic acid regulates proliferation, migration, differen- of TSP-1, relieving the translational silencing of TSP-1, and increasing tiation, and extracellular matrix turnover of human arterial smooth muscle cells. – TSP-1 production (Sanghamitra Bhattacharyya et al., 2012). As the func- Cardiovasc. Res. 49, 851 862. Bauer, P.M., Bauer, E.M., Rogers, N.M., Yao, M., Feijoo-Cuaresma, M., Pilewski, J.M., tions of TSPs in different pathologies are better understood and the Champion, H.C., Zuckerbraun, B.S., Calzada, M.J., Isenberg, J.S., 2012. Activated CD47 miRNA regulation of TSPs is uncovered, targeting protein synthesis by promotes pulmonary arterial hypertension through targeting caveolin-1. Cardiovasc. miRNA may prove to be an efficient way to prevent or restore the pro- Res. 93, 682–693. Bein, K., Ware, J.A., Simons, M., 1998. Myb-dependent regulation of thrombospondin 2 duction of a protein. In the case of miR-467, the pathway is signal- and expression. Role of mRNA stability. J. Biol. Chem. 273, 21423–21429. tissue-specific(Bhattacharyya et al., 2008; Sanghamitra Bhattacharyya Bein, K., Odell-Fiddler, E.T., Drinane, M., 2004. Role of TGF-beta1 and JNK signaling in et al., 2012). For TSPs that are normally tightly regulated and expressed capillary tube patterning. Am. J. Physiol. Cell Physiol. 287, C1012–C1022. fi Belmadani, S., Bernal, J., Wei, C.C., Pallero, M.A., Dell'italia, L., Murphy-Ullrich, J.E., Berecek, in large amounts only in speci c and limited conditions, regulation with K.H., 2007. A thrombospondin-1 antagonist of transforming growth factor-beta miRNAmayprovetobeanefficient and harmless therapeutic approach. activation blocks cardiomyopathy in rats with diabetes and elevated angiotensin II. The fact that all of the five knockout mice did not demonstrate an overt Am. J. Pathol. 171, 777–789. fi Benner, E.J., Luciano, D., Jo, R., Abdi, K., Paez-Gonzalez, P., Sheng, H., Warner, D.S., Liu, C., phenotype without challenge suggests that general non-speci cdown- Eroglu, C., Kuo, C.T., 2013. Protective astrogenesis from the SVZ niche after injury is regulation of a single or several TSPs may be safe, at least for a short controlled by Notch modulator Thbs4. Nature 497, 369–373. period of time. Bentley, A.A., Adams, J.C., 2010. The evolution of thrombospondins and their ligand- binding activities. Mol. Biol. Evol. 27, 2187–2197. Bhattacharyya, S., Marinic, T.E., Krukovets, I., Hoppe, G., Stenina, O.I., 2008. Cell type- 7. Conclusion specific post-transcriptional regulation of production of the potent antiangiogenic and proatherogenic protein thrombospondin-1 by high glucose. J. Biol. Chem. 283, – Regulation of TSP expression is not a well-studied area, with the 5699 5707. Bienes-Martinez, R., Ordonez, A., Feijoo-Cuaresma, M., Corral-Escariz, M., Mateo, G., exception of TSP-1 that attracted more attention, mostly due to the Stenina, O., Jimenez, B., Calzada, M.J., 2012. Autocrine stimulation of clear-cell renal profound changes in its expression during tumor development and its carcinoma cell migration in hypoxia via HIF-independent suppression of potent effect on angiogenesis. The existing information suggests that thrombospondin-1.Sci.Rep.2,788. Bongrazio, M., Da Silva-Azevedo, L., Bergmann, E.C., Baum, O., Hinz, B., Pries, A.R., the expression of TSPs is regulated at multiple molecular levels from Zakrzewicz, A., 2006. Shear stress modulates the expression of thrombospondin-1 transcriptional regulation and splicing to regulation of protein secretion. and CD36 in endothelial cells in vitro and during shear stress-induced angiogenesis Furthermore, we learned that the regulation of expression is often not in vivo. Int. J. Immunopathol. Pharmacol. 19, 35–48. fi Bornstein, P., 1995. Diversity of function is inherent in matricellular proteins: an appraisal only cell- and tissue-speci c, but also multi-leveled: simultaneous of thrombospondin 1. J. Cell Biol. 130, 503–506. engagement of several levels of regulation results in discordant levels Bornstein, P., Agah, A., Kyriakides, T.R., 2004. The role of thrombospondins 1 and 2 in the of mRNA and protein. Although investigation of different levels of regulation of cell-matrix interactions, collagen fibril formation, and the response to injury. Int. J. Biochem. Cell Biol. 36, 1115–1125. regulation may appear tedious and complex, the detailed knowledge Bredel, M., Bredel, C., Juric, D., Harsh, G.R., Vogel, H., Recht, L.D., Sikic, B.I., 2005. Functional of complementary molecular mechanisms may eventually prove useful network analysis reveals extended gliomagenesis pathway maps and three novel in developing effective, fast and specific therapeutic intervention. MYC-interacting genes in human gliomas. Cancer Res. 65, 8679–8689. Cagliani, R., Guerini, F.R., Rubio-Acero, R., Baglio, F., Forni, D., Agliardi, C., Griffanti, L., Fumagalli, M., Pozzoli, U., Riva, S., Calabrese, E., Sikora, M., Casals, F., Comi, G.P., Acknowledgments Bresolin, N., Caceres, M., Clerici, M., Sironi, M., 2013. Long-standing balancing selec- tion in the THBS4 gene: influence on sex-specific brain expression and gray matter – The work was supported by R01 DK067532 and R01 HL117216. volumes in Alzheimer disease. Hum. Mutat. 34, 743 753. Cai, H., Yuan, Z., Fei, Q., Zhao, J., 2012. Investigation of thrombospondin-1 and The author would like to thank Nadia Hoppe and Maria Stenina for transforming growth factor-beta expression in the heart of aging mice. Exp. Ther. the help in editing the text. Med. 3, 433–436.

