Abstract

BACKGROUND

Uterine leiomyoma (also known as fibroid or myoma) is the most common benign tumor of the uterus found in women of reproductive age. It is not usually fatal but can produce serious clinical symptoms, including excessive uterine bleeding, pelvic pain or pressure, infertility and pregnancy complications. Due to lack of effective medical treatments surgery has been a definitive choice for the management of this tumor.

OBJECTIVE AND RATIONALE

Extracellular matrix (ECM) accumulation and remodeling are thought to be crucial for fibrotic diseases such as uterine leiomyoma. Indeed, ECM plays important role in forming the bulk structure of leiomyoma, and the ECM-rich rigid structure within these tumors is thought to be a cause of abnormal bleeding and pelvic pain. Therefore, a better understanding of ECM accumulation and remodeling is critical for developing new therapeutics for uterine leiomyoma.

SEARCH METHODS

PubMed and Google Scholar were searched for all original and review articles/book chapters related to ECM and medical treatments of uterine leiomyoma published in English until May 2017.

OUTCOMES

This review discusses the involvement of ECM in leiomyoma pathogenesis as well as current and future medical treatments that target ECM directly or indirectly. Uterine leiomyoma is characterized by elevated levels of collagens, fibronectin, laminins and proteoglycans. They can induce the mechanotransduction process, such as activation of the integrin-Rho/p38 MAPK/ERK pathway, resulting in cellular responses that are involved in pathogenesis and altered bidirectional signaling between leiomyoma cells and the ECM. ECM accumulation is affected by growth factors (TGF-β, activin-A and PDGF), cytokines (TNF-α), steroid hormones (estrogen and progesterone) and microRNAs (miR-29 family, miR-200c and miR-93/106b). Among these, TGF-βs (1 and 3) and activin-A have been suggested as key players in the accumulation of excessive ECM (fibrosis) in leiomyoma. The presence of elevated levels of ECM and myofibroblasts in leiomyoma supports the fibrotic character of these tumors. Interestingly, ECM may serve as a reservoir of profibrotic growth factors and enhance their activity by increasing their stability and extending their duration of signaling. At present, several classes of compounds, including gonadotropin-releasing hormone (GnRH) agonist (leuprolide acetate), GnRH antagonist (cetrorelix acetate), selective progesterone receptor modulators (ulipristate acetate and asoprisnil), antiprogestin (mifepristone) and natural compounds like vitamin D and resveratrol have been studied as medical treatments that target ECM in uterine leiomyoma.

WIDER IMPLICATIONS

Although several types of drugs (mostly antiproliferative agents) are available for leiomyoma treatment, none of them were introduced specifically as antifibrotic agents. In light of its critical role in the process of fibrosis in leiomyoma, we propose that ECM should be considered as a crucial target for future therapeutics. Thus, the introduction of drugs that are specifically antifibrotic could be a good solution to control abnormal leiomyoma growth and associated clinical symptoms. The antifibrotic compounds can be introduced based on their ability to regulate ECM components and their receptors, as well as growth factors, cytokines, steroid hormones and their corresponding receptors and intracellular signaling pathways, as well as microRNAs, involved in ECM production in leiomyoma.

Introduction

Uterine leiomyoma (also known as fibroid or myoma), the most common benign tumor of the uterus, is found in women of reproductive age (Islam et al., 2013a). They may occur in more than 70% of women, and ~25% of women show clinically significant lesions (Buttram and Reiter, 1981; Cramer and Patel, 1990). Particularly, African-American women are reported to have a higher leiomyoma incidence (Marshall et al., 1997; Day Baird et al., 2003), with more severe clinical symptoms (Kjerulff et al., 1996) compared to Caucasians. Symptoms associated with leiomyoma include excessive uterine bleeding, pelvic pain or pressure, infertility and pregnancy complications (Buttram and Reiter, 1981). Due to lack of effective, non-invasive medical treatments (Islam et al., 2013b), surgery has been the primary choice for leiomyoma management. However, surgery is risky and expensive and negatively affects the quality of life of patients. In the USA, the annual economic burden of uterine leiomyoma is estimated to be between $5.9 and $34.4 billion (Cardozo et al., 2012).

The precise molecular and cellular changes that led to the development and growth of uterine leiomyoma are not well understood. However, one distinguishing characteristic of uterine leiomyoma is the excessive accumulation of extracellular matrix (ECM) components including collagens (Fig. 1), fibronectin, laminins and proteoglycans (Stewart et al., 1994; Arici and Sozen, 2000; Norian et al., 2009; Malik et al., 2012; Herndon et al., 2016). High levels of ECM proteins in uterine leiomyoma result in mechanotransduction, a process whereby increased tissue stiffness leads to bidirectional signaling via integrins and downstream mediators including Rho/p38 MAPK/ERK (Rogers et al., 2008; Malik et al., 2012; Chen et al., 2013; Thorne et al., 2015) (Fig. 2). Uterine leiomyoma also expresses proteolytic enzymes such as matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs) that play key roles in ECM remodeling (Bogusiewicz et al., 2007; Malik et al., 2010). Furthermore, a number of studies indicate that ECM accumulation and function is regulated by growth factors (Joseph et al., 2010; Islam et al., 2014a), cytokines (Wang et al., 2015) and steroid hormones (Qiang et al., 2014). ECM binds and sequesters growth factors to promote their stability as well as restrict their activity. By degrading ECM components, MMPs and other proteolytic enzymes release growth factors and trigger activation of multiple signal transduction pathways (Fig. 3).

Hypothetical presentation of fibrosis in uterine leiomyoma. Fibrosis is initiated by inflammation in the presence or absence of genetic changes. Growth factors, cytokines and steroid hormones are produced at the site of injury and contribute to fibroblast activation and differentiation into myofibroblasts. Myofibroblasts produce ECM components to restore homeostasis and then should be eliminated by apoptosis. During chronic inflammation, myofibroblasts become resistant to elimination by apoptosis and produce excessive amounts of ECM components, leading to fibrotic transformation. Once established, fibroids can promote their own growth. Masson's trichrome stain highlighting collagen (green) and smooth muscle (red) in myometrium and leiomyoma was performed in our laboratory.
Figure 1

Hypothetical presentation of fibrosis in uterine leiomyoma. Fibrosis is initiated by inflammation in the presence or absence of genetic changes. Growth factors, cytokines and steroid hormones are produced at the site of injury and contribute to fibroblast activation and differentiation into myofibroblasts. Myofibroblasts produce ECM components to restore homeostasis and then should be eliminated by apoptosis. During chronic inflammation, myofibroblasts become resistant to elimination by apoptosis and produce excessive amounts of ECM components, leading to fibrotic transformation. Once established, fibroids can promote their own growth. Masson's trichrome stain highlighting collagen (green) and smooth muscle (red) in myometrium and leiomyoma was performed in our laboratory.

Mechanotransduction is caused by extracellular matrix in uterine leiomyoma cells. Mechanical force from the stiffness of ECM or osmotic stress activates mechanical signaling pathways through heterodimeric integrins (α and β) that function as transmembrane receptors for ECM components. Activation of FAK initiates actin polymerization and AKAP13 activation of RhoA, which in turn interacts with ROCK and activates the ERK/p38 MAPK-signaling cascade, resulting in changes in cell proliferation, decreased apoptosis and upregulation of genes involved in ECM composition and remodeling. Cells sense the mechanical force from newly generated altered ECM and further activate mechanical signaling that affects cell behavior and alters overall remodeling of the matrix. This process initiates cell-matrix interactions in a rapid, transient, and bidirectional manner, which is referred to as dynamic reciprocity. Compounds that can interrupt this signaling pathway are also listed (yellow square). FAK, focal adhesion kinase; AKAP13, A kinase anchor protein 13; RhoA, Ras homolog gene family, member A; ROCK, Rho kinase; ERK, extracellular signal–regulated kinase; MAPK, mitogen-activated protein kinase.
Figure 2

Mechanotransduction is caused by extracellular matrix in uterine leiomyoma cells. Mechanical force from the stiffness of ECM or osmotic stress activates mechanical signaling pathways through heterodimeric integrins (α and β) that function as transmembrane receptors for ECM components. Activation of FAK initiates actin polymerization and AKAP13 activation of RhoA, which in turn interacts with ROCK and activates the ERK/p38 MAPK-signaling cascade, resulting in changes in cell proliferation, decreased apoptosis and upregulation of genes involved in ECM composition and remodeling. Cells sense the mechanical force from newly generated altered ECM and further activate mechanical signaling that affects cell behavior and alters overall remodeling of the matrix. This process initiates cell-matrix interactions in a rapid, transient, and bidirectional manner, which is referred to as dynamic reciprocity. Compounds that can interrupt this signaling pathway are also listed (yellow square). FAK, focal adhesion kinase; AKAP13, A kinase anchor protein 13; RhoA, Ras homolog gene family, member A; ROCK, Rho kinase; ERK, extracellular signal–regulated kinase; MAPK, mitogen-activated protein kinase.

Regulation of extracellular matrix by growth factors, cytokines and steroid hormones in uterine leiomyoma. Extracellular matrix may act as both a reservoir of growth factors and a modulator of their actions. Compounds that can interrupt the signaling pathways are also listed (yellow squares). C, Curcumin; V, Vitamin D3; M, 2-Methoxyestradiol; L, Leuprolide acetate.
Figure 3

Regulation of extracellular matrix by growth factors, cytokines and steroid hormones in uterine leiomyoma. Extracellular matrix may act as both a reservoir of growth factors and a modulator of their actions. Compounds that can interrupt the signaling pathways are also listed (yellow squares). C, Curcumin; V, Vitamin D3; M, 2-Methoxyestradiol; L, Leuprolide acetate.

Uterine leiomyoma is thought to be a consequence of an improper inflammatory response (Wegienka, 2012; Leppert et al., 2013) with myofibroblasts possibly playing a key role in the development of fibrosis (Feng et al., 2016; Protic et al., 2016) (Fig. 1). In our recent study, we found α-smooth muscle actin (α-SMA) positive and desmin negative cells as well as a large amount of collagen in leiomyoma tissue, indicating the presence of myofibroblasts and their role in the ECM deposition (Protic et al., 2016). It is known that when women reach reproductive age, events such as ovulation, menstruation and implantation may create physiological injuries in the uterus. In addition to reproductive events, harmful stimuli, mechanical forces, hypoxia and oxidative stress may create a chronic inflammatory state in the uterus (Wegienka, 2012; Fletcher et al., 2013; Leppert et al., 2013; Santulli et al., 2013). In the inflammatory state associated with injury, myofibroblasts produce ECM to promote subsequent repair processes and tissue homeostasis (Wynn, 2007). However, during chronic inflammation, myofibroblasts continuously and excessively produce ECM resulting in pathological fibrosis (Fig. 1). It is hypothesized that growth factors including TGF-βs and activin-A are key players in driving myofibroblast differentiation during the process of fibrosis (Joseph et al., 2010; Islam et al., 2014a; Feng et al., 2016; Protic et al., 2016).

ECM accumulation is a critical event in producing the rigid structure of leiomyoma, and ECM stiffness is thought to be a cause of abnormal bleeding and pelvic pain or pressure. Therefore, the inhibition of further accumulation of ECM and associated fibrosis might be an option for the management of these tumors. In this review, we highlight the involvement of ECM components in leiomyoma pathogenesis. Particularly, we discuss the basis for the increase in abundance and regulation of ECM in leiomyoma as well as the role of the ECM as reservoir for growth factors and a modulator of their actions. We also present options for medical treatments that target ECM formation, which are currently used or may be attractive in the future.

Methods

We used multiple strategies to identify primary research publications and review articles/book chapters related to extracellular matrix and medical treatments of uterine leiomyoma published in English until May 2017. We conducted extensive searches in PubMed and/or Google Scholar using the following keywords either alone or in combination with ‘uterine leiomyoma and extracellular matrix’: sarcolemma, caveolae, myofilaments and intermediate filament, collagen, fibronectin, laminin, versican, proteoglycan, MMP, TIMP, mechanotransduction, mechanical stress, growth factor, transforming growth factor-β (TGF-β), activin-A, platelet-derived growth factor (PDGF), cytokines, chemokines, tumor necrosis factor-α, interleukin (IL)-1β, IL-11, IL-13, granulocyte-macrophage colony-stimulating factor (GM-CSF), estrogen, progesterone, microRNA, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), inflammation, myofibroblast, fibrosis, medical treatment, pharmacological treatment, leuprolide acetate, cetrorelix acetate, asoprisnil, ulipristal acetate, mifepristone, raloxifene, CP8947, 2-methoxyestradiol, liarozole, all-trans retinoic acid, vitamin D, celecoxib, tranilast, pirfenidone, halofuginone, curcumin, resveratrol and collagenase Clostridium histolyticum. We also searched additional relevant articles in the bibliographies of downloaded articles. Overall, we reviewed most relevant articles and included them as appropriate.

Ultrastructural features of uterine leiomyoma

A number of studies have identified some ultrastructural features that may distinguish uterine leiomyoma from myometrium. These include sarcolemma (Richards et al., 1998), caveolae (Richards et al., 1998), extracellular matrix (Leppert et al., 2004; Wortham et al., 2006), myofilaments and intermediate filaments (Eyden et al., 1992; Wortham et al., 2006). Sarcolemma is the cell membrane of a striated muscle fiber cell and sarcolemmal dense bands were found to be significantly greater in length in fibromyomata and host myometria (non-neoplastic myometrial portion of fibromyomatous uteri) than in normal myometria (Richards et al., 1998). Caveolae are 50–100 nm invaginations of the plasma membrane with an omega (Ω) shape and the numbers of caveolae in host myometria and fibromyomata were found to be decreased in comparison to normal myometria (Richards et al., 1998). Collagens are central structural components of the ECM. In myometrium, collagen structure is ordered with tight bundles, whereas in leiomyoma, they are more loosely packed, with little common orientation (Leppert et al., 2004; Wortham et al., 2006). Myofibrils, composed of myofilaments, are rod-like units of a muscle cell. Eyden et al. reported an abundance of myofilaments with focal densities in both normal myometrium and leiomyomata with the organization of intermediate filaments being alike in both tissues (Eyden et al., 1992). However, a later study reported that myofilaments and intermediate filaments were ordered and aligned in the myometrium while disorganized structures were found in leiomyoma (Wortham et al., 2006).

Extracellular matrix in uterine leiomyoma

The hallmark of uterine leiomyoma is the excessive deposition of ECM and key ECM components including collagens (Fig. 1), fibronectin, laminins and proteoglycans have been reported to be elevated in leiomyoma compared to myometrium (Stewart et al., 1994; Arici and Sozen, 2000; Norian et al., 2009; Malik et al., 2012).

Collagens

Collagen is an insoluble extracellular glycoprotein. It is a key structural component of ECM and maintains cellular morphology. In addition to their roles in wound healing and fibrosis, collagens are known to regulate cell migration, proliferation, differentiation and survival by signaling through cell surface receptors including integrins (Jones and Walker, 1999; Pickering, 2001). Leiomyoma cells have an increased expression of collagen subtypes and are organized by an abnormal collagen structure and orientation (Stewart et al., 1994; Leppert et al., 2004; Malik et al., 2010; Iwahashi and Muragaki, 2011). Stewart et al. reported overexpression of type I and III collagen mRNAs in leiomyoma compared to the adjacent myometrium (Stewart et al., 1994) while later studies confirmed an increase of type I and V collagen protein in leiomyoma compared to normal myometrium (Iwahashi et al., 2010; Iwahashi and Muragaki, 2011). Malik et al. measured levels of a series of collagen subtypes using microarray analysis and found that COL1A1, 4A2, 6A1, 6A2, 7A1 and 16A1 were each expressed to a greater extent in leiomyoma cells than in myometrial cells (Malik et al., 2010). Wolanska and co-scientists similarly reported that collagen concentration was higher in both small (defined as <10 g) and large (defined as >100 g) leiomyoma compared to normal myometrium (Wolanska et al., 1998). They also reported that small leiomyoma had a higher collagen concentration than large leiomyoma (Wolanska et al., 1998) suggesting that excessive accumulation of ECM components is an early event in the formation of these uterine leiomyoma.

Fibronectin

Like collagen, fibronectin is a secreted glycoprotein of the ECM and its role is to attach cells to a variety of ECM types (such as collagen, fibrin and heparin) through interacting with specific membrane receptors (such as integrins). Fibronectin is involved in cell migration, adhesion, growth and differentiation as well as fibrosis, tumor invasion and metastasis (Pankov and Yamada, 2002). Leiomyoma cells are reported to have an elevated level of fibronectin compared to myometrial cells (Arici and Sozen, 2000).

Laminins

Laminins are major glycoprotein components of ECM of the basal lamina (one of the layers of the basement membrane). They are composed of α-, β- and γ-subunits. Laminin has binding regions for collagen and fibronectin (Ockleford et al., 1993; Smith and Ockleford, 1994) and specific integrins also serve as cell surface receptors for laminins. Laminin plays an important role in cell migration, adhesion, growth and differentiation as well as assembly of ECM (Kleinman et al., 1985). Malik et al. demonstrated an increased expression of integrin β1 and integrin α6 subunit as well as laminin 5α, laminin 5β and laminin 5γ subunits in leiomyoma cells compared to myometrial cells (Malik et al., 2012). In addition, they plated leiomyoma cells in laminin coated culture plates, and found their alignment was parallel compared to plastic or collagen-1 coated plates (Malik et al., 2012).

Fibulin-3

Fibulins are calcium-binding glycoproteins that associate with basement membranes and elastic fibers in the ECM. Fibulins may interact with fibronectin, laminins, proteoglycans and tropoelastin, and play important roles in cell morphology, growth, adhesion and motility (Gallagher et al., 2005). Fibulin-3 is encoded by the gene EFEMP1 (EGF-containing fibulin-like ECM protein 1). Marsh et al. demonstrated reduced expression of EFEMP1 and fibulin-3 in leiomyoma compared to normal myometrial cells and tissues by using multiple approaches, including real-time PCR, western blotting, immunohistochemistry and immunofluorescence (Marsh et al., 2016a). Since fibulin-3 acts as an antagonist of angiogenesis (Albig et al., 2006), downregulation of EFEMP1 and the associated loss of fibulin-3 protein may be associated with increased leiomyoma angiogenesis.

Proteoglycans

Proteoglycans are glycosylated proteins consisting of a ‘core protein’ with covalently attached glycosaminoglycans (GAGs). GAGs are long, unbranched heteropolysaccharide chains composed of repeating disaccharide units. The common GAGs are chondroitin sulfate, dermatan sulfate, keratan sulfate, heparan sulfate, heparin and hyaluronic acid. Berto et al. reported that the amounts of chondroitin sulfate and dermatan sulfate (combined data) were increased in leiomyoma compared to normal myometrial tissue (Berto et al., 2001). In addition to the increases in chondroitin sulfate and dermatan sulfate (Berto et al., 2001; Mitropoulou et al., 2001), a rise in keratin sulfate was also found in leiomyoma relative to myometrial tissue (Wolanska et al., 1998). Wolanska et al. also reported elevations in heparan sulfate and heparin in leiomyoma relative to myometrial tissue (Wolanska et al., 1998). By contrast, however, a decrease in heparan sulfate concentration was reported in leiomyoma compared to myometrium in other studies (Berto et al., 2001; Mitropoulou et al., 2001). Hyaluronic acid is a large GAG and is unique in that it does not contain any sulfate and does not attach covalently to proteins. Available evidence suggests that expression levels of hyaluronic acid are not consistently higher or lower in leiomyoma than normal myometrium. For example, Wolanska et al. found no differences in the expression of hyaluronic acid in small leiomyoma compared to myometrial tissue (Wolanska et al., 1998), while another study found less hyaluronic acid in leiomyoma compared to normal myometrium (Mitropoulou et al., 2001). This apparent inconsistency may relate to the rather dynamic expression of proteoglycans in the uterus.

Versican is a large chondroitin sulfate proteoglycan that plays an important role in cell migration, adhesion, proliferation, tissue homeostasis and inflammation (Wight, 2002; Andersson-Sjöland et al., 2014). The expression of several versican subtypes was reported to be higher in leiomyoma tissue and primary cells compared to their healthy tissue counterparts by microarray analysis (Leppert et al., 2006; Malik and Catherino, 2007; Malik et al., 2010). Particularly, versican variant V0 was found to be dramatically elevated in leiomyoma (Malik et al., 2010). In addition, Carrino et al. reported higher amounts of versican in uterine leiomyoma and keloid scars compared to corresponding healthy tissues (Carrino et al., 2012), suggesting a molecular link in the ECM composition between these two fibrotic diseases.

Decorin is a small dermatan sulfate proteoglycan that regulates matrix assembly by binding to fibronectin and collagen via its core protein (Schmidt et al., 1991; Fiedler et al., 2008). Available evidence suggests that uterine leiomyoma contain less decorin than normal myometrial tissue (Carrino et al., 2012; Barker et al., 2015) and, since decorin may act as a antagonist of TGF-β signaling (Droguett et al., 2006), this may cause increased activation of TGF-β signaling that is important for fibrosis. Indeed, decorin treatment of several in vivo models of fibrosis was reported to ameliorate the fibrotic condition (Giri et al., 1997; Logan et al., 1999).

Fibromodulin is a small keratan sulfate proteoglycan that is widely expressed in many connective tissues. It has a close homology with decorin and biglycan. Fibromodulin binds to collagens, and influences the rate of fibrillogenesis (Hedbom and Heinegård, 1993; Font et al., 1998). Levens et al. reported that fibromodulin was highly expressed at mRNA and protein levels in leiomyoma compared to myometrial tissue (Levens et al., 2005).

MMPs and TIMPs

ECM remodeling is an essential process for development, wound healing and homeostasis as well as fibrosis. It is regulated by the combined action of MMPs and TIMPs. MMPs are a class of zinc-dependent endopeptidases that are responsible for the degradation of the ECM while TIMPs act as physiological regulators of the MMPs. Available evidence indicates that several MMPs and TIMPs are differentially expressed at both mRNA and protein level in leiomyoma compared to myometrium. Among the studied MMPs, the expression of MMP-1, -2, -3, -9, -11, -14, -16 and -24 is elevated, while the expression of MMP-7, -19 and -25 is decreased in leiomyoma (Palmer et al., 1998; Wolanska et al., 2004; Dimitrova et al., 2009; Malik et al., 2010; Tsigkou et al., 2015) (Table I). Particularly, the activity and circulating level of MMP-2 are elevated in leiomyoma compared to normal myometrium (Wolanska et al., 2004; Bogusiewicz et al., 2007; Korompelis et al., 2015), indicating a possible dominant role of this MMP in ECM remodeling in leiomyoma. The current research suggests that MMPs are not only ECM-degrading proteases; they participate in a wide range of physiological processes including cell migration, differentiation, growth, innate and adaptive immunity, inflammation, angiogenesis, apoptosis, bone remodeling and neurite growth (Nagase et al., 2006; Rodríguez et al., 2010; Löffek et al., 2011). Some MMPs are antifibrotic, whereas others can have profibrotic functions (Giannandrea and Parks, 2014) (Table 1). They can directly or indirectly affect the functions of various cytokines that play roles in inflammation and repair processes including interferon-β (Nelissen et al., 2003), VEGF (Bergers et al., 2000), EGF (Suzuki et al., 1997), FGF (Suzuki et al., 1997) and TGF-β1 (Shull et al., 1992). MMPs can regulate the bioavailability of angiogenic factors sequestered by the ECM. For instance, ECM-bound VEGF is mobilized by MMPs-1, -3, -7, -9, -16 and -19 cleavage of the VEGF-binding ECM proteins (Bergers et al., 2000; Colnot et al., 2003; Lee et al., 2005). MMPs also enable proteolytic release of different ECM-trapped growth factors such as FGFs or TGF-β that can show strong mitogenic properties (Rodríguez et al., 2010). MMPs can both activate or prevent apoptosis through proteolytic processing of particular signaling molecules. For example, the Fas/Fas ligand (FasL) signal transduction axis is a vital pro-apoptotic system affected by MMPs. MMP-7-mediated release of soluble FasL triggers apoptosis in epithelial cells while inhibiting cell death in tumors (Powell et al., 1999b; Mitsiades et al., 2001). MMP-7 also inhibits apoptosis in cancer cells by cleaving Fas (Strand et al., 2004). Regarding TIMPs, Bogusiewicz et al. reported that the levels of TIMP-1 and TIMP-2 were similar in both myometrial and leiomyoma tissues (Bogusiewicz et al., 2007) while circulating levels of TIMP-1 were reported to be significantly elevated in leiomyoma patients compared to controls (Korompelis et al., 2015). Overall, the available evidence suggests a dysregulation of MMPs and TIMPs that may play a critical role in the formation of a more fibrous ECM in leiomyoma.

Table I

Expression of MMPs and TIMPs in uterine leiomyoma compared to myometrium.

MMMs and TIMPsExpression in leiomyoma relative to myometriumMatrix substrates/MMPsFunctions in different biological systems
MMP-1Collagen types I, II, III, VII and X as well as gelatin, entactin, aggrecan and tenascin (McCawley and Matrisian, 2001)(I) Plays an antifibrotic role in liver fibrosis (Iimuro et al., 2003)
(II) Possesses pro-apoptotic effects (Mannello et al., 2005)
MMP-2Fibronectin, laminin, gelatin, elastin, aggrecan and vitronectin as well as collagen types I, IV, V, VII, X and XI (McCawley and Matrisian, 2001)(I) Plays an antifibrotic role in liver fibrosis (Onozuka et al., 2011)
(II) Promotes angiogenesis (Webb et al., 2017)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-3Fibronectin, laminin, proteoglycans, gelatins, fibrinogen, entactin, tenascin and vitronectin as well as collagen types III, IV, V and IX (McCawley and Matrisian, 2001)(I) Appears to be profibrotic in lung fibrosis (Yamashita et al., 2011)
(II) Appears to play a role in angiogenesis (Anuar et al., 2016)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-7Collagen types III, IV, V, IX, X and XI as well as proteoglycans, laminin, fibronectin, gelatin, fibrinogen, entactin, tenascin and vitronectin (McCawley and Matrisian, 2001)(I) Plays a profibrotic role in lung fibrosis (Manicone et al., 2009)
(II) Induces angiogenesis (Nishizuka et al., 2001)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-9Elastin and gelatin as well as collagen types IV and V (Matrisian, 1992)(I) Possesses both anti- and profibrotic functions in lung fibrosis (Lee et al., 2001; Cabrera et al., 2007)
(II) Promotes angiogenesis (Webb et al., 2017)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP­11Laminin, fibronectin and aggrecan (McCawley and Matrisian, 2001)(I) Shows dual functions in tumourigenesis and cancer progression (Zhang et al., 2016)
(II) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-14Collagen, fibronectin, gelatin, vitronectin, tenascin, nidogen, aggrecan, fibrin, fibrinogen and laminin-5 (McCawley and Matrisian, 2001; Klein and Bischoff, 2011)(I) Shows diverse and opposing functions in scarring or fibrosis (Rohani and Parks, 2015)
(II) Promotes cell migration (Itoh, 2006)
(III) Promotes tumor growth and angiogenesis (Sounni et al., 2002)
MMP-16Collagen type III, gelatin and fibronectin (Matsumoto et al., 1997)(I) promotes cancer progression (Sounni and Noël, 2005)
MMP-19Fibronectin, tenascin-C, gelatin, nidogen and collagen type IV (Stracke et al., 2000)(I) possesses both anti- and profibrotic functions in liver fibrosis (Wang et al., 2011; Jirouskova et al., 2012)
(II) exhibits an anti-angiogenic effect on endothelial cells (Brauer et al., 2011)
MMP-24Fibronectin, gelatin, vitronectin, collagen and aggregan (McCawley and Matrisian, 2001)(I) Plays an important role in ECM remodeling events in the brain and during embryonic development (Pei, 1999)
MMP-25Fibronectin, collagen type IV gelatin, fibrin and laminin-1 (McCawley and Matrisian, 2001)() Appears to play a role in cancer progression (Velasco et al., 2000; Sun et al., 2007)
TIMP-1~Collagenases (MMP-1, MMP-8 and MMP-13), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11 and MMP-18), matrilysins (MMP-7, MMP-26), membranous MMP (MMP-15, MMP-16, MMP-17, MMP-24 and MMP-25), and others (MMP-11, MMP-12, MMP-19, MMP-20, MMP-23 and MMP-28) (Jakubowska et al., 2016)(I) Promotes liver fibrosis (Yoshiji et al., 2000)
(II) Inhibits apoptosis (Li et al., 1999)
(III) Inhibits tumor growth and angiogenesis (Ikenaka et al., 2003)
TIMP-2~Collagenases (MMP-1, MMP-8 and MMP-13), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11 and MMP-18), matrilysins (MMP-7, MMP-26), membranous MMP (MMP-14, MMP-15, MMP-16, MMP-17, MMP-24 and MMP-25), and others (MMP-11, MMP-12, MMP-19, MMP-20, MMP-23 and MMP-28) (Jakubowska et al., 2016)(I) Promotes survival of hepatic stellate cells and endothelial cells (Murphy et al., 2002; Boulday et al., 2004)
(II) Inhibits endothelial cell migration and angiogenesis (Seo et al., 2003; Oh et al., 2004)
MMMs and TIMPsExpression in leiomyoma relative to myometriumMatrix substrates/MMPsFunctions in different biological systems
MMP-1Collagen types I, II, III, VII and X as well as gelatin, entactin, aggrecan and tenascin (McCawley and Matrisian, 2001)(I) Plays an antifibrotic role in liver fibrosis (Iimuro et al., 2003)
(II) Possesses pro-apoptotic effects (Mannello et al., 2005)
MMP-2Fibronectin, laminin, gelatin, elastin, aggrecan and vitronectin as well as collagen types I, IV, V, VII, X and XI (McCawley and Matrisian, 2001)(I) Plays an antifibrotic role in liver fibrosis (Onozuka et al., 2011)
(II) Promotes angiogenesis (Webb et al., 2017)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-3Fibronectin, laminin, proteoglycans, gelatins, fibrinogen, entactin, tenascin and vitronectin as well as collagen types III, IV, V and IX (McCawley and Matrisian, 2001)(I) Appears to be profibrotic in lung fibrosis (Yamashita et al., 2011)
(II) Appears to play a role in angiogenesis (Anuar et al., 2016)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-7Collagen types III, IV, V, IX, X and XI as well as proteoglycans, laminin, fibronectin, gelatin, fibrinogen, entactin, tenascin and vitronectin (McCawley and Matrisian, 2001)(I) Plays a profibrotic role in lung fibrosis (Manicone et al., 2009)
(II) Induces angiogenesis (Nishizuka et al., 2001)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-9Elastin and gelatin as well as collagen types IV and V (Matrisian, 1992)(I) Possesses both anti- and profibrotic functions in lung fibrosis (Lee et al., 2001; Cabrera et al., 2007)
(II) Promotes angiogenesis (Webb et al., 2017)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP­11Laminin, fibronectin and aggrecan (McCawley and Matrisian, 2001)(I) Shows dual functions in tumourigenesis and cancer progression (Zhang et al., 2016)
(II) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-14Collagen, fibronectin, gelatin, vitronectin, tenascin, nidogen, aggrecan, fibrin, fibrinogen and laminin-5 (McCawley and Matrisian, 2001; Klein and Bischoff, 2011)(I) Shows diverse and opposing functions in scarring or fibrosis (Rohani and Parks, 2015)
(II) Promotes cell migration (Itoh, 2006)
(III) Promotes tumor growth and angiogenesis (Sounni et al., 2002)
MMP-16Collagen type III, gelatin and fibronectin (Matsumoto et al., 1997)(I) promotes cancer progression (Sounni and Noël, 2005)
MMP-19Fibronectin, tenascin-C, gelatin, nidogen and collagen type IV (Stracke et al., 2000)(I) possesses both anti- and profibrotic functions in liver fibrosis (Wang et al., 2011; Jirouskova et al., 2012)
(II) exhibits an anti-angiogenic effect on endothelial cells (Brauer et al., 2011)
MMP-24Fibronectin, gelatin, vitronectin, collagen and aggregan (McCawley and Matrisian, 2001)(I) Plays an important role in ECM remodeling events in the brain and during embryonic development (Pei, 1999)
MMP-25Fibronectin, collagen type IV gelatin, fibrin and laminin-1 (McCawley and Matrisian, 2001)() Appears to play a role in cancer progression (Velasco et al., 2000; Sun et al., 2007)
TIMP-1~Collagenases (MMP-1, MMP-8 and MMP-13), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11 and MMP-18), matrilysins (MMP-7, MMP-26), membranous MMP (MMP-15, MMP-16, MMP-17, MMP-24 and MMP-25), and others (MMP-11, MMP-12, MMP-19, MMP-20, MMP-23 and MMP-28) (Jakubowska et al., 2016)(I) Promotes liver fibrosis (Yoshiji et al., 2000)
(II) Inhibits apoptosis (Li et al., 1999)
(III) Inhibits tumor growth and angiogenesis (Ikenaka et al., 2003)
TIMP-2~Collagenases (MMP-1, MMP-8 and MMP-13), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11 and MMP-18), matrilysins (MMP-7, MMP-26), membranous MMP (MMP-14, MMP-15, MMP-16, MMP-17, MMP-24 and MMP-25), and others (MMP-11, MMP-12, MMP-19, MMP-20, MMP-23 and MMP-28) (Jakubowska et al., 2016)(I) Promotes survival of hepatic stellate cells and endothelial cells (Murphy et al., 2002; Boulday et al., 2004)
(II) Inhibits endothelial cell migration and angiogenesis (Seo et al., 2003; Oh et al., 2004)
Table I

Expression of MMPs and TIMPs in uterine leiomyoma compared to myometrium.