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx 11

Carlson, J.M., Chakravarty, A., DeZiel, C.E., Gross, R.H., 2007. SCOPE: a web server for practical Flugel-Koch, C., Ohlmann, A., Fuchshofer, R., Welge-Lussen, U., Tamm, E.R., 2004. de novo motif discovery. Nucleic Acids Res. 35, W259–W264. Thrombospondin-1 in the trabecular meshwork: localization in normal and Carron, J.A., Hiscott, P., Hagan, S., Sheridan, C.M., Magee, R., Gallagher, J.A., 2000. Cultured glaucomatous eyes, and induction by TGF-beta1 and dexamethasone in vitro. human retinal pigment epithelial cells differentially express thrombospondin-1, -2, Exp. Eye Res. 79, 649–663. -3, and -4. Int. J. Biochem. Cell Biol. 32, 1137–1142. Fordham, J.B., Hua, J., Morwood, S.R., Schewitz-Bowers, L.P., Copland, D.A., Dick, A.D., Caux, Y., Husson, R., Maquin, M., 1991. In vitro demonstration of polymerization retrac- Nicholson, L.B., 2012. Environmental conditioning in the control of macrophage tion of composites by capillary penetration phenomenon. Chir. Dent. Fr. 61, 51–55. thrombospondin-1 production. Sci. Rep. 2, 512. Chakravarty, A., Carlson, J.M., Khetani, R.S., Gross, R.H., 2007. A novel ensemble learning Forster, S., Gretschel, S., Jons, T., Yashiro, M., Kemmner, W., 2011. THBS4, a novel stromal method for de novo computational identification of DNA binding sites. BMC molecule of diffuse-type gastric adenocarcinomas, identified by transcriptome-wide Bioinforma. 8, 249. expression profiling. Mod. Pathol. 24, 1390–1403. Chatila, K., Ren, G., Xia, Y., Huebener, P., Bujak, M., Frangogiannis, N.G., 2007. The role of Fouladkou, F., Lu, C., Jiang, C., Zhou, L., She, Y., Walls, J.R., Kawabe, H., Brose, N., Henkelman, the thrombospondins in healing myocardial infarcts. Cardiovasc. Hematol. Agents R.M., Huang, A., Bruneau, B.G., Rotin, D., 2010. The ubiquitin ligase Nedd4-1 is required Med. Chem. 5, 21–27. for heart development and is a suppressor of thrombospondin-1. J. Biol. Chem. 285, Chavez, R.J., Haney, R.M., Cuadra, R.H., Ganguly, R., Adapala, R.K., Thodeti, C.K., Raman, P., 6770–6780. 2012. Upregulation of thrombospondin-1 expression by leptin in vascular smooth Framson, P., Bornstein, P., 1993. A serum response element and a binding site for NF-Y muscle cells via JAK2- and MAPK-dependent pathways. Am. J. Physiol. Cell Physiol. mediate the serum response of the human thrombospondin 1 gene. J. Biol. Chem. 303, C179–C191. 268, 4989–4996. Chen, A.L., Fang, C., Liu, C., Leslie, M.P., Chang, E., Di Cesare, P.E., 2004. Expression of bone Frangogiannis, N.G., 2012. Matricellular proteins in cardiac adaptation and disease. Physiol. morphogenetic proteins, receptors, and tissue inhibitors in human fetal, adult, and Rev. 92, 635– 688. osteoarthritic articular cartilage. J. Orthop. Res. 22, 1188–1192. Frangogiannis, N.G., Ren, G., Dewald, O., Zymek, P., Haudek, S., Koerting, A., Winkelmann, Chen, J., Somanath, P.R., Razorenova, O., Chen, W.S., Hay, N., Bornstein, P., Byzova, T.V., K., Michael, L.H., Lawler, J., Entman, M.L., 2005. Critical role of endogenous 2005. Akt1 regulates pathological angiogenesis, vascular maturation and permeability thrombospondin-1 in preventing expansion of healing myocardial infarcts. Circula- in vivo. Nat. Med. 11, 1188–1196. tion 111, 2935–2942. Cinatl Jr., J., Kotchetkov, R., Scholz, M., Cinatl, J., Vogel, J.U., Driever, P.H., Doerr, H.W., 1999. Frazier, E.P., Isenberg, J.S., Shiva, S., Zhao, L., Schlesinger, P., Dimitry, J., Abu-Asab, M.S., Human cytomegalovirus infection decreases expression of thrombospondin-1 inde- Tsokos, M., Roberts, D.D., Frazier, W.A., 2011. Age-dependent regulation of skeletal pendent of the tumor suppressor protein p53. Am. J. Pathol. 155, 285–292. muscle mitochondria by the thrombospondin-1 receptor CD47. Matrix Biol. 30, Cingolani, O.H., Kirk, J.A., Seo, K., Koitabashi, N., Lee, D.I., Ramirez-Correa, G., Bedja, D., 154–161. Barth, A.S., Moens, A.L., Kass, D.A., 2011. Thrombospondin-4 is required for stretch- Frolova E.G., Drazba, J., Krukovets, I., Kostenko, V., Blech, L., Harry, C., Vasanji, A, Drumm, mediated contractility augmentation in cardiac muscle. Circ. Res. 109, 1410–1414. C., Sul, P., Jenniskens, G.J., Plow, E.F., Stenina-Adognravi, O., Control of organization Collins, M., Bornstein, P., 1996. SP1-binding elements, within the common metaxin- and function of muscle and tendon by thrombospondin-4. http://dx.doi.org/10. thrombospondin 3 intergenic region, participate in the regulation of the metaxin 1016/j.matbio.2014.02.003. gene. Nucleic Acids Res. 24, 3661–3669. Frolova, E.G., Pluskota, E., Krukovets, I., Burke, T., Drumm, C., Smith, J.D., Blech, L., Febbraio, Collins, M., Rojnuckarin, P., Zhu, Y.H., Bornstein, P., 1998. A far upstream, cell type-specific M., Bornstein, P., Plow, E.F., Stenina, O.I., 2010. Thrombospondin-4 regulates vascular enhancer of the mouse thrombospondin 3 gene is located within intron 6 of the inflammation and atherogenesis. Circ. Res. 107, 1313–1325. adjacent metaxin gene. J. Biol. Chem. 273, 21816–21824. Frolova, E.G., Sopko, N., Blech, L., Popović, Z.B., Li, J., Vasanji, A., Drumm, C., Krukovets, I., Corless, C.L., Mendoza, A., Collins, T., Lawler, J., 1992. Colocalization of thrombospondin Jain, M.K., Penn, M.S., Plow, E.F., Stenina, O.I., 2012. Thrombospondin-4 regulates and syndecan during murine development. Dev. Dyn. 193, 346–358. fibrosis and remodeling of the myocardium in response to pressure overload. FASEB Crawford, S.E., Stellmach, V., Murphy-Ullrich, J.E., Ribeiro, S.M., Lawler, J., Hynes, R.O., J. 26, 2363–2373. Boivin, G.P., Bouck, N., 1998. Thrombospondin-1 is a major activator of TGF-beta1 Gabrielsen, A., Lawler, P.R., Yongzhong, W., Steinbruchel, D., Blagoja, D., Paulsson-Berne, in vivo. Cell 93, 1159–1170. G., Kastrup, J., Hansson, G.K., 2007. Gene expression signals involved in ischemic injury, Dabir, P., Marinic, T.E., Krukovets, I., Stenina, O.I., 2008. Aryl hydrocarbon receptor is acti- extracellular matrix composition and fibrosis defined by global mRNA profiling of the vated by glucose and regulates the thrombospondin-1 gene promoter in endothelial human left ventricular myocardium. J. Mol. Cell. Cardiol. 