MMMs and TIMPsExpression in leiomyoma relative to myometriumMatrix substrates/MMPsFunctions in different biological systems
MMP-1Collagen types I, II, III, VII and X as well as gelatin, entactin, aggrecan and tenascin (McCawley and Matrisian, 2001)(I) Plays an antifibrotic role in liver fibrosis (Iimuro et al., 2003)
(II) Possesses pro-apoptotic effects (Mannello et al., 2005)
MMP-2Fibronectin, laminin, gelatin, elastin, aggrecan and vitronectin as well as collagen types I, IV, V, VII, X and XI (McCawley and Matrisian, 2001)(I) Plays an antifibrotic role in liver fibrosis (Onozuka et al., 2011)
(II) Promotes angiogenesis (Webb et al., 2017)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-3Fibronectin, laminin, proteoglycans, gelatins, fibrinogen, entactin, tenascin and vitronectin as well as collagen types III, IV, V and IX (McCawley and Matrisian, 2001)(I) Appears to be profibrotic in lung fibrosis (Yamashita et al., 2011)
(II) Appears to play a role in angiogenesis (Anuar et al., 2016)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-7Collagen types III, IV, V, IX, X and XI as well as proteoglycans, laminin, fibronectin, gelatin, fibrinogen, entactin, tenascin and vitronectin (McCawley and Matrisian, 2001)(I) Plays a profibrotic role in lung fibrosis (Manicone et al., 2009)
(II) Induces angiogenesis (Nishizuka et al., 2001)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-9Elastin and gelatin as well as collagen types IV and V (Matrisian, 1992)(I) Possesses both anti- and profibrotic functions in lung fibrosis (Lee et al., 2001; Cabrera et al., 2007)
(II) Promotes angiogenesis (Webb et al., 2017)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP­11Laminin, fibronectin and aggrecan (McCawley and Matrisian, 2001)(I) Shows dual functions in tumourigenesis and cancer progression (Zhang et al., 2016)
(II) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-14Collagen, fibronectin, gelatin, vitronectin, tenascin, nidogen, aggrecan, fibrin, fibrinogen and laminin-5 (McCawley and Matrisian, 2001; Klein and Bischoff, 2011)(I) Shows diverse and opposing functions in scarring or fibrosis (Rohani and Parks, 2015)
(II) Promotes cell migration (Itoh, 2006)
(III) Promotes tumor growth and angiogenesis (Sounni et al., 2002)
MMP-16Collagen type III, gelatin and fibronectin (Matsumoto et al., 1997)(I) promotes cancer progression (Sounni and Noël, 2005)
MMP-19Fibronectin, tenascin-C, gelatin, nidogen and collagen type IV (Stracke et al., 2000)(I) possesses both anti- and profibrotic functions in liver fibrosis (Wang et al., 2011; Jirouskova et al., 2012)
(II) exhibits an anti-angiogenic effect on endothelial cells (Brauer et al., 2011)
MMP-24Fibronectin, gelatin, vitronectin, collagen and aggregan (McCawley and Matrisian, 2001)(I) Plays an important role in ECM remodeling events in the brain and during embryonic development (Pei, 1999)
MMP-25Fibronectin, collagen type IV gelatin, fibrin and laminin-1 (McCawley and Matrisian, 2001)() Appears to play a role in cancer progression (Velasco et al., 2000; Sun et al., 2007)
TIMP-1~Collagenases (MMP-1, MMP-8 and MMP-13), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11 and MMP-18), matrilysins (MMP-7, MMP-26), membranous MMP (MMP-15, MMP-16, MMP-17, MMP-24 and MMP-25), and others (MMP-11, MMP-12, MMP-19, MMP-20, MMP-23 and MMP-28) (Jakubowska et al., 2016)(I) Promotes liver fibrosis (Yoshiji et al., 2000)
(II) Inhibits apoptosis (Li et al., 1999)
(III) Inhibits tumor growth and angiogenesis (Ikenaka et al., 2003)
TIMP-2~Collagenases (MMP-1, MMP-8 and MMP-13), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11 and MMP-18), matrilysins (MMP-7, MMP-26), membranous MMP (MMP-14, MMP-15, MMP-16, MMP-17, MMP-24 and MMP-25), and others (MMP-11, MMP-12, MMP-19, MMP-20, MMP-23 and MMP-28) (Jakubowska et al., 2016)(I) Promotes survival of hepatic stellate cells and endothelial cells (Murphy et al., 2002; Boulday et al., 2004)
(II) Inhibits endothelial cell migration and angiogenesis (Seo et al., 2003; Oh et al., 2004)
MMMs and TIMPsExpression in leiomyoma relative to myometriumMatrix substrates/MMPsFunctions in different biological systems
MMP-1Collagen types I, II, III, VII and X as well as gelatin, entactin, aggrecan and tenascin (McCawley and Matrisian, 2001)(I) Plays an antifibrotic role in liver fibrosis (Iimuro et al., 2003)
(II) Possesses pro-apoptotic effects (Mannello et al., 2005)
MMP-2Fibronectin, laminin, gelatin, elastin, aggrecan and vitronectin as well as collagen types I, IV, V, VII, X and XI (McCawley and Matrisian, 2001)(I) Plays an antifibrotic role in liver fibrosis (Onozuka et al., 2011)
(II) Promotes angiogenesis (Webb et al., 2017)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-3Fibronectin, laminin, proteoglycans, gelatins, fibrinogen, entactin, tenascin and vitronectin as well as collagen types III, IV, V and IX (McCawley and Matrisian, 2001)(I) Appears to be profibrotic in lung fibrosis (Yamashita et al., 2011)
(II) Appears to play a role in angiogenesis (Anuar et al., 2016)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-7Collagen types III, IV, V, IX, X and XI as well as proteoglycans, laminin, fibronectin, gelatin, fibrinogen, entactin, tenascin and vitronectin (McCawley and Matrisian, 2001)(I) Plays a profibrotic role in lung fibrosis (Manicone et al., 2009)
(II) Induces angiogenesis (Nishizuka et al., 2001)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-9Elastin and gelatin as well as collagen types IV and V (Matrisian, 1992)(I) Possesses both anti- and profibrotic functions in lung fibrosis (Lee et al., 2001; Cabrera et al., 2007)
(II) Promotes angiogenesis (Webb et al., 2017)
(III) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP­11Laminin, fibronectin and aggrecan (McCawley and Matrisian, 2001)(I) Shows dual functions in tumourigenesis and cancer progression (Zhang et al., 2016)
(II) Possesses both pro-apoptotic and anti-apoptotic effects (Mannello et al., 2005)
MMP-14Collagen, fibronectin, gelatin, vitronectin, tenascin, nidogen, aggrecan, fibrin, fibrinogen and laminin-5 (McCawley and Matrisian, 2001; Klein and Bischoff, 2011)(I) Shows diverse and opposing functions in scarring or fibrosis (Rohani and Parks, 2015)
(II) Promotes cell migration (Itoh, 2006)
(III) Promotes tumor growth and angiogenesis (Sounni et al., 2002)
MMP-16Collagen type III, gelatin and fibronectin (Matsumoto et al., 1997)(I) promotes cancer progression (Sounni and Noël, 2005)
MMP-19Fibronectin, tenascin-C, gelatin, nidogen and collagen type IV (Stracke et al., 2000)(I) possesses both anti- and profibrotic functions in liver fibrosis (Wang et al., 2011; Jirouskova et al., 2012)
(II) exhibits an anti-angiogenic effect on endothelial cells (Brauer et al., 2011)
MMP-24Fibronectin, gelatin, vitronectin, collagen and aggregan (McCawley and Matrisian, 2001)(I) Plays an important role in ECM remodeling events in the brain and during embryonic development (Pei, 1999)
MMP-25Fibronectin, collagen type IV gelatin, fibrin and laminin-1 (McCawley and Matrisian, 2001)() Appears to play a role in cancer progression (Velasco et al., 2000; Sun et al., 2007)
TIMP-1~Collagenases (MMP-1, MMP-8 and MMP-13), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11 and MMP-18), matrilysins (MMP-7, MMP-26), membranous MMP (MMP-15, MMP-16, MMP-17, MMP-24 and MMP-25), and others (MMP-11, MMP-12, MMP-19, MMP-20, MMP-23 and MMP-28) (Jakubowska et al., 2016)(I) Promotes liver fibrosis (Yoshiji et al., 2000)
(II) Inhibits apoptosis (Li et al., 1999)
(III) Inhibits tumor growth and angiogenesis (Ikenaka et al., 2003)
TIMP-2~Collagenases (MMP-1, MMP-8 and MMP-13), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11 and MMP-18), matrilysins (MMP-7, MMP-26), membranous MMP (MMP-14, MMP-15, MMP-16, MMP-17, MMP-24 and MMP-25), and others (MMP-11, MMP-12, MMP-19, MMP-20, MMP-23 and MMP-28) (Jakubowska et al., 2016)(I) Promotes survival of hepatic stellate cells and endothelial cells (Murphy et al., 2002; Boulday et al., 2004)
(II) Inhibits endothelial cell migration and angiogenesis (Seo et al., 2003; Oh et al., 2004)

Mechanotransduction of the extracellular matrix

Mechanotransduction is a process that allows cells to adapt to their changing physical surroundings by sensing their environment and translating mechanical stress into biochemical signals (Huang et al., 2004; Thorne et al., 2015). This process is promoted by the stiffness of the ECM or osmotic stress that initiates cell-matrix interactions in a rapid, transient and bidirectional manner which is referred to as ‘dynamic reciprocity’ (Bissell and Aggeler, 1986; Maniotis et al., 1997; Polacheck et al., 2014; Thorne et al., 2015) (Fig. 2). The activation of downstream mechanical signaling pathways can alter gene expression, leading to changes in ECM density, composition and organization that ultimately affect cell shape and contractility (Fig. 2). Cells sense the mechanical force from newly generated altered ECM and further activate mechanical signaling that result in altered cell behavior and overall remodeling of the matrix. This process appears to play a crucial role in cell adhesion, migration, proliferation and survival as well as inflammation and fibrosis (Seong et al., 2013; Duscher et al., 2014; Thorne et al., 2015). Mechanotransduction occurs at the sites of focal adhesion (Seong et al., 2013; Mui et al., 2016) where large protein complexes serve as a ‘molecular bridge’ between ECM and intracellular molecules/cytoskeleton (Geiger et al., 2001). At these sites, mechanical signals are transmitted from the ECM to the intracellular space through heterodimeric (α and β) transmembrane integrins. Upon ligation and clustering, integrins activate cytoplasmic tyrosine kinases including Src and focal adhesion kinase (FAK). Activation of FAK results in the recruitment of a number of SH2 (Src Homology 2) domain- and SH3 domain-containing proteins that mediate downstream signaling pathways (Parsons, 2003; Schlaepfer and Mitra, 2004) including ERK 1/2, p38 MAPK and JNK (c-Jun N-terminal kinase) (Ruwhof and van der Laarse, 2000; PaszeK et al., 2005). Rho is one of the targets of FAK (Hanks et al., 1992). The Rho family of GTPases, include Rac, Cdc42 and RhoA (Ras homolog gene family, member A). RhoA serves as a ‘molecular switch’ that mediates cycling between active GTP (guanosine triphosphate)-bound and inactive GDP (guanosine diphosphate)-bound states. It plays a crucial role in mediating several profibrotic responses in cardiac fibroblasts (Porter et al., 2004; Zhao et al., 2007). A kinase anchor protein 13 (AKAP13), a RhoA GTPase-specific guanine exchange factor (Rho-GEF), is known to activate RhoA from the inactivated form to the activated RhoA GTPase. The AKAP13/RhoA complex was recently reported to mediate the profibrotic effects in cardiac fibroblasts (Cavin et al., 2014). In addition, AKAP13 appears to play a key role in transmitting the osmotic stress or extracellular hyperosmolarity signal through nuclear factor of activated T cells 5 (NFAT5) (Kino et al., 2009). The cellular osmotic response results in fluid exchange between the cell and ECM that may affect cell shape and ECM composition and organization (Polacheck et al., 2014).

Uterine leiomyoma has been reported to be significantly stiffer than matched myometrium (Rogers et al., 2008), and appears to be under increased mechanical stress (Rogers et al., 2008). In leiomyomal cells, the levels of integrin α6 and integrin β1 were found to be overexpressed compared to normal counterparts (Malik et al., 2012; Chen et al., 2013). In addition, phosphorylation of FAK and ERK was increased in leiomyomal cells (Malik et al., 2012; Chen et al., 2013). The upstream target of ERK, RhoA was overexpressed in leiomyoma compared to myometrial cells (Norian et al., 2012). Malik et al. reported that inhibition of integrin-β1 led to a decrease in active RhoA in leiomyoma cells (Malik et al., 2012). Fasudil, an inhibitor of Rho kinase (ROCK, a downstream target of RhoA), was found to relax the contraction of leiomyoma cells in 3D collagen gels (Malik et al., 2014). The levels of AKAP13 (a Rho-GEF) also appeared higher in leiomyoma compared to myometrium (Rogers et al., 2008). Rogers and co-investigators further reported that p38 MAPK phosphorylation was greater in leiomyoma compared to matched myometrium (Rogers et al., 2008), and this increase was associated with upregulation of AKAP13 (Rogers et al., 2008).

NFAT5 is a transcription factor that induces the expression of genes involved in osmotic stress. Uterine leiomyoma cells demonstrated increased basal expression of NFAT5 compared to myometrial cells. The expression of NFAT5 as well as NFAT5-regulated genes, AR (aldose reductase) and SMIT (sodium myo-inositol transporter 1) was further increased in leiomyoma cells under hyperosmolar conditions (McCarthy-Keith et al., 2011).

A recent study suggested that collagen may play a role in smooth muscle cell proliferation in uterine leiomyoma (Koohestani et al., 2013). Using an in vitro model system of collagen, Koohestani et al. investigated the interaction of cultured leiomyoma smooth muscle cells with monomeric unpolymerized collagen films and fibrillar polymerized collagen gels, and found a significant increase in cell proliferation (Koohestani et al., 2013). The differences in cell proliferation were accompanied by changes in cell cycle progression and p21 (Koohestani et al., 2013). The overall result suggests a possible activation of ECM mediated integrin-FAK-Rho-ERK-p38 MAPK-signaling leading to cell proliferation. Indeed, Malik and co-investigators reported that inhibition of integrin β1 led to a decrease in phosphorylation of ERK as well as reduced proliferation of leiomyoma cells (Malik et al., 2012).

Regulation of extracellular matrix accumulation by growth factors and cytokines

As discussed below, several growth factors and cytokines regulate the production of ECM in leiomyoma cells and can contribute to fibrosis (Islam et al., 2013a, 2016) (Fig. 3).

Transforming growth factor-β

Transforming growth factor-β (TGF-β) is a secreted polypeptide with multifunctional properties. It has three isoforms, referred to as TGF-β1, TGF-β2 and TGF-β3. Myometrial and leiomyoma smooth muscle cells express mRNAs and proteins for each of the three TGF-β isoforms as well as for the TGF-β receptors, TGF-βR-I and TGF-βR-II (Dou et al., 1996; Tang et al., 1997). TGF-β3 appears to increase mRNA expression of ECM components, including collagen 1A1, connective tissue growth factor (CTGF) (Joseph et al., 2010), fibronectin (Arici and Sozen, 2000) and versican V0 (Norian et al., 2009) in both myometrial and leiomyoma cells. However, TGF-β3 was reported to decrease production of MMP-2 and MMP-11 in myometrial and leiomyoma cells (Joseph et al., 2010). In addition to TGF-β3, TGF-β1 has been reported to increase mRNA expression of CTGF (Luo et al., 2006), plasminogen activator inhibitor 1 (PAI-1) (Ding et al., 2004b) and fibromodulin (Levens et al., 2005) in myometrial and leiomyoma smooth muscle cells. The profibrotic role of TGF-β is mediated, at least in part, through activation of Smad 2/3 and ERK 2/3 signaling pathways (Xu et al., 2003; Ding et al., 2004b).

Activin-A

Activin-A, a member of the TGF-β superfamily, was originally isolated for its ability to stimulate secretion of FSH from the anterior pituitary gland but is now known to have wide-ranging physiological and pathophysiological roles in multiple tissues. The mRNA level of activin-A was found to be more highly expressed in leiomyoma compared to myometrium, whereas the levels of activin receptors (ALK4, ActRIIA and ActRIIB) were unchanged (Ciarmela et al., 2011a). The profibrotic role of activin-A in leiomyoma cells has recently been demonstrated by our group (Islam et al., 2014a). We found that activin-A can increase mRNA levels of several ECM components, including collagen 1A1, fibronectin and versican in human primary leiomyoma cells compared to untreated controls (Islam et al., 2014a). Furthermore, activin-A can induce phosphorylation of Smad2 and Smad3 in both myometrial and leiomyoma cells compared to untreated cells (Islam et al., 2014a), suggesting that the fibrotic role of activin-A is mediated, at least in part, by activation of Smad 2/3 signaling pathway.

Platelet-derived growth factor

PDGF is a family of growth factors consisting of four homodimers, namely PDGF-AA, PDGF-BB, PDGF-CC and PDGF-DD, and one heterodimer, PDGF-AB. The protein levels of PDGF-AA, PDGF-BB and PDGF-CC and their receptors were found to be highly expressed in leiomyoma compared to myometrial cells and/or tissues (Liang et al., 2006; Hwu et al., 2008; Yu et al., 2008; Suo et al., 2009). PDGF exerts a profibrotic effect in myometrial and leiomyoma cells through increasing collagen α1 (I) expression (Liang et al., 2006). Additionally, PDGF can stimulate myometrial and leiomyoma cell proliferation (Liang et al., 2006; Mesquita et al., 2010) by regulating of VEGF production (Taniguchi et al., 2001) and activating the ERK 1/2 signaling pathway (Mesquita et al., 2010).

Tumor necrosis factor-α

Tumor necrosis factor (TNF)-α is a pleiotropic cytokine that is primarily secreted by activated macrophages. It plays an important role in controlling inflammation, immunity, cell growth and differentiation, and apoptosis. The level of TNF-α protein expression was found to be higher in leiomyoma compared to normal myometrial cells and tissues (Kurachi et al., 2001; Plewka et al., 2013). Wang et al. reported that TNF-α can significantly upregulate the protein and mRNA levels of MMP-2 in cultured leiomyoma smooth muscle cells but not in matched myometrial smooth muscle cells (Wang et al., 2015). Recently, we found that TNF-α can increase activin-A mRNA expression in both myometrial and leiomyoma cells (Islam et al., 2013b). The increased expression of activin-A in leiomyoma (Ciarmela et al., 2011a), and the ability of activin-A is to increase production of collagen 1A1, fibronectin and versican in leiomyoma smooth muscle cells (Islam et al., 2014a), supports the possible role of TNF-α and activin-A in the process of fibrosis following leiomyoma growth. Additionally, Nair and Al-Hendy demonstrated the possible of role of TNF-α in human uterine leiomyoma cell proliferation (Nair and Al-Hendy, 2011). The proliferative and profibrotic effect of TNF-α in leiomyoma cells was mediated, at least in part, by activating ERK 1/2 signaling pathway (Wang et al., 2015).

Other cytokines

Several cytokines, including interleukin (IL)-1β, IL-11, IL-13 and granulocyte-macrophage colony-stimulating factor (GM-CSF) have been reported to be highly expressed in leiomyoma (Chegini et al., 1999; Ding et al., 2004a; Luo et al., 2005; Syssoev et al., 2008; Plewka et al., 2013). Since these cytokines are known to play important roles in the development of fibrosis in different cellular systems (Xing et al., 1997; Chakir et al., 2003; Fichtner-Feigl et al., 2005; Guo et al., 2013), they may have fibrotic roles in the pathogenesis of leiomyoma.

Hormonal regulation of extracellular matrix accumulation

It is well accepted that estrogen and progesterone play prominent roles as regulators of uterine leiomyoma growth (Maruo et al., 2004; Ciarmela et al., 2011b; Islam et al., 2013a). However, the underlying mechanisms are not completely understood. A number of studies suggest that the stimulatory effects of estrogen and progesterone on leiomyoma growth are mediated, at least in part, by regulation of ECM proteins, growth factors and their signaling pathways (Barbarisi et al., 2001; Chegini et al., 2002; Hoekstra et al., 2009; Nierth-Simpson et al., 2009; Qiang et al., 2014; Barker et al., 2015) (Fig. 3).

Estrogen

Estrogens exert multiple effects on their target cells through binding to estrogen receptors (ERα and ERβ) (Nilsson et al., 2001). Uterine leiomyoma exhibits higher mRNA and protein expression levels of ERα (more highly expressed) and ERβ than normal myometrium (Benassayag et al., 1999; Kovacs et al., 2001). Particularly, ER-α and ER-β are more highly expressed at both mRNA and protein levels in leiomyoma fibroblasts compared to smooth muscle fibroblasts (Feng et al., 2016). Using primary cultures, Zbucka et al. found that collagen biosynthesis was strongly stimulated by low doses of estrogen (5 nM) in leiomyoma cells while no changes occurred in myometrial cells (Zbucka et al., 2008). In contrast, collagen production was inhibited by 10 nM estrogen in both leiomyoma as well as control cells (Zbucka et al., 2008). In uterine leiomyoma xenografts, 17β-estradiol was found to upregulate mRNAs for multiple collagens through downregulation of miR-29b (Qiang et al., 2014). Recently, higher expression of collagen I, fibronectin and laminin, were found in estrogen-treated leiomyoma and smooth muscle fibroblasts compared to untreated control groups (Feng et al., 2016). However, Zbucka et al. found that estrogen can inhibit MMP-2 in leiomyoma and myometrial cells (Zbucka et al., 2008) while transfection of a dominant-negative estrogen receptor resulted in significant augmentation of MMP-1 protein expression in primary and immortalized human leiomyoma cultures (Hassan et al., 2007). Chegini and co-investigators demonstrated that estradiol stimulated both total and active TGF-β1 production in both leiomyoma and myometrial smooth muscle cells (Chegini et al., 2002). The secretion of TGF-β and IGF-1 was also found to be significantly increased in response to estrogen treatment in both leiomyoma and smooth muscle fibroblasts compared to untreated controls (Feng et al., 2016). Recently, we found that estrogen can downregulate activin-A mRNA expression in human myometrial explants (Ciarmela et al., 2011a). Barbarisi et al. reported that estrogen can upregulate PDGF expression in cultured leiomyoma cells (Barbarisi et al., 2001). The ability of estrogen to induce PDGF expression was further confirmed by the observation by Barbarisi et al. that an antiestrogenic compound ICI 182 780 prevented estrogen-induced PDGF expression in cultured leiomyoma cells (Barbarisi et al., 2001). They further reported rapid and transient activation of the MAPK pathway in estrogen-treated leiomyoma smooth muscle cells compared to untreated groups (Barbarisi et al., 2001). Treatment of leiomyoma smooth muscle cells with estrogen was also reported to increase phosphorylation of several downstream intracellular proteins, such as GAP (GTPase-activating protein), PI3K, PLCγ (phospholipase-Cγ) and ERK 1/2 (Barbarisi et al., 2001; Nierth-Simpson et al., 2009). A recent study reported that estrogen can also promote phosphorylation of MEK, ERK1/2 and AKT (also known as protein kinase B or PKB) in both leiomyoma and smooth muscle fibroblasts compared to untreated controls (Feng et al., 2016).

Progesterone

Progesterone exerts physiologic actions on its target cells by interacting with progesterone receptors (PR-A and PR-B) (Kastner et al., 1990). Progesterone seems to be the dominant steroidal influence on uterine leiomyoma as indicated by the increased mitotic rates of leiomyoma cells during the secretory phase of the menstrual cycle (Kawaguchi et al., 1989). Recently, Barker et al. tested the effects of progesterone on decorin expression in human uterine leiomyoma compared with normal myometrial tissue from different stages of the menstrual cycle (Barker et al., 2015). This group found lower expression of decorin at both the mRNA and protein levels in leiomyoma than in adjacent myometrium (Barker et al., 2015). They also found reduced level of decorin mRNA expression in leiomyoma tissue from the secretory phase compared to the proliferative phase (Barker et al., 2015), suggesting an inhibitory effect of progesterone on decorin expression in leiomyoma. As expected, progesterone was found to decrease mRNA expression of decorin in uterine leiomyoma cells compared to controls (Barker et al., 2015). Since decorin inhibits the activity of TGF-β (Yamaguchi et al., 1990), its reduced level in leiomyoma may enhance ECM deposition through TGF-β activity. Chegini et al. demonstrated that treatment of primary myometrial and leiomyoma cell cultures with medroxyprogesterone acetate, a synthetic variant of the steroid hormone progesterone, stimulated both total and active TGF-β1 production (Chegini et al., 2002). Consistently, the highest level of TGF-β3 mRNA was observed in leiomyoma from the mid-secretory phase of menstrual cycle (Arici and Sozen, 2000). Hoekstra et al. reported that progestins can rapidly increase the levels of phosphorylated-AKT and its downstream effectors, pGSK3β and pFOXO1 in leiomyoma cells (Hoekstra et al., 2009). Recently, ECM regulation by progesterone via miRNA was reported by Qiang and co-workers (Qiang et al., 2014). This group found a lower level of miR-29b in uterine leiomyoma compared to myometrial tissues as well as primary cells (Qiang et al., 2014). The restoration of miR-29b in the uterine leiomyoma cells reduced the COL1A1 level in cultured uterine leiomyoma cells. In uterine leiomyoma xenografts, restoring miR-29b also inhibited the accumulation of several collagen subtypes and the development of solid tumors (Qiang et al., 2014). This group demonstrated that progesterone can upregulate mRNA expression of collagens via downregulating miR-29b in a leiomyoma xenograft model (Qiang et al., 2014).

Regulation of extracellular matrix accumulation by microRNA

MicroRNAs (or miRNAs) are small non-coding RNAs (∼22-nt long) that regulate post-transcriptional gene expression. By binding to the 3'-UTR (untranslated region) of target mRNAs, miRNAs prevent protein production by inducing mRNA degradation and/or directly repress translation. A significant number of studies have suggested that miRNAs play an important role in the regulation of ECM accumulation during leiomyoma pathogenesis (Wang et al., 2007; Luo and Chegini, 2008; Chuang et al., 2012a, 2012b; Chuang and Khorram, 2014; Qiang et al., 2014; Chuang and Khorram, 2016; Marsh et al., 2016b).

miR-29 family

miR-29a

Uterine leiomyoma has been demonstrated to have decreased levels of miR-29a in leiomyoma relative to myometrium (Marsh et al., 2016b). Marsh's group used Ambion miRNA precursors to successfully overexpress miRNA-29 in cell culture, and demonstrate its role in ECM production. They found that overexpression of miRNA-29a causes a decrease in the production of collagen subtypes (types II and III) in leiomyoma cells compared to control cells (Marsh et al., 2016b). However, the knockdown of miRNA-29 failed to impact collagen expression in leiomyoma cells compared to control cells (Marsh et al., 2016b).

miR-29b

Uterine leiomyoma expressed a lower level of miR-29b than myometrium (Wang et al., 2007; Qiang et al., 2014; Marsh et al., 2016b) and forced expression of miR-29b reduced the COL1A1, COL2A1 and COL3A1 protein level in primary leiomyoma cells (Qiang et al., 2014; Marsh et al., 2016b). In leiomyoma xenografts, overexpression of miR-29b also inhibited the accumulation of collagens (COL1A1 and COL1A3) and the development of solid tumors (Qiang et al., 2014). However, knockdown of miR-29b in myometrium xenografts increased the expression of collagens (COL1A1, COL1A2, COL3A1, COL5A1, COL5A3 and COL7A1) without transformation into leiomyoma (Qiang et al., 2014). This result suggests that the downregulation of miR-29b is essential but not sufficient for leiomyoma tumourigenesis. Qiang's group reported that estrogen and progesterone can downregulate miR-29b, and upregulate collagen expression in leiomyoma xenografts (Qiang et al., 2014), indicating that steroid hormones promote leiomyoma growth, at least in part, through increasing ECM production via downregulation of the tumor suppressor miR-29b.

miR-29c

Similar to miR-29a and miR-29b, miR-29c is also expressed at low levels in leiomyoma compared to myometrium (Luo and Chegini, 2008; Chuang and Khorram, 2016; Marsh et al., 2016b). Levels of miR-29c were inversely associated with expression of its target, COL3A1 (Chuang and Khorram, 2016) and gain of function of miR-29c inhibited the protein and mRNA expression of COL3A1, and reduced secreted COL3A1, and the rate of cell proliferation (Chuang and Khorram, 2016). Marsh et al. also reported that overexpression of miRNA-29c decreased protein expression of the major collagen subtypes (COL1A1, COL2A1 and COL3A1) in primary leiomyoma cells compared to control cells (Marsh et al., 2016b). The suppression of miR-29c in leiomyoma smooth muscle cells was primarily mediated by steroid hormones, NF-κB and SP1transcription factors, and DNA methylation (Chuang and Khorram, 2016).

miR-200c

Uterine leiomyoma expresses very low levels of miR-200c relative to myometrium (Chuang et al., 2012b; Chuang and Khorram, 2014). Gain-of-function of miR-200c repressed the expression of ZEB1 and ZEB2, TIMP2 and FBLN5 (fibulin 5) at the mRNA and protein levels in leiomyoma and/or smooth muscle cells (Chuang et al., 2012b). Gain-of-function of miR-200c in leiomyoma smooth muscle cells also reduced mRNA and protein levels of IL8 (an inflammatory mediator) (Chuang and Khorram, 2014). In contrast, knockdown of miR-200c produced the opposite effect with a significant increase in IL8 mRNA in these cells (Chuang and Khorram, 2014). This regulation was mediated by suppressing the NF-κB pathway through targeting IKBKB (Chuang and Khorram, 2014).

miR-93/106b

Uterine leiomyoma expresses significantly lower levels of miR-93, but not miR-106b, as compared with myometrium (Chuang et al., 2012a). The gain of function of miR-93 and miR-106b in leiomyoma smooth muscle cells and myometrial smooth muscle cells repressed mRNA and protein levels of tissue factor (F3) and IL8 at through direct interactions with their respective mRNA3'-UTRs (Chuang et al., 2012a) and indirectly inhibited IL8, CTGF and PAI-1 expression through F3 repression (Chuang et al., 2012a).

Extracellular matrix acts as a reservoir for growth factors and modulator of their actions

An elegant experimental in vitro co-culture model by Moore and co-workers has demonstrated that leiomyoma-derived fibroblasts enhance collagen type I production, activate receptor tyrosine kinases (RTKs) and TGF-β signaling and stimulate leiomyoma cell proliferation (Moore et al., 2010). They also found an increased secretion of several growth factors, including IGF, PDGF, TGF-β, EGF and FGF in the ECM of cultured uterine leiomyoma cells (Moore et al., 2010), suggesting an important link among ECM, growth factors, their receptors and signaling pathways in the pathogenesis of leiomyoma.