42, 870–883. cells. Circ. Res. 102, 1558–1565. Ghoneim, C., Soula-Rothhut, M., Blanchevoye, C., Martiny, L., Antonicelli, F., Rothhut, B., Dalla-Torre, C.A., Yoshimoto, M., Lee, C.H., Joshua, A.M., de Toledo, S.R., Petrilli, A.S., 2007. Activating transcription factor-1-mediated hepatocyte growth factor-induced Andrade, J.A., Chilton-MacNeill, S., Zielenska, M., Squire, J.A., 2006. Effects of THBS3, down-regulation of thrombospondin-1 expression leads to thyroid cancer cell invasion. SPARC and SPP1 expression on biological behavior and survival in patients with oste- J. Biol. Chem. 282, 15490–15497. osarcoma. BMC Cancer 6, 237. Gilsohn, E., Volk, T., 2010. Slowdown promotes muscle integrity by modulating integrin- Daniel, C., Wiede, J., Krutzsch, H.C., Ribeiro, S.M., Roberts, D.D., Murphy-Ullrich, J.E., Hugo, mediated adhesion at the myotendinous junction. Development 137, 785–794. C., 2004. Thrombospondin-1 is a major activator of TGF-beta in fibrotic renal disease Grimbert, P., Bouguermouh, S., Baba, N., Nakajima, T., Allakhverdi, Z., Braun, D., Saito, H., in the rat in vivo. Kidney Int. 65, 459–468. Rubio, M., Delespesse, G., Sarfati, M., 2006. Thrombospondin/CD47 interaction: a Davis, S., Lollo, B., Freier, S., Esau, C., 2006. Improved targeting of miRNA with antisense pathway to generate regulatory T cells from human CD4+ CD25-T cells in response oligonucleotides. Nucleic Acids Res. 34, 2294–2304. to inflammation. J. Immunol. 177, 3534–3541. Dawes, J., Clemetson, K.J., Gogstad, G.O., McGregor, J., Clezardin, P., Prowse, C.V., Pepper, Harada, H., Nakagawa, K., Saito, M., Kohno, S., Nagato, S., Furukawa, K., Kumon, Y., Hamada, D.S., 1983. A radioimmunoassay for thrombospondin, used in a comparative study K.,Ohnishi,T.,2003.Introduction of wild-type p53 enhances thrombospondin-1 of thrombospondin, beta-thromboglobulin and platelet factor 4 in healthy volun- expression in human glioma cells. Cancer Lett. 191, 109–119. teers. Thromb. Res. 29, 569–581. Hecht, J.T., Sage, E.H., 2006. Retention of the matricellular protein SPARC in the endoplasmic Dejong, V., Degeorges, A., Filleur, S., Ait-Si-Ali, S., Mettouchi, A., Bornstein, P., Binetruy, B., reticulum of chondrocytes from patients with pseudoachondroplasia. J. Histochem. Cabon, F., 1999. The Wilms' tumor gene product represses the transcription of Cytochem. 54, 269–274. thrombospondin 1 in response to overexpression of c-Jun. Oncogene 18, 3143–3151. Hecht, J.T., Nelson, L.D., Crowder, E., Wang, Y., Elder, F.F., Harrison, W.R., Francomano, C.A., Dews, M., Homayouni, A., Yu, D., Murphy, D., Sevignani, C., Wentzel, E., Furth, E.E., Lee, Prange, C.K., Lennon, G.G., Deere, M., et al., 1995. Mutations in exon 17B of cartilage W.M., Enders, G.H., Mendell, J.T., Thomas-Tikhonenko, A., 2006. Augmentation of oligomeric matrix protein (COMP) cause pseudoachondroplasia. Nat. Genet. 10, tumor angiogenesis by a Myc-activated microRNA cluster. Nat. Genet. 38, 1060–1065. 325–329. Dews,M.,Fox,J.L.,Hultine,S.,Sundaram,P.,Wang,W.,Liu,Y.Y.,Furth,E.,Enders,G.H., Hecht, J.T., Montufar-Solis, D., Decker, G., Lawler, J., Daniels, K., Duke, P.J., 1998. Retention El-Deiry, W., Schelter, J.M., Cleary, M.A., Thomas-Tikhonenko, A., 2010. The myc-miR-17 of cartilage oligomeric matrix protein (COMP) and cell death in redifferentiated 92 axis blunts TGF{beta} signaling and production of multiple TGF{beta}-dependent pseudoachondroplasia chondrocytes. Matrix Biol. 17, 625–633. antiangiogenic factors. Cancer Res. 70, 8233–8246. Hecht, J.T., Hayes, E., Snuggs, M., Decker, G., Montufar-Solis, D., Doege, K., Mwalle, F., Di Cesare, P.E., Fang, C., Leslie, M.P., Tulli, H., Perris, R., Carlson, C.S., 2000. Expression of Poole, R., Stevens, J., Duke, P.J., 2001. Calreticulin, PDI, Grp94 and BiP chaperone cartilage oligomeric matrix protein (COMP) by embryonic and adult osteoblasts. proteins are associated with retained COMP in pseudoachondroplasia chondrocytes. J. Orthop. Res. 18, 713–720. Matrix Biol. 20, 251–262. DiPietro, L.A., Nissen, N.N., Gamelli, R.L., Koch, A.E., Pyle, J.M., Polverini, P.J., 1996. Henkin, J., Volpert, O.V., 2011. Therapies using anti-angiogenic peptide mimetics of Thrombospondin 1 synthesis and function in wound repair. Am. J. Pathol. 148, thrombospondin-1. Expert Opin. Ther. Targets 15, 1369–1386. 1851–1860. Hirose, Y., Chiba, K., Karasugi, T., Nakajima, M., Kawaguchi, Y., Mikami, Y., Furuichi, T., Mio, El Btaouri, H., Morjani, H., Greffe, Y., Charpentier, E., Martiny, L., 2011. Role of JNK/ATF-2 F., Miyake, A., Miyamoto, T., Ozaki, K., Takahashi, A., Mizuta, H., Kubo, T., Kimura, T., pathway in inhibition of thrombospondin-1 (TSP-1) expression and apoptosis medi- Tanaka, T., Toyama, Y., Ikegawa, S., 2008. A functional polymorphism in THBS2 ated by doxorubicin and camptothecin in FTC-133 cells. Biochim. Biophys. Acta 1813, that affects alternative splicing and MMP binding is associated with lumbar-disc 695–703. herniation. Am. J. Hum. Genet. 82, 1122–1129. Eroglu, C., Allen, N.J., Susman, M.W., O'Rourke, N.A., Park, C.Y., Ozkan, E., Chakraborty, C., Horwich, M.D., Zamore, P.D., 2008. Design and delivery of antisense oligonucleotides to Mulinyawe, S.B., Annis, D.S., Huberman, A.D., Green, E.M., Lawler, J., Dolmetsch, R., block microRNA function in cultured Drosophila and human cells. Nat. Protoc. 3, Garcia, K.C., Smith, S.J., Luo, Z.D., Rosenthal, A., Mosher, D.F., Barres, B.A., 2009. 1537–1549. Gabapentin receptor alpha2delta-1 is a neuronal thrombospondin receptor responsible Huang, X. a, Miller, W., 1991. A time-efficient, linear-space local similarity algorithm. Adv. for excitatory CNS synaptogenesis. Cell 139, 380–392. Appl. Math. 12, 337–357. Fang, C., Carlson, C.S., Leslie, M.P., Tulli, H., Stolerman, E., Perris, R., Ni, L., Di Cesare, P.E., Iruela-Arispe, M.L., Liska, D.J., Sage, E.H., Bornstein, P., 1993. Differential expression of 2000. Molecular cloning, sequencing, and tissue and developmental expression of thrombospondin 1, 2, and 3 during murine development. Dev. Dyn. 197, 40–56. mouse cartilage oligomeric matrix protein (COMP). J. Orthop. Res. 18, 593–603. Janz, A., Sevignani, C., Kenyon, K., Ngo, C.V., Thomas-Tikhonenko, A., 2000. Activation of Favier, J., Germain, S., Emmerich, J., Corvol, P., Gasc, J.M., 2005. Critical overexpression of the myc oncoprotein leads to increased turnover of thrombospondin-1 mRNA. thrombospondin 1 in chronic leg ischaemia. J. Pathol. 207, 358–366. Nucleic Acids Res. 28, 2268–2275.