Uterine leiomyoma is reported to express several GAG components of proteoglycans, such as heparan sulfate, heparin and keratan sulfate (Wolanska et al., 1998; Berto et al., 2001; Mitropoulou et al., 2001). Among these, heparin or heparan sulfate is known to bind to many growth factors. For example, heparan sulfate can bind to bFGF as a cofactor to facilitate the formation and signaling of bFGF-bFGF receptor complexes (Rapraeger et al., 1991; Yayon et al., 1991). Endothelial cell-derived heparan sulfate can also bind to bFGF and protect it from proteolytic degradation (Saksela et al., 1988), effectively serving as a storage depot for this growth factor. bFGF is commonly known as an inducer of angiogenesis and uterine leiomyoma has an increased expression of bFGF in leiomyoma compared to myometrium (Mangrulkar et al., 1995; Wolanska and Bankowski, 2006). FGFR-1 and FGFR-2 receptor expression levels were also found to be elevated in leiomyoma compared to myometrium (Wolanska and Bankowski, 2006; Yu et al., 2008). bFGF was found to be primarily bound to the ECM of myometrium and leiomyoma (Mangrulkar et al., 1995; Dixon et al., 2000). Particularly, leiomyoma showed stronger staining for bFGF because of large areas of ECM in these tumors (Mangrulkar et al., 1995; Dixon et al., 2000). ECM of uterine leiomyoma may therefore serve as a reservoir for bFGF.

A direct physical interaction between VEGF and heparan sulfate has been reported (Ortéga et al., 1998) and VEGF can bind to the heparin-II domain of fibronectin (Wijelath et al., 2006). The activation of VEGFR-2 can induce tyrosine phosphorylation of integrin β3 (Mahabeleshwar et al., 2007). Thus, the relationship between VEGFR-2 and integrin β3 seems to be synergistic. Uterine leiomyoma and myometrium were both reported to express VEGFR-1 and VEGFR-2 (Harrison-Woolrych et al., 1995; Brown et al., 1997; Gentry et al., 2001; Sanci et al., 2011). However, VEGF-A protein levels were found to be higher in leiomyoma than in adjacent myometrium (Gentry et al., 2001). Hassan et al. demonstrated that VEGF was required for continuous growth of tumors or leiomyoma in a xenograft mouse model (Hassan et al., 2008).

Growth factors can also bind to heparan sulfate indirectly through accessory proteins. For example, an activin-binding protein, follistatin, associates with heparan sulfate in follicular granulosa cells (Nakamura et al., 1991). Similarly, TGF-β binds to heparan sulfate via a 60 kDa protein (Bützow et al., 1993). In certain growth factors, such as PDGF-A and VEGF, the major heparin-binding site is localized to a linear 20 amino acid stretch rich in basic residues. The presence of heparan sulfate binding sequences in PDGF-A and VEGF can be modulated by alternative splicing (Raines and Ross, 1992; Park et al., 1993).

The interaction between TGF-β and the core protein of the proteoglycan, decorin has been reported (Border et al., 1990; Yamaguchi et al., 1990). TGF-β1 can also induce the synthesis of decorin by mesangial cells (Border et al., 1990). Decorin, in turn, binds to TGF-β and acts a ligand trap that blocks its ability to bind its receptors on target cells (Yamaguchi et al., 1990), suggesting a negative feedback loop. Uterine leiomyoma tissue contains less decorin protein than normal myometrial tissues (Carrino et al., 2012), suggesting a mechanism for increased activation of TGF-β signaling. Collagen type II contains a chordin-like VWC domain which can also bind to TGF-β1 (Abreu et al., 2002) and act as a negative regulator for this growth factor. By contrast, a membrane-anchored proteoglycan, betaglycan (TGF-β type III receptor) was reported to facilitate the formation of TGF-β receptors complexes (Shi and Massagué, 2003). TGF-β binds first to betaglycan and that binding and ‘presentation’ play key roles in mediating TGF-β binding to the type II receptor (Shi and Massagué, 2003).

A hallmark of the integrins is the ability to recognize multiple ligands. They can activate growth factor mediated signaling pathways independently (Assoian and Schwartz, 2001) or synergistically with other receptors (Alam et al., 2007). These include TGF-β receptors (Scaffidi et al., 2004), VEGF receptors (Mahabeleshwar et al., 2007), PDGF-β receptors (Schneller et al., 1997), insulin-like growth factor receptors (IGFR) (Doerr and Jones, 1996), and EGF receptors (Bill et al., 2004). In addition to integrins, several ECM proteins, including laminin, fibrillin, tenascin and thrombospondin contain EGF-like domains, which can directly bind to EGF receptors and modulate their signaling (Schenk et al., 2003).

Fibrosis in uterine leiomyoma: role of myofibroblasts and inflammation

Fibrosis is caused by an excessive accumulation of ECM proteins resulting from an imbalance in wound healing and repair processes during chronic tissue injury and/or inflammation (Wynn, 2007). Myofibroblasts are specialized cell type, activated by tissue injury, inflammation, hypoxia and oxidative stress, that play critical roles in wound healing and tissue homeostasis as well as fibrosis (Poli, 2000; Higgins et al., 2007; Wynn, 2007; Toullec et al., 2010) (Fig. 1). A biological function of myofibroblasts is to produce ECM proteins in order to heal wounds and maintain functional integrity of organs/tissues after injury. After finishing this role in tissue repair and homeostasis, myofibroblasts disappear via apoptosis (Desmouliere et al., 1995). When this process fails to proceed normally, persistence of cells with myofibroblastic phenotype can lead to excessive ECM production and development of fibrosis (Powell et al., 1999a; Tomasek et al., 2002) (Fig. 1).

Myofibroblast cells are characterized by α-SMA expression and a high level of collagen synthesis (Wynn, 2008). The fibronectin domain ED-A is also crucial for the myofibroblastic phenotype (Serini et al., 1998). Several cell types, including fibroblasts, fibrocytes, stem cells, smooth muscle cells and endothelial cells have been reported to acquire the myofibroblast phenotype in large variety of organs (Hinz et al., 2007) and a number of studies suggest that myofibroblastic transformation could occur from fibroblasts, stem cells and smooth muscle cells in the myometrium (Ono et al., 2007; Chang et al., 2010; Moore et al., 2010; Holdsworth-Carson et al., 2014; Zheng et al., 2014; Mas et al., 2015; Yin et al., 2015; Feng et al., 2016; Protic et al., 2016) (Fig. 1). CD90, a fibroblast specific marker, was found to be expressed in both smooth muscle leiomyoma fibroblasts but not in smooth muscle cells themselves (Luo et al., 2014). In addition, FAP (fibroblast activation protein), a marker of active fibroblasts, was highly expressed at the protein level in fibroblasts from uterine leiomyoma compared to those from normal muscle tissues (Luo et al., 2014). Recently, we identified numerous CD34+ fibroblasts in myometrium and leiomyoma, which are known to give origin to myofibroblasts when they lose CD34 expression (Protic et al., 2016). In addition, Yin et al. identified human uterine leiomyoma stem/progenitor cells expressing CD34 and CD49b, which are able to initiate tumors in vivo (Yin et al., 2015). In our recent study, we reported the presence of α-SMA positive and desmin negative cells as well as large amount of collagen inside the leiomyoma tissue that supports the existence of myofibroblast and fibrotic characters of this tumor (Protic et al., 2016).

Myofibroblast differentiation depends on environmental cues, including tension in the matrix and a variety of soluble mediators, such as growth factors, steroid hormones, cytokines and chemokines (Tomasek et al., 2002; Gabbiani, 2003; Wipff et al., 2007; Wynn, 2007, 2008; Luo et al., 2014; Feng et al., 2016). The central player of myofibroblast differentiation is TGF-β. It is induced by mechanical tension, induces α-SMA expression (Desmoulière et al., 1993), and enhances the synthesis of both collagens (Lindahl et al., 2002) and ED-A fibronectin (Serini et al., 1998). The cross-linking of the ECM may further activate resident cells to myofibroblastic transition (Desmouliere et al., 2005; Ho et al., 2014). In addition to TGF-β, activin-A has been demonstrated to play an important role in tissue repair as well as fibrosis (Werner et al., 1999). Both TGF-β and activin-A are highly expressed in uterine leiomyoma (Dou et al., 1996; Tang et al., 1997; Ciarmela et al., 2011a) where their abilities to enhance synthesis of ECM components, such as collagen 1A1, CTGF, fibronectin, versican and PAI-1 (Arici and Sozen, 2000; Ding et al., 2004b; Joseph et al., 2010; Islam et al., 2014a), supports their roles in driving myofibroblast differentiation. Recently, Feng et al. reported that after a biological insult of serum starvation and serum add-back, myometrial cells undergo a transition to a myofibroblast-like phenotype that was related to an increased activation of TGF-β signaling via the Smad 2/3 pathway (Feng et al., 2016). These authors further showed that in response to TGF-β3 treatment during the biological insult, myometrial cells migrated into nodules containing collagen and fibronectin (Feng et al., 2016). Using transmission electron microscopy, they also found myofibroblast-like cells and fibril-like structures in the extracellular spaces of the nodules (Feng et al., 2016).

In addition to TGF-β and activin-A, TNF-α can be considered as an important regulator of myofibroblast differentiation. Indeed, TNF-α increases the expression of activin-A in both myometrial and leiomyoma cells (Islam et al., 2013b) and thereby promotes the fibrotic role of activin-A in the process of myofibroblast differentiation following fibrosis.

Estrogen and progesterone have been reported to increase the proliferation of both leiomyoma and smooth muscle fibroblasts compared to untreated groups (Luo et al., 2014). Estrogen-mediated fibroblast proliferation was mediated, at least in part, by increased secretion of TGF-β and IGF-1 as well as activation of MEK/ERK1/2 and AKT pathways (Luo et al., 2014). The effect of estrogen on fibroblast activation was documented by the observation that expression of FAP as well as collagen I, fibronectin and laminin proteins was increased in estrogen-treated leiomyoma and smooth muscle fibroblasts compared to untreated controls (Luo et al., 2014). FAP knockdown was reported to attenuate the estrogen-mediated proliferation of fibroblasts, as well as phosphorylation levels of MEK, ERK, AKT and protein levels of c-fos (Luo et al., 2014). The decreased protein expression of ECM components (collagen I, fibronectin and laminin) was also observed in leiomyoma fibroblasts compared to untreated controls in response to suppression of FAP expression (Luo et al., 2014). These results suggest that the effect of estrogen on fibroblast cell activation is mediated partially through the FAP pathway.

Inflammation is a key component of the wound healing process. However, local chronic inflammation makes a suitable microenvironment for development of fibrosis. Chronic inflammation is characterized by infiltration of mononuclear immune cells (macrophages, monocytes, lymphocytes and plasma cells). Of note, inflammatory cells, in particular macrophages, are widely accepted as important regulators of cytokines and growth factors during the wound healing process (Leibovich and Ross, 1975). Recently, we found higher numbers of macrophages present inside and in the vicinity of leiomyoma compared to the more distant surrounding myometrium (Protic et al., 2016).

Chronic inflammatory reactions are induced by a variety of stimuli, including chemical insults, radiation, persistent infections, tissue injury, autoimmune reactions and allergic responses (Wynn, 2008). In the uterus, reproductive events, including ovulation, menstruation and implantation, may trigger inflammatory reactions. In addition, many factors have been hypothesized to increase inflammation in uterus, such as infection, injury, talc use, an intrauterine device, cesarean section, male reproductive proteins and stress (work, home or perceived racism) (Wegienka, 2012). Repeated stimulation by reproductive events, mechanical forces, injury, hypoxia and oxidative stress may also create a chronic inflammatory state in the uterus (Wegienka, 2012; Fletcher et al., 2013; Leppert et al., 2013; Santulli et al., 2013). Regardless of the cause of chronic uterine inflammation, it is under this condition that myofibroblasts cells produce ECM in an unregulated manner and fail to undergo normal apoptosis leading to fibrosis.

Although most women experience causes of uterine inflammation, e.g. reproductive events, they all do not have leiomyoma. This observation suggests that initiation of uterine leiomyoma does not solely depend on inflammation. There are other factors that may influence the risk of developing leiomyoma under the chronic inflammatory condition. Accumulated evidence suggests that genetic and epigenetic factors may influence the risk for developing leiomyoma (Gallagher and Morton, 2016; Yang et al., 2016). In addition, black race, heredity, nulliparity, obesity, polycystic ovary syndrome, diabetes and hypertension are associated with increased risk of this tumor (Okolo, 2008).

Extracellular matrix as a therapeutic target of current and future medical treatments

Several classes of compounds including GnRH agonizts, GnRH antagonists, selective progesterone receptor modulators, antiprogestin and natural compounds have been studied as medical treatments that target ECM in uterine leiomyoma (Table II).

Table II

Current and potential treatments for leiomyoma that target the extracellular matrix.

Treatment modalitiesFibrosis-related molecular targetsEffects on leiomyoma and related symptomsSafety and side effects
Current treatments
 Leuprolide  acetate↓TGF-β1, ↓TGF-β3, ↓TGF-βR type I, ↓TGF-βR type II, ↓Smad4, ↓pSmad3, ↑Smad7, ↓ collagen type I, ↓↑fibronectin, ↓versican, ↓fibromodulin, ↓TIMP-1, ↑MMP-1, ↑MMP-2, ↑MMP-3, and ↑MMP-9(I) Reduces uterine and leiomyoma volume (Stovall et al., 1995)(I) Prolonged therapy (>6 months) is not recommended
(II) Alleviates bleeding and increase hemoglobin levels (Stovall et al., 1991)(II) Side effects-hot flashes, vaginitis and bone loss (Stovall et al., 1995)
 Cetrorelix  acetate↓COL1A1, ↓fibronectin, and ↓versican variant V0(I) Decreases uterine and leiomyoma volume (Gonzalez-Barcena et al., 1997; Felberbaum et al., 2001; Engel et al., 2007)(I) Side effects-amenorrhea and hot flashes (Gonzalez-Barcena et al., 1997)
(II) Abolishes symptoms of menorrhagia and reduce uterine pain (Engel et al., 2007)
 Asoprisnil↓TGF-β3, ↓TGF-βR type II, ↑EMMPRIN, ↑MMP-1, ↑MT1-MMP, ↓TIMP-1, ↓TIMP-2, ↓ collagen type I and ↓collagen type III(I) Suppress leiomyoma and uterine volume (Chwalisz et al., 2007)(I) Abnormal endometrial changes (Williams et al., 2007)
(II) Reduces uterine bleeding as well as bloating and pelvic pressure (Chwalisz et al., 2007)(II) Side effects-hot flashes or night sweats (Chwalisz et al., 2007)
 Ulipristal  acetate↑MMP-1, ↑MMP-2, ↑MMP-3, ↑MMP-8, ↑MMP-9, ↑EMMPRIN, ↓TIMP-1, ↓TIMP-2, ↓ collagen type I, ↓collagen type III and ↓fibronectin(I) Reduces leiomyoma and uterine volume (Donnez et al., 2012a, 2012b., 2015)(I) Quite well tolerated (Donnez et al., 2015).
(II) Controls bleeding and pain (Donnez et al., 2015)(II) Side effects-hot flashes and headaches (Donnez et al., 2015)
(III) Improves quality of life (Donnez et al., 2012a, 2012b., 2015)
 Mifepristone↓COL1A1, ↓fibronectin, ↓versican and ↓dermatopontin(I) Reduces uterine and leiomyoma volume (Esteve et al., 2012)(I) No endometrial hyperplasia or cellular atypia(Yerushalmi et al., 2014)
(II) Alleviates hypermenorrhea, menstrual blood loss, pelvic painandpressure, anemia and dysmenorrhea (Shen et al., 2013; Yerushalmi et al., 2014)(II) Side effects-hot flashes, nausea, weakness, abdominal pain and vaginal discharge (Yerushalmi et al., 2014)
(III) Reduces leiomyoma cell viability and proliferation (Chung et al., 2014)
 Raloxifene↓Collagen and ↓MMP-2(I) prevents progression of uterine leiomyoma (Jirecek et al., 2004)(I) quite well tolerated (Jirecek et al., 2004)
(II) side effects-hot flashes and leg cramps (Martino et al., 2005), and risk of venous thromboembolism (Ettinger et al., 1999)
Investigational compounds
 CP8947↓COL1A1 and ↓COL7A1(I) Inhibits the leiomyoma cell proliferation (Catherino et al., 2010)Limited
(II) No effect on myometrial cell proliferation (Catherino et al., 2010)
 2-methoxyestradiol↓Collagen type I, ↓Collagen type III, ↓PAI-1, ↓CTGF, ↓α-SMA, ↓pSmad2/3 and ↓PI3K/Akt/mTOR(I) Inhibits cell proliferation in rat and human leiomyoma cells (Salama et al., 2006)(I) Well tolerated (Harrison et al., 2011)
(II) induces apoptosis in rat and human leiomyoma cells(Salama et al., 2006)(II) Side effects-hot flashes, muscle cramps, headache, fatigue or weakness, nausea, vomiting, diarrhea, gastrointestinal hemorrhage and hyponatremia (Rajkumar et al., 2007)
 Liarozole↓COL1A1, ↓COL4A2, ↓versican, ↓fibromodulin, ↓fibronectin and ↓TGF-β3(I) Inhibits proliferation of both myometrial and leiomyoma cells (Gilden et al., 2012)(I) Well tolerated (Denis et al., 1998)
(II) Side effects-skin disorders and dryness of mouth/eyes/lips (Goss et al., 2000)
 All-trans retinoic  acid↓Collagen 1, ↓collagen 4, ↓fibronectin, ↓versican and ↓TGF-β3(I) Downregulates immortalized leiomyoma cell proliferation (Malik et al., 2008)(I) Quite well tolerated (Kurzrock et al., 1993; Böcher et al., 2008)
(II) Side effects-transient headache, dry skin and mucosa, nausea and vomiting, myalgias or muscle pain, dyspnea (shortness of breath), and sensorineural hearing loss (Kurzrock et al., 1993; Böcher et al., 2008)
 Vitamin D↓fibronectin, ↓collagen type 1, ↓PAI-1, ↓pSmad2, ↓Wnt4, ↓β-catenin, ↓mTOR, ↓fibromodulin, ↓biglycan and ↓versican(I) Shrinks uterine leiomyoma tumors in Eker rat model (Halder et al., 2012)Limited
 Celecoxib↓collagen A, ↓fibronectin, ↓PDGF, and ↓TGF-β(I) Inhibits leiomyoma cell proliferation (Ke et al., 2013)(I) Increases the risk of myocardial infarction, stroke or heart failure (Solomon et al., 2005)
(II) No effect on myometrial cell proliferation (Ke et al., 2013)
 Tranilast↓COL1A1, ↓fibronectin ↓versican and ↓activin-A(I) Inhibits proliferation of myometrial and leiomyoma cells (Shime et al., 2002; Islam et al., 2012)(I) Well tolerated with low toxicity (Konneh, 1998)
 Pirfenidone↓collagen type I, ↓collagen type III(I) Inhibits proliferation of myometrial and leiomyoma cells (Lee et al., 1998)(I) Rather well tolerated by patients with idiopathic pulmonary fibrosis (Chaudhuri et al., 2014)
(II) Gastrointestinal adverse effects (Chaudhuri et al., 2014)
 Halofuginone↓collagen type I (a1), ↓collagen type III (a1) and ↓TGF-β1(I) Reduces uterine leiomyoma volume in a mouse xenograft model (Koohestani et al., 2016)(I) Appears to be safe and well tolerated (De Jonge et al., 2006)
(II) Inhibits both myometrial and leiomyoma cell proliferation (Grudzien et al., 2010)(II) No clinically adverse events (De Jonge et al., 2006)
(III) Induces apoptosis (Grudzien et al., 2010)
 Curcumin↓fibronectin(I) Inhibits leiomyoma cell proliferation (Malik et al., 2009)(I) Appears to be safe and well tolerated by patients (Gupta et al., 2013)
(II) Side effects-nausea, diarrhea, headache, rash and yellow stool (Gupta et al., 2013)
 Resveratrol↓fibronectin, ↓collagen types I, ↓collagen III, ↓fibromodulin. ↓biglycan, and ↓MMP-9, ↑TIMP2(I) Inhibits proliferation of human uterine leiomyoma cells (Catherino et al., 2011)(I) Appears safe and well tolerated by patients (Turner et al., 2015)
(II) Adverse events-nausea, diarrhea and weight loss (Turner et al., 2015)
 Collagenase C.  histolyticum↓ collagen(I) Reduces fibrosis (collagen) and tissue stiffness in uterine leiomyoma (Jayes et al., 2016)Limited
Treatment modalitiesFibrosis-related molecular targetsEffects on leiomyoma and related symptomsSafety and side effects
Current treatments
 Leuprolide  acetate↓TGF-β1, ↓TGF-β3, ↓TGF-βR type I, ↓TGF-βR type II, ↓Smad4, ↓pSmad3, ↑Smad7, ↓ collagen type I, ↓↑fibronectin, ↓versican, ↓fibromodulin, ↓TIMP-1, ↑MMP-1, ↑MMP-2, ↑MMP-3, and ↑MMP-9(I) Reduces uterine and leiomyoma volume (Stovall et al., 1995)(I) Prolonged therapy (>6 months) is not recommended
(II) Alleviates bleeding and increase hemoglobin levels (Stovall et al., 1991)(II) Side effects-hot flashes, vaginitis and bone loss (Stovall et al., 1995)
 Cetrorelix  acetate↓COL1A1, ↓fibronectin, and ↓versican variant V0(I) Decreases uterine and leiomyoma volume (Gonzalez-Barcena et al., 1997; Felberbaum et al., 2001; Engel et al., 2007)(I) Side effects-amenorrhea and hot flashes (Gonzalez-Barcena et al., 1997)
(II) Abolishes symptoms of menorrhagia and reduce uterine pain (Engel et al., 2007)
 Asoprisnil↓TGF-β3, ↓TGF-βR type II, ↑EMMPRIN, ↑MMP-1, ↑MT1-MMP, ↓TIMP-1, ↓TIMP-2, ↓ collagen type I and ↓collagen type III(I) Suppress leiomyoma and uterine volume (Chwalisz et al., 2007)(I) Abnormal endometrial changes (Williams et al., 2007)
(II) Reduces uterine bleeding as well as bloating and pelvic pressure (Chwalisz et al., 2007)(II) Side effects-hot flashes or night sweats (Chwalisz et al., 2007)
 Ulipristal  acetate↑MMP-1, ↑MMP-2, ↑MMP-3, ↑MMP-8, ↑MMP-9, ↑EMMPRIN, ↓TIMP-1, ↓TIMP-2, ↓ collagen type I, ↓collagen type III and ↓fibronectin(I) Reduces leiomyoma and uterine volume (Donnez et al., 2012a, 2012b., 2015)(I) Quite well tolerated (Donnez et al., 2015).
(II) Controls bleeding and pain (Donnez et al., 2015)(II) Side effects-hot flashes and headaches (Donnez et al., 2015)
(III) Improves quality of life (Donnez et al., 2012a, 2012b., 2015)
 Mifepristone↓COL1A1, ↓fibronectin, ↓versican and ↓dermatopontin(I) Reduces uterine and leiomyoma volume (Esteve et al., 2012)(I) No endometrial hyperplasia or cellular atypia(Yerushalmi et al., 2014)
(II) Alleviates hypermenorrhea, menstrual blood loss, pelvic painandpressure, anemia and dysmenorrhea (Shen et al., 2013; Yerushalmi et al., 2014)(II) Side effects-hot flashes, nausea, weakness, abdominal pain and vaginal discharge (Yerushalmi et al., 2014)
(III) Reduces leiomyoma cell viability and proliferation (Chung et al., 2014)
 Raloxifene↓Collagen and ↓MMP-2(I) prevents progression of uterine leiomyoma (Jirecek et al., 2004)(I) quite well tolerated (Jirecek et al., 2004)
(II) side effects-hot flashes and leg cramps (Martino et al., 2005), and risk of venous thromboembolism (Ettinger et al., 1999)
Investigational compounds
 CP8947↓COL1A1 and ↓COL7A1(I) Inhibits the leiomyoma cell proliferation (Catherino et al., 2010)Limited
(II) No effect on myometrial cell proliferation (Catherino et al., 2010)
 2-methoxyestradiol↓Collagen type I, ↓Collagen type III, ↓PAI-1, ↓CTGF, ↓α-SMA, ↓pSmad2/3 and ↓PI3K/Akt/mTOR(I) Inhibits cell proliferation in rat and human leiomyoma cells (Salama et al., 2006)(I) Well tolerated (Harrison et al., 2011)
(II) induces apoptosis in rat and human leiomyoma cells(Salama et al., 2006)(II) Side effects-hot flashes, muscle cramps, headache, fatigue or weakness, nausea, vomiting, diarrhea, gastrointestinal hemorrhage and hyponatremia (Rajkumar et al., 2007)
 Liarozole↓COL1A1, ↓COL4A2, ↓versican, ↓fibromodulin, ↓fibronectin and ↓TGF-β3(I) Inhibits proliferation of both myometrial and leiomyoma cells (Gilden et al., 2012)(I) Well tolerated (Denis et al., 1998)
(II) Side effects-skin disorders and dryness of mouth/eyes/lips (Goss et al., 2000)
 All-trans retinoic  acid↓Collagen 1, ↓collagen 4, ↓fibronectin, ↓versican and ↓TGF-β3(I) Downregulates immortalized leiomyoma cell proliferation (Malik et al., 2008)(I) Quite well tolerated (Kurzrock et al., 1993; Böcher et al., 2008)
(II) Side effects-transient headache, dry skin and mucosa, nausea and vomiting, myalgias or muscle pain, dyspnea (shortness of breath), and sensorineural hearing loss (Kurzrock et al., 1993; Böcher et al., 2008)
 Vitamin D↓fibronectin, ↓collagen type 1, ↓PAI-1, ↓pSmad2, ↓Wnt4, ↓β-catenin, ↓mTOR, ↓fibromodulin, ↓biglycan and ↓versican(I) Shrinks uterine leiomyoma tumors in Eker rat model (Halder et al., 2012)Limited
 Celecoxib↓collagen A, ↓fibronectin, ↓PDGF, and ↓TGF-β(I) Inhibits leiomyoma cell proliferation (Ke et al., 2013)(I) Increases the risk of myocardial infarction, stroke or heart failure (Solomon et al., 2005)
(II) No effect on myometrial cell proliferation (Ke et al., 2013)
 Tranilast↓COL1A1, ↓fibronectin ↓versican and ↓activin-A(I) Inhibits proliferation of myometrial and leiomyoma cells (Shime et al., 2002; Islam et al., 2012)(I) Well tolerated with low toxicity (Konneh, 1998)
 Pirfenidone↓collagen type I, ↓collagen type III(I) Inhibits proliferation of myometrial and leiomyoma cells (Lee et al., 1998)(I) Rather well tolerated by patients with idiopathic pulmonary fibrosis (Chaudhuri et al., 2014)
(II) Gastrointestinal adverse effects (Chaudhuri et al., 2014)
 Halofuginone↓collagen type I (a1), ↓collagen type III (a1) and ↓TGF-β1(I) Reduces uterine leiomyoma volume in a mouse xenograft model (Koohestani et al., 2016)(I) Appears to be safe and well tolerated (De Jonge et al., 2006)
(II) Inhibits both myometrial and leiomyoma cell proliferation (Grudzien et al., 2010)(II) No clinically adverse events (De Jonge et al., 2006)
(III) Induces apoptosis (Grudzien et al., 2010)
 Curcumin↓fibronectin(I) Inhibits leiomyoma cell proliferation (Malik et al., 2009)(I) Appears to be safe and well tolerated by patients (Gupta et al., 2013)
(II) Side effects-nausea, diarrhea, headache, rash and yellow stool (Gupta et al., 2013)
 Resveratrol↓fibronectin, ↓collagen types I, ↓collagen III, ↓fibromodulin. ↓biglycan, and ↓MMP-9, ↑TIMP2(I) Inhibits proliferation of human uterine leiomyoma cells (Catherino et al., 2011)(I) Appears safe and well tolerated by patients (Turner et al., 2015)
(II) Adverse events-nausea, diarrhea and weight loss (Turner et al., 2015)
 Collagenase C.  histolyticum↓ collagen(I) Reduces fibrosis (collagen) and tissue stiffness in uterine leiomyoma (Jayes et al., 2016)Limited
Table II

Current and potential treatments for leiomyoma that target the extracellular matrix.