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 12 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx

Kaiser, R., Grotemeyer, K., Kalsch, T., Graber, S., Wilkens, H., Elmas, E., 2013. Decreased reveals molecularly and clinically distinct subtypes of glioblastoma multiforme. TSP-1 following percutaneous coronary intervention is associated with major adverse Proc. Natl. Acad. Sci. U. S. A. 102, 5814–5819. cardiac events in ST-elevation myocardial infarction. Clin. Hemorheol. Microcirc. 54, Lin, T.N., Kim, G.M., Chen, J.J., Cheung, W.M., He, Y.Y., Hsu, C.Y., 2003. Differential regulation of 59–73. thrombospondin-1 and thrombospondin-2 after focal cerebral ischemia/reperfusion. Kanai, Y., Ushijima, S., Kondo, Y., Nakanishi, Y., Hirohashi, S., 2001. DNA methyltransferase Stroke 34, 177–186. expression and DNA methylation of CPG islands and peri-centromeric satellite Lin, Y., Luo, E., Chen, X., Liu, L., Qiao, J., Yan, Z., Li, Z., Tang, W., Zheng, X., Tian, W., 2005. regions in human colorectal and stomach cancers. Int. J. Cancer 91, 205–212. Molecular and cellular characterization during chondrogenic differentiation of Kang, J.H., Chang, S.Y., Yeom, D.H., Kim, S.A., Um, S.J., Hong, K.J., 2004. Weakening of the adipose tissue-derived stromal cells in vitro and cartilage formation in vivo. J. Cell. repressive YY-1 site on the thrombospondin-1 promoter via c-Jun/YY-1 interaction. Mol. Med. 9, 929–939. Exp. Mol. Med. 36, 300–310. Lindner, D.J., Wu, Y., Haney, R., Jacobs, B.S., Fruehauf, J.P., Tuthill, R., Borden, E.C., 2013. Kang, J.H., Kim, S.A., Hong, K.J., 2006. Induction of TSP1 gene expression by heat shock is Thrombospondin-1 expression in melanoma is blocked by methylation and targeted mediated via an increase in mRNA stability. FEBS Lett. 580, 510–516. reversal by 5-Aza-deoxycytidine suppresses angiogenesis. Matrix Biol. 32, 123–132. Kang, J.H., Kim, M.J., Chang, S.Y., Sim, S.S., Kim, M.S., Jo, Y.H., 2008. CCAAT box is required Liu, Y., Pang, J.C., Dong, S., Mao, B., Poon, W.S., Ng, H.K., 2005. Aberrant CpG island hyper- for the induction of human thrombospondin-1 gene by trichostatin A. J. Cell. methylation profile is associated with atypical and anaplastic meningiomas. Hum. Biochem. 104, 1192–1203. Pathol. 36, 416–425. Kawakami, T., Tokunaga, T., Hatanaka, H., Tsuchida, T., Tomii, Y., Osada, H., Onoda, N., Lopez-Dee, Z., Pidcock, K., Gutierrez, L.S., 2011. Thrombospondin-1: multiple paths to Morino, F., Nagata, J., Kijima, H., Yamazaki, H., Abe, Y., Osamura, Y., Ueyama, Y., inflammation. Mediators Inflamm. 2011, 296069. Nakamura, M., 2001. Interleukin 10 expression is correlated with thrombospondin Lynch, J.M., Maillet, M., Vanhoutte, D., Schloemer, A., Sargent, M.A., Blair, N.S., Lynch, K.A., expression and decreased vascular involvement in colon cancer. Int. J. Oncol. 18, Okada, T., Aronow, B.J., Osinska, H., Prywes, R., Lorenz, J.N., Mori, K., Lawler, J., 487–491. Robbins, J., Molkentin, J.D., 2012. A thrombospondin-dependent pathway for a Kawataki, T., Naganuma, H., Sasaki, A., Yoshikawa, H., Tasaka, K., Nukui, H., 2000. Correla- protective ER stress response. Cell 149, 1257–1268. tion of thrombospondin-1 and transforming growth factor-beta expression with Ma, X.J., Dahiya, S., Richardson, E., Erlander, M., Sgroi, D.C., 2009. Gene expression profil- malignancy of glioma. Neuropathology 20, 161–169. ing of the tumor microenvironment during breast cancer progression. Breast Cancer Kim, S.A., Hong, K.J., 2000. Responsive site on the thrombospondin-1 promotor to down- Res. 11, R7. regulation by phorbol 12-myristate 13-acetate in porcine aortic endothelial cells. Exp. MacLauchlan, S., Yu, J., Parrish, M., Asoulin, T.A., Schleicher, M., Krady, M.M., Zeng, J., Mol. Med. 32, 135–140. Huang, P.L., Sessa, W.C., Kyriakides, T.R., 2011. Endothelial nitric oxide synthase con- Kim, S.A., Um, S.J., Kang, J.H., Hong, K.J., 2001. Expression of thrombospondin-1 in human trols the expression of the angiogenesis inhibitor thrombospondin 2. Proc. Natl. Acad. hepatocarcinoma cell lines and its regulation by transcription factor Jun/AP-1. Mol. Sci. U. S. A. 108, E1137–E1145. Cell. Biochem. 216, 21–29. Maloney, J.P., Stearman, R.S., Bull, T.M., Calabrese, D.W., Tripp-Addison, M.L., Wick, M.J., Kipnes, J., Carlberg, A.L., Loredo, G.A., Lawler, J., Tuan, R.S., Hall, D.J., 2003. Effect of cartilage Broeckel, U., Robbins, I.M., Wheeler, L.A., Cogan, J.D., Loyd, J.E., 2012. Loss-of-function oligomeric matrix protein on mesenchymal chondrogenesis in vitro. Osteoarthritis thrombospondin-1 mutations in familial pulmonary hypertension. Am. J. Physiol. Cartilage 11, 442–454. Lung Cell. Mol. Physiol. 302, L541–L554. Kodama, J., Hashimoto, I., Seki, N., Hongo, A., Yoshinouchi, M., Okuda, H., Kudo, T., 2001. Manna, P.P., Frazier, W.A., 2003. The mechanism of CD47-dependent killing of T Thrombospondin-1 and -2 messenger RNA expression in invasive cervical cancer: cells: heterotrimeric Gi-dependent inhibition of protein kinase A. J. Immunol. 170, correlation with angiogenesis and prognosis. Clin. Cancer Res. 7, 2826–2831. 3544–3553. Kong,L.,Tian,Q.,Guo,F.,Mucignat,M.T.,Perris,R.,Sercu,S.,Merregaert,J.,DiCesare, Manni, A., Rager, T., Kimball, S.R., Jefferson, L.S., Washington, S., Hu, X., Verderame, M.F., P.E., Liu, C.J., 2010. Interaction between cartilage oligomeric matrix protein and 2007. Effects of alpha-difluoromethylornithine on thrombospondin-1 production by extracellular matrix protein 1 mediates endochondral bone growth. Matrix Biol. human breast cancer cells. Int. J. Oncol. 31, 1187–1191. 29, 276–286. Martin-Manso, G., Navarathna, D.H., Galli, S., Soto-Pantoja, D.R., Kuznetsova, S.A., Tsokos, Korkola, J.E., DeVries, S., Fridlyand, J., Hwang, E.S., Estep, A.L., Chen, Y.Y., Chew, K.L., M., Roberts, D.D., 2012. Endogenous thrombospondin-1 regulates leukocyte recruit- Dairkee, S.H., Jensen, R.M., Waldman, F.M., 2003. Differentiation of lobular versus ment and activation and accelerates death from systemic candidiasis. PLoS One 7, ductal breast carcinomas by expression microarray analysis. Cancer Res. 63, e48775. 