Treatment modalitiesFibrosis-related molecular targetsEffects on leiomyoma and related symptomsSafety and side effects
Current treatments
 Leuprolide  acetate↓TGF-β1, ↓TGF-β3, ↓TGF-βR type I, ↓TGF-βR type II, ↓Smad4, ↓pSmad3, ↑Smad7, ↓ collagen type I, ↓↑fibronectin, ↓versican, ↓fibromodulin, ↓TIMP-1, ↑MMP-1, ↑MMP-2, ↑MMP-3, and ↑MMP-9(I) Reduces uterine and leiomyoma volume (Stovall et al., 1995)(I) Prolonged therapy (>6 months) is not recommended
(II) Alleviates bleeding and increase hemoglobin levels (Stovall et al., 1991)(II) Side effects-hot flashes, vaginitis and bone loss (Stovall et al., 1995)
 Cetrorelix  acetate↓COL1A1, ↓fibronectin, and ↓versican variant V0(I) Decreases uterine and leiomyoma volume (Gonzalez-Barcena et al., 1997; Felberbaum et al., 2001; Engel et al., 2007)(I) Side effects-amenorrhea and hot flashes (Gonzalez-Barcena et al., 1997)
(II) Abolishes symptoms of menorrhagia and reduce uterine pain (Engel et al., 2007)
 Asoprisnil↓TGF-β3, ↓TGF-βR type II, ↑EMMPRIN, ↑MMP-1, ↑MT1-MMP, ↓TIMP-1, ↓TIMP-2, ↓ collagen type I and ↓collagen type III(I) Suppress leiomyoma and uterine volume (Chwalisz et al., 2007)(I) Abnormal endometrial changes (Williams et al., 2007)
(II) Reduces uterine bleeding as well as bloating and pelvic pressure (Chwalisz et al., 2007)(II) Side effects-hot flashes or night sweats (Chwalisz et al., 2007)
 Ulipristal  acetate↑MMP-1, ↑MMP-2, ↑MMP-3, ↑MMP-8, ↑MMP-9, ↑EMMPRIN, ↓TIMP-1, ↓TIMP-2, ↓ collagen type I, ↓collagen type III and ↓fibronectin(I) Reduces leiomyoma and uterine volume (Donnez et al., 2012a, 2012b., 2015)(I) Quite well tolerated (Donnez et al., 2015).
(II) Controls bleeding and pain (Donnez et al., 2015)(II) Side effects-hot flashes and headaches (Donnez et al., 2015)
(III) Improves quality of life (Donnez et al., 2012a, 2012b., 2015)
 Mifepristone↓COL1A1, ↓fibronectin, ↓versican and ↓dermatopontin(I) Reduces uterine and leiomyoma volume (Esteve et al., 2012)(I) No endometrial hyperplasia or cellular atypia(Yerushalmi et al., 2014)
(II) Alleviates hypermenorrhea, menstrual blood loss, pelvic painandpressure, anemia and dysmenorrhea (Shen et al., 2013; Yerushalmi et al., 2014)(II) Side effects-hot flashes, nausea, weakness, abdominal pain and vaginal discharge (Yerushalmi et al., 2014)
(III) Reduces leiomyoma cell viability and proliferation (Chung et al., 2014)
 Raloxifene↓Collagen and ↓MMP-2(I) prevents progression of uterine leiomyoma (Jirecek et al., 2004)(I) quite well tolerated (Jirecek et al., 2004)
(II) side effects-hot flashes and leg cramps (Martino et al., 2005), and risk of venous thromboembolism (Ettinger et al., 1999)
Investigational compounds
 CP8947↓COL1A1 and ↓COL7A1(I) Inhibits the leiomyoma cell proliferation (Catherino et al., 2010)Limited
(II) No effect on myometrial cell proliferation (Catherino et al., 2010)
 2-methoxyestradiol↓Collagen type I, ↓Collagen type III, ↓PAI-1, ↓CTGF, ↓α-SMA, ↓pSmad2/3 and ↓PI3K/Akt/mTOR(I) Inhibits cell proliferation in rat and human leiomyoma cells (Salama et al., 2006)(I) Well tolerated (Harrison et al., 2011)
(II) induces apoptosis in rat and human leiomyoma cells(Salama et al., 2006)(II) Side effects-hot flashes, muscle cramps, headache, fatigue or weakness, nausea, vomiting, diarrhea, gastrointestinal hemorrhage and hyponatremia (Rajkumar et al., 2007)
 Liarozole↓COL1A1, ↓COL4A2, ↓versican, ↓fibromodulin, ↓fibronectin and ↓TGF-β3(I) Inhibits proliferation of both myometrial and leiomyoma cells (Gilden et al., 2012)(I) Well tolerated (Denis et al., 1998)
(II) Side effects-skin disorders and dryness of mouth/eyes/lips (Goss et al., 2000)
 All-trans retinoic  acid↓Collagen 1, ↓collagen 4, ↓fibronectin, ↓versican and ↓TGF-β3(I) Downregulates immortalized leiomyoma cell proliferation (Malik et al., 2008)(I) Quite well tolerated (Kurzrock et al., 1993; Böcher et al., 2008)
(II) Side effects-transient headache, dry skin and mucosa, nausea and vomiting, myalgias or muscle pain, dyspnea (shortness of breath), and sensorineural hearing loss (Kurzrock et al., 1993; Böcher et al., 2008)
 Vitamin D↓fibronectin, ↓collagen type 1, ↓PAI-1, ↓pSmad2, ↓Wnt4, ↓β-catenin, ↓mTOR, ↓fibromodulin, ↓biglycan and ↓versican(I) Shrinks uterine leiomyoma tumors in Eker rat model (Halder et al., 2012)Limited
 Celecoxib↓collagen A, ↓fibronectin, ↓PDGF, and ↓TGF-β(I) Inhibits leiomyoma cell proliferation (Ke et al., 2013)(I) Increases the risk of myocardial infarction, stroke or heart failure (Solomon et al., 2005)
(II) No effect on myometrial cell proliferation (Ke et al., 2013)
 Tranilast↓COL1A1, ↓fibronectin ↓versican and ↓activin-A(I) Inhibits proliferation of myometrial and leiomyoma cells (Shime et al., 2002; Islam et al., 2012)(I) Well tolerated with low toxicity (Konneh, 1998)
 Pirfenidone↓collagen type I, ↓collagen type III(I) Inhibits proliferation of myometrial and leiomyoma cells (Lee et al., 1998)(I) Rather well tolerated by patients with idiopathic pulmonary fibrosis (Chaudhuri et al., 2014)
(II) Gastrointestinal adverse effects (Chaudhuri et al., 2014)
 Halofuginone↓collagen type I (a1), ↓collagen type III (a1) and ↓TGF-β1(I) Reduces uterine leiomyoma volume in a mouse xenograft model (Koohestani et al., 2016)(I) Appears to be safe and well tolerated (De Jonge et al., 2006)
(II) Inhibits both myometrial and leiomyoma cell proliferation (Grudzien et al., 2010)(II) No clinically adverse events (De Jonge et al., 2006)
(III) Induces apoptosis (Grudzien et al., 2010)
 Curcumin↓fibronectin(I) Inhibits leiomyoma cell proliferation (Malik et al., 2009)(I) Appears to be safe and well tolerated by patients (Gupta et al., 2013)
(II) Side effects-nausea, diarrhea, headache, rash and yellow stool (Gupta et al., 2013)
 Resveratrol↓fibronectin, ↓collagen types I, ↓collagen III, ↓fibromodulin. ↓biglycan, and ↓MMP-9, ↑TIMP2(I) Inhibits proliferation of human uterine leiomyoma cells (Catherino et al., 2011)(I) Appears safe and well tolerated by patients (Turner et al., 2015)
(II) Adverse events-nausea, diarrhea and weight loss (Turner et al., 2015)
 Collagenase C.  histolyticum↓ collagen(I) Reduces fibrosis (collagen) and tissue stiffness in uterine leiomyoma (Jayes et al., 2016)Limited
Treatment modalitiesFibrosis-related molecular targetsEffects on leiomyoma and related symptomsSafety and side effects
Current treatments
 Leuprolide  acetate↓TGF-β1, ↓TGF-β3, ↓TGF-βR type I, ↓TGF-βR type II, ↓Smad4, ↓pSmad3, ↑Smad7, ↓ collagen type I, ↓↑fibronectin, ↓versican, ↓fibromodulin, ↓TIMP-1, ↑MMP-1, ↑MMP-2, ↑MMP-3, and ↑MMP-9(I) Reduces uterine and leiomyoma volume (Stovall et al., 1995)(I) Prolonged therapy (>6 months) is not recommended
(II) Alleviates bleeding and increase hemoglobin levels (Stovall et al., 1991)(II) Side effects-hot flashes, vaginitis and bone loss (Stovall et al., 1995)
 Cetrorelix  acetate↓COL1A1, ↓fibronectin, and ↓versican variant V0(I) Decreases uterine and leiomyoma volume (Gonzalez-Barcena et al., 1997; Felberbaum et al., 2001; Engel et al., 2007)(I) Side effects-amenorrhea and hot flashes (Gonzalez-Barcena et al., 1997)
(II) Abolishes symptoms of menorrhagia and reduce uterine pain (Engel et al., 2007)
 Asoprisnil↓TGF-β3, ↓TGF-βR type II, ↑EMMPRIN, ↑MMP-1, ↑MT1-MMP, ↓TIMP-1, ↓TIMP-2, ↓ collagen type I and ↓collagen type III(I) Suppress leiomyoma and uterine volume (Chwalisz et al., 2007)(I) Abnormal endometrial changes (Williams et al., 2007)
(II) Reduces uterine bleeding as well as bloating and pelvic pressure (Chwalisz et al., 2007)(II) Side effects-hot flashes or night sweats (Chwalisz et al., 2007)
 Ulipristal  acetate↑MMP-1, ↑MMP-2, ↑MMP-3, ↑MMP-8, ↑MMP-9, ↑EMMPRIN, ↓TIMP-1, ↓TIMP-2, ↓ collagen type I, ↓collagen type III and ↓fibronectin(I) Reduces leiomyoma and uterine volume (Donnez et al., 2012a, 2012b., 2015)(I) Quite well tolerated (Donnez et al., 2015).
(II) Controls bleeding and pain (Donnez et al., 2015)(II) Side effects-hot flashes and headaches (Donnez et al., 2015)
(III) Improves quality of life (Donnez et al., 2012a, 2012b., 2015)
 Mifepristone↓COL1A1, ↓fibronectin, ↓versican and ↓dermatopontin(I) Reduces uterine and leiomyoma volume (Esteve et al., 2012)(I) No endometrial hyperplasia or cellular atypia(Yerushalmi et al., 2014)
(II) Alleviates hypermenorrhea, menstrual blood loss, pelvic painandpressure, anemia and dysmenorrhea (Shen et al., 2013; Yerushalmi et al., 2014)(II) Side effects-hot flashes, nausea, weakness, abdominal pain and vaginal discharge (Yerushalmi et al., 2014)
(III) Reduces leiomyoma cell viability and proliferation (Chung et al., 2014)
 Raloxifene↓Collagen and ↓MMP-2(I) prevents progression of uterine leiomyoma (Jirecek et al., 2004)(I) quite well tolerated (Jirecek et al., 2004)
(II) side effects-hot flashes and leg cramps (Martino et al., 2005), and risk of venous thromboembolism (Ettinger et al., 1999)
Investigational compounds
 CP8947↓COL1A1 and ↓COL7A1(I) Inhibits the leiomyoma cell proliferation (Catherino et al., 2010)Limited
(II) No effect on myometrial cell proliferation (Catherino et al., 2010)
 2-methoxyestradiol↓Collagen type I, ↓Collagen type III, ↓PAI-1, ↓CTGF, ↓α-SMA, ↓pSmad2/3 and ↓PI3K/Akt/mTOR(I) Inhibits cell proliferation in rat and human leiomyoma cells (Salama et al., 2006)(I) Well tolerated (Harrison et al., 2011)
(II) induces apoptosis in rat and human leiomyoma cells(Salama et al., 2006)(II) Side effects-hot flashes, muscle cramps, headache, fatigue or weakness, nausea, vomiting, diarrhea, gastrointestinal hemorrhage and hyponatremia (Rajkumar et al., 2007)
 Liarozole↓COL1A1, ↓COL4A2, ↓versican, ↓fibromodulin, ↓fibronectin and ↓TGF-β3(I) Inhibits proliferation of both myometrial and leiomyoma cells (Gilden et al., 2012)(I) Well tolerated (Denis et al., 1998)
(II) Side effects-skin disorders and dryness of mouth/eyes/lips (Goss et al., 2000)
 All-trans retinoic  acid↓Collagen 1, ↓collagen 4, ↓fibronectin, ↓versican and ↓TGF-β3(I) Downregulates immortalized leiomyoma cell proliferation (Malik et al., 2008)(I) Quite well tolerated (Kurzrock et al., 1993; Böcher et al., 2008)
(II) Side effects-transient headache, dry skin and mucosa, nausea and vomiting, myalgias or muscle pain, dyspnea (shortness of breath), and sensorineural hearing loss (Kurzrock et al., 1993; Böcher et al., 2008)
 Vitamin D↓fibronectin, ↓collagen type 1, ↓PAI-1, ↓pSmad2, ↓Wnt4, ↓β-catenin, ↓mTOR, ↓fibromodulin, ↓biglycan and ↓versican(I) Shrinks uterine leiomyoma tumors in Eker rat model (Halder et al., 2012)Limited
 Celecoxib↓collagen A, ↓fibronectin, ↓PDGF, and ↓TGF-β(I) Inhibits leiomyoma cell proliferation (Ke et al., 2013)(I) Increases the risk of myocardial infarction, stroke or heart failure (Solomon et al., 2005)
(II) No effect on myometrial cell proliferation (Ke et al., 2013)
 Tranilast↓COL1A1, ↓fibronectin ↓versican and ↓activin-A(I) Inhibits proliferation of myometrial and leiomyoma cells (Shime et al., 2002; Islam et al., 2012)(I) Well tolerated with low toxicity (Konneh, 1998)
 Pirfenidone↓collagen type I, ↓collagen type III(I) Inhibits proliferation of myometrial and leiomyoma cells (Lee et al., 1998)(I) Rather well tolerated by patients with idiopathic pulmonary fibrosis (Chaudhuri et al., 2014)
(II) Gastrointestinal adverse effects (Chaudhuri et al., 2014)
 Halofuginone↓collagen type I (a1), ↓collagen type III (a1) and ↓TGF-β1(I) Reduces uterine leiomyoma volume in a mouse xenograft model (Koohestani et al., 2016)(I) Appears to be safe and well tolerated (De Jonge et al., 2006)
(II) Inhibits both myometrial and leiomyoma cell proliferation (Grudzien et al., 2010)(II) No clinically adverse events (De Jonge et al., 2006)
(III) Induces apoptosis (Grudzien et al., 2010)
 Curcumin↓fibronectin(I) Inhibits leiomyoma cell proliferation (Malik et al., 2009)(I) Appears to be safe and well tolerated by patients (Gupta et al., 2013)
(II) Side effects-nausea, diarrhea, headache, rash and yellow stool (Gupta et al., 2013)
 Resveratrol↓fibronectin, ↓collagen types I, ↓collagen III, ↓fibromodulin. ↓biglycan, and ↓MMP-9, ↑TIMP2(I) Inhibits proliferation of human uterine leiomyoma cells (Catherino et al., 2011)(I) Appears safe and well tolerated by patients (Turner et al., 2015)
(II) Adverse events-nausea, diarrhea and weight loss (Turner et al., 2015)
 Collagenase C.  histolyticum↓ collagen(I) Reduces fibrosis (collagen) and tissue stiffness in uterine leiomyoma (Jayes et al., 2016)Limited

Current treatments

Leuprolide acetate

Leuprolide acetate is a synthetic analog of GnRH. GnRH is produced in the hypothalamus of the brain and after its release travels to the pituitary gland where it stimulates the production of LH and follicle stimulating hormone (FSH). Through peripheral circulation, LH and FSH travel to the ovaries where they stimulate the production of estrogen. Estrogen maintains its own levels within an appropriate range by acting as a negative feedback regulator of GnRH, LH and FSH production. Leuprolide acetate acts by producing an initial stimulation of FSH and LH as well as estrogen but after a few weeks, levels of LH and FSH drop because the pituitary gland stops responding to GnRH and leuprolide. This induces a state of hypoestrogenism, which has been used for the treatment of leiomyoma (Stovall et al., 1991). Leuprolide acetate is able to alleviate bleeding and increase hemoglobin levels (Stovall et al., 1991) as well as reduce uterine and leiomyoma volume (Stovall et al., 1995). However, this treatment is associated with some side effect related to hypoestrogenism, such as hot flashes, vaginitis and bone loss, which constitute the major limitation of long-term use (Stovall et al., 1995). The therapeutic effects of leuprolide acetate on leiomyoma are mediated by regulation of TGF-β receptor signaling and substantial tissue remodeling (Dou et al., 1997; Chegini et al., 2003; Ding et al., 2004b). Leuprolide acetate was reported to inhibit TGF-β1, TGF-β3, TGF-βR type I and type II as well as Smad4 and pSmad3 levels while increasing Smad7 expression in both myometrium and leiomyoma relative to untreated controls (Dou et al., 1996; Chegini et al., 2003). Leuprolide acetate also inhibited fibronectin mRNA expression in myometrial cells with a moderate increase in its expression in leiomyoma cells while also inhibiting type I collagen expression in both myometrial and leiomyoma cells (Ding et al., 2004b). A recent study reported that 3D leiomyoma cultures exposed to estrogen and progesterone demonstrated an increased expression of collagen-1, fibronectin and versican and this effect was inhibited by leuprolide acetate (Malik et al., 2016). TGF-β1 increased the expression of fibromodulin in myometrial cells, whereas leuprolide acetate inhibited this effect in both myometrial and leiomyoma cells (Levens et al., 2005). Dou et al. reported that leuprolide acetate treatment also induced a significant decrease in TIMP-1, and an increase in MMP-1, MMP-2, MMP-3 and MMP-9 mRNA expression in both leiomyoma and myometrium compared with untreated groups (Dou et al., 1997).

Cetrorelix acetate

Cetrorelix acetate, a GnRH antagonist, has been reported to decrease mean uterine and leiomyoma volume in pre-menopausal women (Gonzalez-Barcena et al., 1997; Felberbaum et al., 2001; Engel et al., 2007). This compound can abolish symptoms of menorrhagia and reduce uterine pain (Engel et al., 2007). However, cetrorelix acetate is associated with amenorrhea and hot flashes (Gonzalez-Barcena et al., 1997) and its therapeutic use is also limited by its prohibitive cost and requirement for daily injections (Kashani et al., 2016). Britten et al. investigated the effect of cetrorelix acetate on ECM production in human uterine leiomyoma and patient-matched myometrial cells (Britten et al., 2012). They found that cetrorelix decreased mRNA and protein expression of COL1A1, fibronectin and versican variant V0 in a time dependent manner in leiomyoma cells compared to untreated cells (Britten et al., 2012). In 3D leiomyoma cultures, cetrorelix also decreased estrogen- and progesterone-induced expression of collagen-1, fibronectin and versican (Malik et al., 2016).

Asoprisnil

Asoprisnil is a selective progesterone receptor modulator that shows a high degree of receptor and tissue selectivity. It has high-binding affinity for progesterone receptors, no binding affinity for estrogen or mineralocorticoid receptors, moderate affinity for glucocorticoid receptors and low affinity for androgen receptors. Asoprisnil has been reported to suppress leiomyoma and uterine volume as well as uterine bleeding and was associated with significant reduction in bloating and pelvic pressure (Chwalisz et al., 2007). The safety profile indicates that asoprisnil treatment is associated with endometrial changes including abnormal vascular growth (Williams et al., 2007). The adverse events, such as hot flashes or night sweats, were also detected but they were mild or moderate in severity, because asoprisnil does not induce a hypoestrogenic state (Chwalisz et al., 2007; Kashani et al., 2016). Asoprisnil exerts therapeutic effects on leiomyoma by regulating growth factor expression as well as ECM turnover and tissue remodeling (Wang et al., 2006; Morikawa et al., 2008). Treatment of leiomyoma cells with asoprisnil caused a decrease in the levels of TGF-β3 mRNA and protein and phosphorylated TGF-βR type II receptor compared to untreated controls (Wang et al., 2006). Morikawa and co-investigators reported that asoprisnil treatment also significantly increased levels of EMMPRIN, MMP-1 and MT1-MMP (membrane type 1-MMP) while decreasing levels of TIMP-1, TIMP-2, collagen type I and type III in cultured leiomyoma cells compared to untreated control cells (Morikawa et al., 2008). This group also reported that asoprisnil had no effect on protein contents of ECM and ECM-remodeling enzymes in myometrial cells (Morikawa et al., 2008).