7167–7175. Mayer, J.E., Iatridis, J.C., Chan, D., Qureshi, S.A., Gottesman, O., Hecht, A.C., 2013. Genetic Krutzfeldt, J., Rajewsky, N., Braich, R., Rajeev, K.G., Tuschl, T., Manoharan, M., Stoffel, M., polymorphisms associated with intervertebral disc degeneration. Spine J. 13, 2005. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 438, 685–689. 299–317. Krutzfeldt, J., Kuwajima, S., Braich, R., Rajeev, K.G., Pena, J., Tuschl, T., Manoharan, M., McGray, A.J., Gingerich, T., Petrik, J.J., LaMarre, J., 2011a. Rapid insulin-like growth factor- Stoffel, M., 2007. Specificity, duplex degradation and subcellular localization of 1-induced changes in granulosa cell thrombospondin-1 expression in vitro. J. Reprod. antagomirs. Nucleic Acids Res. 35, 2885–2892. Dev. 57, 76–83. Kyriakides, T.R., Tam, J.W., Bornstein, P., 1999. Accelerated wound healing in mice with a McGray, A.J., Gingerich, T., Petrik, J.J., Lamarre, J., 2011b. Regulation of thrombospondin-1 disruption of the thrombospondin 2 gene. J. Invest. Dermatol. 113, 782–787. expression through AU-rich elements in the 3′UTR of the mRNA. Cell. Mol. Biol. Lett. Kyriakides, T.R., Hartzel, T., Huynh, G., Bornstein, P., 2001. Regulation of angiogenesis and 16, 55–68. matrix remodeling by localized, matrix-mediated antisense gene delivery. Mol. Ther. McLaughlin, J.N., Mazzoni, M.R., Cleator, J.H., Earls, L., Perdigoto, A.L., Brooks, J.D., 3, 842–849. Muldowney III, J.A., Vaughan, D.E., Hamm, H.E., 2005. Thrombin modulates the Kyriakides, T.R., Cheung, C.Y., Murthy, N., Bornstein, P., Stayton, P.S., Hoffman, A.S., 2002. expression of a set of genes including thrombospondin-1 in human microvascular pH-sensitive polymers that enhance intracellular drug delivery in vivo. J. Control. endothelial cells. J. Biol. Chem. 280, 22172–22180. Release 78, 295–303. McMorrow, J.P., Crean, D., Gogarty, M., Smyth, A., Connolly, M., Cummins, E., Veale, D., LaFramboise, W.A., Petrosko, P., Krill-Burger, J.M., Morris, D.R., McCoy, A.R., Scalise, D., Fearon, U., Tak, P.P., Fitzgerald, O., Murphy, E.P., 2013. Tumor necrosis factor inhibi- Malehorn, D.E., Guthrie, R.D., Becich, M.J., Dhir, R., 2010. Proteins secreted by tion modulates thrombospondin-1 expression in human inflammatory joint disease embryonic stem cells activate cardiomyocytes through ligand binding pathways. through altered NR4A2 activity. Am. J. Pathol. 183, 1243–1257. J. Proteomics 73, 992–1003. Melenovsky, V., Benes, J., Skaroupkova, P., Sedmera, D., Strnad, H., Kolar, M., Vlcek, C., Laherty, C.D., Gierman, T.M., Dixit, V.M., 1989. Characterization of the promoter region of Petrak, J., Benes Jr., J., Papousek, F., Oliyarnyk, O., Kazdova, L., Cervenka, L., 2011. the human thrombospondin gene. DNA sequences within the first intron increase Metabolic characterization of volume overload heart failure due to aorto-caval fistula transcription. J. Biol. Chem. 264, 11222–11227. in rats. Mol. Cell. Biochem. 354, 83–96. Laherty, C.D., O'Rourke, K., Wolf, F.W., Katz, R., Seldin, M.F., Dixit, V.M., 1992. Characteri- Melnick, M., Chen, H., Zhou, Y., Jaskoll, T., 2000. Thrombospondin-2 gene expression zation of mouse thrombospondin 2 sequence and expression during cell growth and protein localization during embryonic mouse palate development. Arch. Oral and development. J. Biol. Chem. 267, 3274–3281. Biol. 45, 19–25. Lange-Asschenfeldt, B., Weninger, W., Velasco, P., Kyriakides, T.R., von Andrian, U.H., Mettouchi, A., Cabon, F., Montreau, N., Vernier, P., Mercier, G., Blangy, D., Tricoire, H., Bornstein, P., Detmar, M., 2002. Increased and prolonged inflammation and angiogenesis Vigier, P., Binetruy, B., 1994. SPARC and thrombospondin genes are repressed by in delayed-type hypersensitivity reactions elicited in the skin of thrombospondin-2- the c-jun oncogene in rat embryo fibroblasts. EMBO J. 13, 5668–5678. deficient mice. Blood 99, 538–545. Miller, T.W., Kaur, S., Ivins-O'Keefe, K., Roberts, D.D., 2013. Thrombospondin-1 is a CD47- Lario, S., Bescos, M., Campos, B., Mur, C., Luque, P., Alvarez, R., Campistol, J.M., 2007. dependent endogenous inhibitor of hydrogen sulfide signaling in T cell activation. Thrombospondin-1 mRNA expression in experimental kidney transplantation with Matrix Biol. 32, 316–324. heart-beating and non-heart-beating donors. J. Nephrol. 20, 588–595. Mirkin, S., Archer, D.F., 2004. Effects of mifepristone on vascular endothelial growth factor Lawler, P.R., Lawler, J., 2012. Molecular basis for the regulation of angiogenesis by and thrombospondin-1 mRNA in Ishikawa cells: implication for the endometrial thrombospondin-1 and -2. Cold Spring Harb. Perspect. Med. 2, a006627. effects of mifepristone. Contraception 70, 327–333. Lawler, J.W., Slayter, H.S., 1981. The release of heparin binding peptides from platelet Moon, Y., Bottone Jr., F.G., McEntee, M.F., Eling, T.E., 2005. Suppression of tumor cell inva- thrombospondin by proteolytic action of thrombin, plasmin and trypsin. Thromb. sion by cyclooxygenase inhibitors is mediated by thrombospondin-1 via the early Res. 22, 267–279. growth response gene Egr-1. Mol. Cancer Ther. 4, 1551–1558. Lee, J.H., Horak, C.E., Khanna, C., Meng, Z., Yu, L.R., Veenstra, T.D., Steeg, P.S., 2008. Moura, R., Tjwa, M., Vandervoort, P., Van Kerckhoven, S., Holvoet, P., Hoylaerts, M.F., 2008. Alterations in Gemin5 expression contribute to alternative mRNA splicing patterns Thrombospondin-1 deficiency accelerates atherosclerotic plaque maturation in and tumor cell motility. Cancer Res. 68, 639–644. ApoE−/− mice. Circ. Res. 103, 1181–1189. Li, Y., Tong, X., Rumala, C., Clemons, K., Wang, S., 2011. Thrombospondin1 deficiency Mumby, S.M., Abbott-Brown, D., Raugi, G.J., Bornstein, P., 1984. Regulation of reduces obesity-associated inflammation and improves insulin sensitivity in a diet- thrombospondin secretion by cells in culture. J. Cell. Physiol. 120, 280–288. induced obese mouse model. PLoS One 6, e26656. Murphy-Ullrich, J.E., Mosher, D.F., 1987a. Interactions of thrombospondin with cells in Liang, Y., Diehn, M., Watson, N., Bollen, A.W., Aldape, K.D., Nicholas, M.K., Lamborn, K.R., culture: rapid degradation of both soluble and matrix thrombospondin. Semin. Berger, M.S., Botstein, D., Brown, P.O., Israel, M.A., 2005. Gene expression profiling Thromb. Hemost. 13, 343–351.

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx 13

Murphy-Ullrich, J.E., Mosher, D.F., 1987b. Interactions of thrombospondin with endothelial angiogenesis and cancer growth in response to hyperglycemia. J. Am. Heart cells: receptor-mediated binding and degradation. J. Cell Biol. 105, 1603–1611. Assoc. 1, e005967. Mustonen, E., Aro, J., Puhakka, J., Ilves, M., Soini, Y., Leskinen, H., Ruskoaho, H., Rysa, J., Scherr, M., Venturini, L., Battmer, K., Schaller-Schoenitz, M., Schaefer, D., Dallmann, I., 2008. Thrombospondin-4 expression is rapidly upregulated by cardiac overload. Ganser, A., Eder, M., 2007. Lentivirus-mediated antagomir expression for specific Biochem. Biophys. Res. Commun. 373, 186–191. inhibition of miRNA function. Nucleic Acids Res. 35, e149. Mustonen, E., Leskinen, H., Aro, J., Luodonpaa, M., Vuolteenaho, O., Ruskoaho, H., Rysa, J., Schroen, B., Heymans, S., Sharma, U., Blankesteijn, W.M., Pokharel, S., Cleutjens, J.P., 2010. Metoprolol treatment lowers thrombospondin-4 expression in rats with Porter, J.G., Evelo, C.T., Duisters, R., van Leeuwen, R.E., Janssen, B.J., Debets, J.J., Smits, myocardial infarction and left ventricular hypertrophy. Basic Clin. Pharmacol. Toxicol. J.F., Daemen, M.J., Crijns, H.J., Bornstein, P., Pinto, Y.M., 2004. Thrombospondin-2 is es- 107, 709–717. sential for myocardial matrix integrity: increased expression identifies failure-prone Mustonen, E., Ruskoaho, H., Rysa, J., 2012. Thrombospondin-4, tumour necrosis factor-like cardiac hypertrophy. Circ. Res. 95, 515–522. weak inducer of apoptosis (TWEAK) and its receptor Fn14: novel extracellular matrix Scott-Burden, T., Resink, T.J., Hahn, A.W., Buhler, F.R., 1990. Induction of thrombospondin modulating factors in cardiac remodelling. Ann. Med. 44, 793–804. expression in vascular smooth muscle cells by angiotensin II. J. Cardiovasc. Negoescu, A., Lafeuillade, B., Pellerin, S., Chambaz, E.M., Feige, J.J., 1995. Transforming Pharmacol. 16 (Suppl. 7), S17–S20. growth factors beta stimulate both thrombospondin-1 and CISP/thrombospondin-2 Sezaki, S., Hirohata, S., Iwabu, A., Nakamura, K., Toeda, K., Miyoshi, T., Yamawaki, H., synthesis by bovine adrenocortical cells. Exp. Cell Res. 217, 404–409. Demircan, K., Kusachi, S., Shiratori, Y., Ninomiya, Y., 2005. Thrombospondin-1 is induced Okamoto, M., Ono, M., Uchiumi, T., Ueno, H., Kohno, K., Sugimachi, K., Kuwano, M., 2002. in rat myocardial infarction and its induction is accelerated by ischemia/reperfusion. Up-regulation of thrombospondin-1 gene by epidermal growth factor and Exp. Biol. Med. (Maywood) 230, 621–630. transforming growth factor beta in human cancer cells—transcriptional activation Shingu,T.,Bornstein,P.,1994.Overlapping Egr-1 and Sp1 sites function in the regu- and messenger RNA stabilization. Biochim. Biophys. Acta 1574, 24–34. lation of transcription of the mouse thrombospondin 1 gene. J. Biol. Chem. 269, Oshika, Y., Masuda, K., Tokunaga, T., Hatanaka, H., Kamiya, T., Abe, Y., Ozeki, Y., Kijima, H., 32551–32557. Yamazaki, H., Tamaoki, N., Ueyama, Y., Nakamura, M., 1998. Thrombospondin 2 gene Sorlie, T., Perou, C.M., Tibshirani, R., Aas, T., Geisler, S., Johnsen, H., Hastie, T., Eisen, M.B., expression is correlated with decreased vascularity in non-small cell lung cancer. van de Rijn, M., Jeffrey, S.S., Thorsen, T., Quist, H., Matese, J.C., Brown, P.O., Botstein, Clin. Cancer Res. 4, 1785–1788. D., Lonning, P.E., Borresen-Dale, A.L., 2001. Gene expression patterns of breast carci- Papageorgiou, A.P., Swinnen, M.,Vanhoutte,D.,VandenDriessche,T.,Chuah,M.,Lindner,D., nomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. Verhesen, W., de Vries, B., D'Hooge, J., Lutgens, E., Westermann, D., Carmeliet, P., U. S. A. 98, 10869–10874. Heymans, S., 2012. Thrombospondin-2 prevents cardiac injury and dysfunction in viral Starr, M.E., Hu, Y., Stromberg, A.J., Carmical, J.R., Wood, T.G., Evers, B.M., Saito, H., 2013. myocarditis through the activation of regulatory T-cells. Cardiovasc. Res. 94, 115–124. Gene expression profile of mouse white adipose tissue during inflammatory stress: Pluskota, E., Stenina, O.I., Krukovets, I., Szpak, D., Topol, E.J., Plow, E.F., 2005. Mechanism age-dependent upregulation of major procoagulant factors. Aging Cell 12, 194–206. and effect of thrombospondin-4 polymorphisms on neutrophil function. Blood 106, Stenina, O.I., Krukovets, I., Wang, K., Zhou, Z., Forudi, F., Penn, M.S., Topol, E.J., Plow, E.F., 3970–3978. 2003a. Increased expression of thrombospondin-1 in vessel wall of diabetic Zucker Poczatek, M.H., Hugo, C., Darley-Usmar, V., Murphy-Ullrich, J.E., 2000. Glucose stimulation rat. Circulation 107, 3209–3215. of transforming growth factor-beta bioactivity in mesangial cells is mediated by Stenina, O.I., Desai, S.Y., Krukovets, I., Kight, K., Janigro, D., Topol, E.J., Plow, E.F., 2003b. thrombospondin-1. Am. J. Pathol. 157, 1353–1363. Thrombospondin-4 and its variants: expression and differential effects on endothelial Pohjolainen, V., Mustonen, E., Taskinen, P., Napankangas, J., Leskinen, H., Ohukainen, P., cells. Circulation 108, 1514–1519. Peltonen, T., Aro, J., Juvonen, T., Satta, J., Ruskoaho, H., Rysa, J., 2012. Increased Stenina, O.I., Ustinov, V., Krukovets, I., Marinic, T., Topol, E.J., Plow, E.F., 2005. Polymor- thrombospondin-2 in human fibrosclerotic and stenotic aortic valves. Atherosclerosis phisms A387P in thrombospondin-4 and N700S in thrombospondin-1 perturb calcium 220, 66–71. binding sites. FASEB J. 19, 1893–1895. Posey, K.L., Davies, S., Bales, E.S., Haynes, R., Sandell, L.J., Hecht, J.T., 2005. In vivo human Stenina-Adognravi, O., 2013. Thrombospondins: old players, new games. Curr. Opin. cartilage oligomeric matrix protein (COMP) promoter activity. Matrix Biol. 24, Lipidol. 24, 401–409. 539–549. Stenvang, J., Kauppinen, S., 2008. MicroRNAs as targets for antisense-based therapeutics. Raman, P., Krukovets, I., Marinic, T.E., Bornstein, P., Stenina, O.I., 2007. Glycosylation Expert. Opin. Biol. Ther. 8, 59–81. mediates up-regulation of a potent antiangiogenic and proatherogenic protein, Stenvang, J., Lindow, M., Kauppinen, S., 2008a. Targeting of microRNAs for therapeutics. thrombospondin-1, by glucose in vascular smooth muscle cells. J. Biol. Chem. 282, Biochem. Soc. Trans. 36, 1197–1200. 5704–5714. Stenvang, J., Silahtaroglu, A.N., Lindow, M., Elmen, J., Kauppinen, S., 2008b. The utility of Raman, P., Harry, C., Weber, M., Krukovets, I., Stenina, O.I., 2011. Anoveltranscriptional LNA in microRNA-based cancer diagnostics and therapeutics. Semin. Cancer Biol. mechanism of cell type-specific regulation of vascular gene expression by glucose. 18, 89–102. Arterioscler. Thromb. Vasc. Biol. 31, 634–642. Streit, M., Velasco, P., Riccardi, L., Spencer, L., Brown, L.F., Janes, L., Lange-Asschenfeldt, B., Ramaswamy, S., Ross, K.N., Lander, E.S., Golub, T.R., 2003. A molecular signature of metastasis Yano, K., Hawighorst, T., Iruela-Arispe, L., Detmar, M., 2000. Thrombospondin-1 sup- inprimarysolidtumors.Nat.Genet.33,49–54. presses wound healing and granulation tissue formation in the skin of transgenic Raugi, G.J., Olerud, J.E., Gown, A.M., 1987. Thrombospondin in early human wound tissue. mice. EMBO J. 19, 3272–3282. J. Invest. Dermatol. 89, 551– 554. Su, A.I., Cooke, M.P., Ching, K.A., Hakak, Y., Walker, J.R., Wiltshire, T., Orth, A.P., Vega, R.G., Reed, M.J., Puolakkainen, P., Lane, T.F., Dickerson, D., Bornstein, P., Sage, E.H., 1993. Sapinoso, L.M., Moqrich, A., Patapoutian, A., Hampton, G.M., Schultz, P.G., Hogenesch, Differential expression of SPARC and thrombospondin 1 in wound repair: immunolo- J.B., 2002. Large-scale analysis of the human and mouse transcriptomes. Proc. Natl. calization and in situ hybridization. J. Histochem. Cytochem. 41, 1467–1477. Acad. Sci. U. S. A. 99, 4465–4470. Riessen,R.,Kearney,M.,Lawler,J.,Isner,J.M.,1998.Immunolocalization of thrombospondin- Su, F., Pascal, L.E., Xiao, W., Wang, Z., 2010. Tumor suppressor U19/EAF2 regulates 1 in human atherosclerotic and restenoticarteries.Am.HeartJ.135,357–364. thrombospondin-1 expression via p53. Oncogene 29, 421–431. Risher, W.C., Eroglu, C., 2012. Thrombospondins as key regulators of synaptogenesis in Subramanian,A.,Wayburn,B.,Bunch,T.,Volk,T.,2007.Thrombospondin-mediated the central nervous system. Matrix Biol. 31, 170–177. adhesion is essential for the formation of the myotendinous junction in Drosophila. Roberts, D.D., Miller, T.W., Rogers, N.M., Yao, M., Isenberg, J.S., 2012. The matricellular pro- Development 134, 1269–1278. tein thrombospondin-1 globally regulates cardiovascular function and responses to Sun, L., Hui, A.M., Su, Q., Vortmeyer, A., Kotliarov, Y., Pastorino, S., Passaniti, A., Menon, J., stress via CD47. Matrix Biol. 31, 162–169. Walling, J., Bailey, R., Rosenblum, M., Mikkelsen, T., Fine, H.A., 2006. Neuronal and Rogers, N.M., Roberts, D.D., Isenberg, J.S., 2012. Age-associated induction of cell mem- glioma-derived stem cell factor induces angiogenesis within the brain. Cancer Cell brane CD47 limits basal and temperature-induced changes in cutaneous blood 9, 287–300. flow. Ann. Surg. 258, 184–191. Sundaram, P., Hultine, S., Smith, L.M., Dews, M., Fox, J.L., Biyashev, D., Schelter, J.M., Huang, Roman-Blas, J., Dion, A.S., Seghatoleslami, M.R., Giunta, K., Oca, P., Jimenez, S.A., Williams, Q., Cleary, M.A., Volpert, O.V., Thomas-Tikhonenko, A., 2011. p53-responsive miR-194 C.J., 2010. MED and PSACH COMP mutations affect chondrogenesis in chicken limb inhibits thrombospondin-1 and promotes angiogenesis in colon cancers. Cancer Res. bud micromass cultures. J. Cell. Physiol. 224, 817–826. 71, 7490–7501. Roudier, E., Milkiewicz, M., Birot, O., Slopack, D., Montelius, A., Gustafsson, T., Paik, J.H., Sweetwyne, M.T., Murphy-Ullrich, J.E., 2012. Thrombospondin1 in tissue repair and DePinho, R.A., Casale, G.P., Pipinos, I.I., Haas, T.L., 2013. Endothelial FoxO1 is an intrin- fibrosis: TGF-beta-dependent and independent mechanisms. Matrix Biol. 31, sic regulator of thrombospondin 1 expression that restrains angiogenesis in ischemic 178–186. muscle. Angiogenesis 16, 759–772. Swinnen, M., Vanhoutte, D., Van Almen, G.C., Hamdani, N., Schellings, M.W., D'Hooge, J., Rysa, J., Leskinen, H., Ilves, M., Ruskoaho, H., 2005. Distinct upregulation of extracellular Van der Velden, J., Weaver, M.S., Sage, E.H., Bornstein, P., Verheyen, F.K., matrix genes in transition from hypertrophy to hypertensive heart failure. Hyperten- VandenDriessche, T., Chuah, M.K., Westermann, D., Paulus, W.J., Van de Werf, sion 45, 927–933. F., Schroen, B., Carmeliet, P., Pinto, Y.M., Heymans, S., 2009. Absence of Sage, E.H., Bornstein, P., 1991. Extracellular proteins that modulate cell-matrix interac- thrombospondin-2 causes age-related dilated cardiomyopathy. Circulation tions. SPARC, tenascin, and thrombospondin. J. Biol. Chem. 266, 14831–14834. 120, 1585–1597. Sage, E., Mercier, O., Van den Eyden, F., de Perrot, M., Barlier-Mur, A.M., Dartevelle, P., Tang, Y., Scheef, E.A., Wang, S., Sorenson, C.M., Marcus, C.B., Jefcoate, C.R., Sheibani, N., Eddahibi, S., Herve, P., Fadel, E., 2008. Endothelial cell apoptosis in chronically 2009. CYP1B1 expression promotes the proangiogenic phenotype of endothelium obstructed and reperfused pulmonary artery. Respir. Res. 9, 19. through decreased intracellular oxidative stress and thrombospondin-2 expression. Salnikow, K., Cosentino, S., Klein, C., Costa, M., 1994. Loss of thrombospondin transcrip- Blood 113, 744–754. tional activity in nickel-transformed cells. Mol. Cell. Biol. 14, 851–858. Thakar, C.V., Zahedi, K., Revelo, M.P., Wang, Z., Burnham, C.E., Barone, S., Bevans, S., Salnikow, K., Wang, S., Costa, M., 1997. Induction of activating transcription factor 1 by Lentsch, A.B., Rabb, H., Soleimani, M., 2005. Identification of thrombospondin 1 nickel and its role as a negative regulator of thrombospondin I gene expression. (TSP-1) as a novel mediator of cell injury in kidney ischemia. J. Clin. Invest. 115, Cancer Res. 57, 5060–5066. 3451–3459. Sanghamitra Bhattacharyya, K.S., Krukovets, Irene, Nestor, Carla, Li, Jianbo, Stenina- Tokunaga, T., Nakamura, M., Oshika, Y., Abe, Y., Ozeki, Y., Fukushima, Y., Hatanaka, H., Adognravi, Olga, 2012. Novel tissue-specific mechanism of regulation of Sadahiro, S., Kijima, H., Tsuchida, T., Yamazaki, H., Tamaoki, N., Ueyama, Y., 1999.

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001 14 O. Stenina-Adognravi / Matrix Biology xxx (2014) xxx–xxx

Thrombospondin 2 expression is correlated with inhibition of angiogenesis and Volpert, O.V., Pili, R., Sikder, H.A., Nelius, T., Zaichuk, T., Morris, C., Shiflett, C.B., Devlin, metastasis of colon cancer. Br. J. Cancer 79, 354–359. M.K., Conant, K., Alani, R.M., 2002. Id1 regulates angiogenesis through transcriptional Tooney, P.A., Sakai, T., Sakai, K., Aeschlimann, D., Mosher, D.F., 1998. Restricted localiza- repression of thrombospondin-1. Cancer Cell 2, 473–483. tion of thrombospondin-2 protein during mouse embryogenesis: a comparison to Wang, S., Olson, E.N., 2009. AngiomiRs—key regulators of angiogenesis. Curr. Opin. Genet. thrombospondin-1. Matrix Biol. 17, 131–143. Dev. 19, 205–211. Tucker, R.P., Adams, J.C., Lawler, J., 1995. Thrombospondin-4 is expressed by early Wang, S., Wu, X., Lincoln, T.M., Murphy-Ullrich, J.E., 2003. Expression of constitutively active osteogenic tissues in the chick embryo. Dev. Dyn. 203, 477–490. cGMP-dependent protein kinase prevents glucose stimulation of thrombospondin 1 Tucker, R.P., Hagios, C., Chiquet-Ehrismann, R., Lawler, J., 1997. In situ localization of expression and TGF-beta activity. Diabetes 52, 2144– 2150. thrombospondin-1 and thrombospondin-3 transcripts in the avian embryo. Dev. Wang, S., Skorczewski, J., Feng, X., Mei, L., Murphy-Ullrich, J.E., 2004. Glucose up-regulates Dyn. 208, 326–337. thrombospondin 1 gene transcription and transforming growth factor-beta activity Tucker, R.P., Hess, J.F., Gong, Q., Garvey, K., Shibata, B., Adams, J.C., 2013. A through antagonism of cGMP-dependent protein kinase repression via upstream thrombospondin in the anthozoan Nematostella vectensis is associated with stimulatory factor 2. J. Biol. Chem. 279, 34311–34322. the nervous system and upregulated during regeneration. Biol. Open 2, Wang, J.Y., Zhang, X.M., Zhang, H.Y., 2006. The study of thrombospondin-I (TSP1) expres- 217–226. sion in the early stages of diabetic retinopathy induced by streptozotocin. Fen Zi Xi Turashvili, G., Bouchal, J., Baumforth, K., Wei, W., Dziechciarkova, M., Ehrmann, J., Klein, J., Bao Sheng Wu Xue Bao 39, 431–437. Fridman, E., Skarda, J., Srovnal, J., Hajduch, M., Murray, P., Kolar, Z., 2007. Novel Wu, L., Belasco, J.G., 2008. Let me count the ways: mechanisms of gene regulation by markers for differentiation of lobular and ductal invasive breast carcinomas by laser miRNAs and siRNAs. Mol. Cell 29, 1–7. microdissection and microarray analysis. BMC Cancer 7, 55. Wu, M.P., Tzeng, C.C., Wu, L.W., Huang, K.F., Chou, C.Y., 2004. Thrombospondin-1 acts as a Urry, L.A., Whittaker, C.A., Duquette, M., Lawler, J., DeSimone, D.W., 1998. fence to inhibit angiogenesis that occurs during cervical carcinogenesis. Cancer J. 10, Thrombospondins in early Xenopus embryos: dynamic patterns of expression 27–32. suggest diverse roles in nervous system, notochord, and muscle development. Dev. Yabkowitz, R., Dixit, V.M., Guo, N., Roberts, D.D., Shimizu, Y., 1993. Activated T-cell Dyn. 211, 390–407. adhesion to thrombospondin is mediated by the alpha 4 beta 1 (VLA-4) and alpha Vallejo, A.N., Mugge, L.O., Klimiuk, P.A., Weyand, C.M., Goronzy, J.J., 2000. Central role of 5 beta 1 (VLA-5) integrins. J. Immunol. 151, 149–158. thrombospondin-1 in the activation and clonal expansion of inflammatory T cells. Yang, Q.W., Liu, S., Tian, Y., Salwen, H.R., Chlenski, A., Weinstein, J., Cohn, S.L., 2003. J. Immunol. 164, 2947–2954. Methylation-associated silencing of the thrombospondin-1 gene in human neuro- van Almen, G.C., Swinnen, M., Carai, P., Verhesen, W., Cleutjens, J.P., D'Hooge, J., Verheyen, blastoma. Cancer Res. 63, 6299–6310. F.K., Pinto, Y.M., Schroen, B., Carmeliet, P., Heymans, S., 2011a. Absence of Yu, K., Ganesan, K., Tan, L.K., Laban, M., Wu, J., Zhao, X.D., Li, H., Leung, C.H., Zhu, Y., Wei, thrombospondin-2 increases cardiomyocyte damage and matrix disruption in C.L., Hooi, S.C., Miller, L., Tan, P., 2008. A precisely regulated gene expression cassette doxorubicin-induced cardiomyopathy. J. Mol. Cell. Cardiol. 51, 318–328. potently modulates metastasis and survival in multiple solid cancers. PLoS Genet. 4, van Almen, G.C., Verhesen, W., van Leeuwen, R.E., van de Vrie, M., Eurlings, C., Schellings, e1000129. M.W., Swinnen, M., Cleutjens, J.P., van Zandvoort, M.A., Heymans, S., Schroen, B., Zaslavsky, A., Chen, C., Grillo, J., Baek, K.H., Holmgren, L., Yoon, S.S., Folkman, J., Ryeom, S., 2011b. MicroRNA-18 and microRNA-19 regulate CTGF and TSP-1 expression in age- 2010. Regional control of tumor growth. Mol. Cancer Res. 8, 1198–1206. related heart failure. Aging Cell 10, 769–779. Zhang, Y.W., Su, Y., Volpert, O.V., Vande Woude, G.F., 2003. Hepatocyte growth factor/ van Rooij, E., Marshall, W.S., Olson, E.N., 2008. Toward microRNA-based therapeutics for scatter factor mediates angiogenesis through positive VEGF and negative heart disease: the sense in antisense. Circ. Res. 103, 919–928. thrombospondin 1 regulation. Proc. Natl. Acad. Sci. U. S. A. 100, 12718–12723. Vanhoutte, D., van Almen, G.C., Van Aelst, L.N., Van Cleemput, J., Droogne, W., Jin, Y., Van Zhao, X.M., Hu, Y., Miller, G.G., Mitchell, R.N., Libby, P., 2001. Association of de Werf, F., Carmeliet, P., Vanhaecke, J., Papageorgiou, A.P., Heymans, S., 2013. thrombospondin-1 and cardiac allograft vasculopathy in human cardiac allografts. Cir- Matricellular proteins and matrix metalloproteinases mark the inflammatory and culation 103, 525–531. fibrotic response in human cardiac allograft rejection. Eur. Heart J. 34, 1930–1941. Zhou, Y., Poczatek, M.H., Berecek, K.H., Murphy-Ullrich, J.E., 2006. Thrombospondin 1 Veliceasa, D., Ivanovic, M., Hoepfner, F.T., Thumbikat, P., Volpert, O.V., Smith, N.D., 2007. mediates angiotensin II induction of TGF-beta activation by cardiac and renal cells Transient potential receptor channel 4 controls thrombospondin-1 secretion and under both high and low glucose conditions. Biochem. Biophys. Res. Commun. 339, angiogenesis in renal cell carcinoma. FEBS J. 274, 6365–6377. 633–641.

Please cite this article as: Stenina-Adognravi, O., Invoking the power of thrombospondins: Regulation of thrombospondins expression, Matrix Biol. (2014), http://dx.doi.org/10.1016/j.matbio.2014.02.001