Ulipristal acetate

Ulipristal acetate (also known as CDB-2914) is a selective progesterone receptor modulator that binds to progesterone receptors A and B with high affinity. Use of this drug has been approved in Europe and Canada for preoperative leiomyoma treatment (Melis et al., 2012). Ulipristal acetate is able to reduce leiomyoma and uterine volume and improves leiomyoma-related symptoms such as bleeding and pain as well as quality of life (Donnez et al., 2012a, 2012b, 2015). While this treatment is associated with some adverse events, such as hot flashes and headaches, and breast tenderness, these occurred in ≤10% of patients as circulating estradiol levels are maintained in the mid-follicular range throughout the treatment duration (Kashani et al., 2016). Moreover, it may be associated with physiologic endometrial changes (PAECs) that consist of benign cystic glandular dilation but these have not been associated to date with an increased risk of endometrial hyperplasia or malignancy (Donnez et al., 2015). Ulipristal acetate exerts therapeutic effects on leiomyoma growth by regulating the fibrotic process and its use results in lower ECM volume and higher MMP-2 expression in women with symptomatic leiomyoma compared to untreated leiomyoma (Courtoy et al., 2015). In cultured leiomyoma cells, ulipristal acetate significantly increased EMMPRIN, MMP-1 and MMP-8 protein contents as well as MMP-1, MMP-2, MMP-3 and MMP-9 mRNA levels, while decreasing mRNA and protein levels of TIMP-1 and TIMP-2 as well as collagen types I and III content, without comparable effects on cultured normal myometrial cells (Xu et al., 2008). Recently, we also demonstrated that ulipristal acetate can block activin-A induction of fibronectin mRNA expression in myometrial and leiomyoma cultured cells (Ciarmela et al., 2014).

Mifepristone

Mifepristone (also known as RU-486, mifegyne, mifeprex) is a synthetic steroid with antiprogesterone and antiglucocorticoid activity. It binds progesterone receptors and competitively antagonizes progesterone binding and signaling. Several clinical studies have demonstrated that mifepristone can reduce uterine and leiomyoma volume as well as alleviate leiomyoma-related symptoms including hypermenorrhea, menstrual blood loss, pelvic pain or pressure, anemia and dysmenorrhea (Esteve et al., 2012, 2013; Shen et al., 2013; Yerushalmi et al., 2014). A recent study reported that no endometrial hyperplasia or cellular atypia was observed after vaginal mifepristone treatment (Yerushalmi et al., 2014). However, some side effects, including hot flashes (10.3%), nausea (6.9%), weakness (6.9%), abdominal pain (24.1%) and vaginal discharge (20.7%), were observed at some point during the course of the 3 month study (Yerushalmi et al., 2014). Moreover, the increased risk of developing endometrial hyperplasia (28%) (10/36) during treatment with mifepristone raised safety concerns about its use in the management of leiomyoma (Steinauer et al., 2004; Guo and Segars, 2012). Antifibrotic effects of mifepristone have recently been demonstrated by Patel et al. (2016). This group reported that the progesterone agonist R5020 directly stimulated production of several ECM components including COL1A1, fibronectin, versican and dermatopontin in human leiomyoma cells compared to untreated cells and this effect was inhibited by mifepristone (Patel et al., 2016). Additionally, mifepristone can significantly reduce leiomyoma cell viability and proliferation (Chung et al., 2014) at least partly by downregulation of LAT2 (L-type amino acid transporter 2) mRNA expression (Luo et al., 2009). Furthermore, Yin et al. reported that mifepristone robustly up-regulated mRNA and protein levels of the known tumor suppressor KLF11 in leiomyoma smooth muscle cells (Yin et al., 2010).

Raloxifene

Raloxifene is a selective estrogen receptor modulator that interacts with estrogen receptors to elicit tissue-specific responses. It inhibits the growth of uterine leiomyoma in pre-menopausal women (Jirecek et al., 2004). Raloxifene is quite well tolerated (Jirecek et al., 2004) but has been associated with the risk of venous thromboembolism (Ettinger et al., 1999) as well as hot flashes and leg cramps (Martino et al., 2005). Raloxifene appears to inhibit collagen biosynthesis in leiomyoma cells while only slightly affecting collagen biosynthesis in control myometrial cells (Zbucka et al., 2008). Raloxifene was also found to inhibit MMP-2 in leiomyoma as well as in control myometrial cells (Zbucka et al., 2008).

Investigational compounds

CP8947

CP8947 is a novel non-steroidal selective progesterone receptor modulator derived from Penicillium oblatum. It has high selectivity for progesterone receptors and lacks affinity for estrogen receptor-α, androgen receptor and glucocorticoid receptor. Catherino et al. investigated the effect of CP8947 on cell proliferation and ECM components in leiomyoma cells (Catherino et al., 2010). They found that CP8947 was effective in inhibiting leiomyoma cell proliferation without disrupting myometrial cell proliferation and that it also decreased mRNA expression of COL1A1 and COL7A1 (Catherino et al., 2010).

2-Methoxyestradiol

2-Methoxyestradiol is a naturally occurring estradiol metabolite with low affinity for estrogen receptors. 2-Methoxyestradiol is currently being evaluated in ongoing advanced phases of clinical trials in patients with multiple myeloma, glioblastoma, ovarian cancer, metastatic renal cell carcinoma and prostate cancer. It is well tolerated by patients but associated with some adverse events such as hot flashes, muscle cramps, headache, fatigue or weakness, nausea, vomiting, diarrhea, gastrointestinal hemorrhage and hyponatremia (Rajkumar et al., 2007; Harrison et al., 2011). 2-Methoxyestradiol has been reported to induce apoptosis as well as inhibit cell proliferation and collagen production in rat and human leiomyoma cells (Salama et al., 2006). Salama et al. investigated the antifibrotic effect of 2­-methoxyestradiol on TGF-β3 mediated fibrosis-related factors in leiomyoma cells (Salama et al., 2012). They found that 2-methoxyestradiol abrogated TGF-β3-induced expression of collagen types I and III, PAI-1, CTGF and α-SMA in immortalized human uterine leiomyoma smooth muscle cells compared to untreated controls (Salama et al., 2012). 2-Methoxyestradiol also inhibited TGF-β3-induced activation of the PI3K/AKT/mTOR pathway as well as ameliorated TGF-β3-induced phosphorylation and nuclear translocation of Smad2/3 in this cell type (Salama et al., 2012). It has been shown that relatively high concentrations of 2-methoxyestradiol can also induce spindle aberrations in oocytes (Eichenlaub-Ritter et al., 2007).

Liarozole

Liarozole inhibits the cytochrome P450 (CYP)-dependent catabolism of retinoic acid and thereby increases intracellular retinoic acid levels. Liarozole is well tolerated in clinical studies and has been studied as treatment for prostate cancer (Denis et al., 1998) and breast cancer (Goss et al., 2000). The adverse events were predominantly dermatological including skin disorders (88%) and dryness of mouth/eyes/lips (69%) (Goss et al., 2000). The antifibrotic effects of liarozole have been studied in leiomyoma cells (Gilden et al., 2012; Levy et al., 2014). Gilden and co-investigators reported that liarozole can inhibit proliferation of both myometrial and leiomyoma cells at suprapharmacologic concentrations and also decrease mRNA and protein expression of COL1A1, COL4A2, versican, fibromodulin and fibronectin in a dose-dependent manner in leiomyoma cells compared with myometrial cells (Gilden et al., 2012). Interestingly, they found no statistically significant alteration in ECM regulation in liarozole treated myometrial cells (Gilden et al., 2012). A recent study reported that liarozole can inhibit TGF-β3 and TGF-β3 induction of ECM components, including versican, COL1A1 and fibronectin in human three-dimensional leiomyoma cultures (Levy et al., 2014).

All-trans retinoic acid

All-trans retinoic acid (also known as tretinoin) is a derivative of vitamin A that functions as a ligand for the retinoic acid receptor (RAR). It is quite well tolerated by patients but has been associated with common side effects including transient headache, dry skin and mucosa, nausea and vomiting, myalgias or muscle pain, dyspnea (shortness of breath) and sensorineural hearing loss (Kurzrock et al., 1993; Böcher et al., 2008). Human uterine smooth muscle cells express retinoic acid receptors RAR α, β and γ and retinoid X receptors RXR α and β as well as all-trans retinoic acid (Boettger-Tong et al., 1997). Treatment of immortalized leiomyoma cells with all-trans retinoic acid was reported to downregulate cell proliferation as well as protein production of ECM components including collagen 1, collagen 4, fibronectin and versican as well as mRNA expression of TGF-β3 compared to untreated controls (Malik et al., 2008).

Vitamin D

Vitamin D is a fat-soluble vitamin found in many foods including fish, eggs, fortified milk and cod liver oil. Its major physiologically relevant forms are D2 (ergocalciferol) and D3 (cholecalciferol). Several recent in vitro and in vivo studies implicate vitamin D insufficiency as an important contributor to the development of uterine leiomyoma (Halder et al., 2012; Baird et al., 2013; Paffoni et al., 2013). Women with sufficient vitamin D have been reported to have an estimated 32% lower odds of leiomyoma compared to women with vitamin D insufficiency (Baird et al., 2013). Similarly, Paffoniand et al. reported that women with a vitamin D deficiency experienced 2.4 times more leiomyoma compared to women with an adequate level of vitamin D (Paffoni et al., 2013). Using the Eker rat model, Halder and co-investigators found that vitamin D3 [1,25(OH)2D3] treatment was able to shrink uterine leiomyoma tumors (Halder et al., 2012). The therapeutic effect of vitamin D3 on leiomyoma was mediated, at least in part, by regulation of TGF-β responsive genes (Halder et al., 2011, 2013), as well as Wnt/β-catenin and mTOR signaling pathways (Al-Hendy et al., 2016). Vitamin D3 reduced TGF-β3-induced expression of fibronectin, collagen type 1 and PAI-1 protein in human uterine leiomyoma cells (Halder et al., 2011). Vitamin D3 also reduced TGF-β3-induced Smad2 phosphorylation as well as Smad2 and Smad3 nuclear translocation in human uterine leiomyoma cells (Halder et al., 2011). Al-Hendy et al. reported that vitamin D3 can reduce the levels of Wnt4 and β-catenin as well as mTOR in both immortalized and primary human uterine leiomyoma cells (Al-Hendy et al., 2016). Furthermore, vitamin D3 was reported to reduce mRNA and protein levels of fibromodulin, biglycan and versican in human uterine leiomyoma cells (Halder et al., 2013). The ability of vitamin D3 to downregulate ECM (fibronectin and collagen type 1) expression as well as Wnt4/β-catenin and cell proliferation was further confirmed by the observation that silencing expression of the vitamin D receptor (VDR) gene in normal myometrial cells increased ECM production as well as Wnt4/β-catenin and cell proliferation (Al-Hendy et al., 2016). Initial results regarding the opportunity for Vitamin D3 supplementation in women with leiomyoma are encouraging, but they must be confirmed by further studies (Ciavattini et al., 2016).

Celecoxib

Celecoxib is an inhibitor of cyclooxygenase 2 (COX-2) that is commonly used to manage pain or inflammation. The function of COX-2 is to convert arachidonic acid into prostaglandin H2, a common substrate for specific prostaglandin synthases. Uterine leiomyoma cells have higher levels of COX-2 mRNA and protein than myometrial cells (Ke et al., 2013). A recent study reported that celecoxib can significantly inhibit uterine leiomyoma cell proliferation without affecting proliferation of healthy myometrial smooth muscle cells (Ke et al., 2013). Celcoxib can also reduce expression of collagen A, fibronectin, PDGF and TGF-β at the mRNA level in uterine leiomyoma cells compared to untreated controls (Park et al., 2014). Although initial results are encouraging, the potential health risks may hinder the use of celecoxib for the treatment uterine leiomyoma. Clinical evidence indicates that celecoxib use is associated with increased risk of cardiovascular events, myocardial infarction, stroke and heart failure (Solomon et al., 2005).

Tranilast

Tranilast is an orally administered synthetic drug of low toxicity used for the treatment of allergic disorders in Japan and South Korea. It is well tolerated by patients (Konneh, 1998) supporting the possibility for its use to treat uterine leiomyoma. The antifibrotic effects of tranilast have been reported in myometrial and leiomyoma cells (Islam et al., 2014b). Recently, we demonstrated that tranilast can decrease expression of COL1A1 and fibronectin at mRNA and protein levels, as well as versican and activin-A at mRNA levels in myometrial and leiomyoma cells (Islam et al., 2014b). Additionally, tranilast can also inhibit the proliferation of uterine myometrial and leiomyoma cells (Shime et al., 2002; Islam et al., 2012).

Pirfenidone

Pirfenidone is a synthetic pyridone compound used for the treatment of idiopathic pulmonary fibrosis. It is rather well tolerated by patients, with adverse effects that are predominantly gastrointestinal (Chaudhuri et al., 2014). The antifibrotic potential of pirfenidone has been studied in leiomyoma cells by Lee et al. (1998). They reported that pirfenidone was effective in regulating proliferation of myometrial and leiomyoma cells in vitro as well as in reducing the mRNA level of collagen types I and III in a dose-dependent manner (Lee et al., 1998).

Halofuginone

Halofuginone, an analog of febrifugine, is a small alkaloid isolated from the plant Dichroa febrifuga. It has been used as a coccidiostat (an antiprotozoal agent) in chickens since the 1960s. Phase I/II clinical trials of halofuginone have been completed in patients with progressive advanced solid tumors and HIV-related Kaposi's sarcoma. Halofuginone at dose 0.5 mg/d appears to be safe and well tolerated, with no clinically adverse events (De Jonge et al., 2006). The antifibrotic effect of halofuginone on uterine leiomyoma has been reported (Grudzien et al., 2010). Grudzien et al. reported that halofuginone can significantly reduce collagen type I (a1), collagen type III (a1) and TGF-β1 mRNA levels in leiomyoma and myometrial cells compared to corresponding untreated cells (Grudzien et al., 2010). Halofuginone can also inhibit both myometrial and leiomyoma cell proliferation by inducing apoptosis and inhibiting DNA synthesis in a dose-dependent manner (Grudzien et al., 2010). A recent study by Koohestani et al. reported that halofuginone was effective in reducing uterine leiomyoma volume in a mouse xenograft model (Koohestani et al., 2016). This group found a 35–40% reduction in leiomyoma/tumor volume in mice carrying human uterine leiomyoma xenografts after treatment with halofuginone compared to control groups. The halofuginone-induced reduction of tumor volume was accompanied by decreased cell proliferation and increased apoptosis (Koohestani et al., 2016). However, these authors found no significant difference in the content of collagen between helofuginone-treated mice carrying human uterine leiomyoma xenografts and control groups (Koohestani et al., 2016).

Curcumin

Curcumin is a polyphenol derived from rhizome of turmeric (Curcuma longa). Turmeric is commonly used in Asian foods. It has shown beneficial effects in a plethora of human diseases. Extensive clinical trials indicate that curcumin is safe and well tolerated by patients. However, this treatment is associated with some undesired adverse effects such as nausea, diarrhea, headache, rash and yellow stool (Gupta et al., 2013). Antifibrotic effects of curcumin on human uterine leiomyoma cells have been demonstrated by Malik et al. (2009). This group reported that curcumin can inhibit mRNA expression of fibronectin in leiomyoma cells compared to untreated controls (Malik et al., 2009). They also found that curcumin can inhibit proliferation of leiomyoma cells without affecting patient-matched myometrial cells (Malik et al., 2009). These effects were mediated by inducing caspase-3 and caspase-9 protein expression as well as inhibiting ERK 1/2 and NF-κB protein expression (Malik et al., 2009).

Resveratrol

Resveratrol is a polyphenolic compound found in peanuts, grapes and some berries and is produced by plants in response to environmental stress, pathogen infection and ultraviolet radiation. Resveratrol is safe and well tolerated by patients, with common adverse events including nausea, diarrhea and weight loss (Turner et al., 2015). The ability of resveratrol to interfere with ECM formation and deposition in multiple diseases has recently been discussed by Agarwal and Agarwal (2017). Resveratrol appears to reduce expression of fibronectin, collagen types I and III as well as fibromodulin and biglycan at mRNA and/or protein levels in ELT-3 cells and/or human uterine leiomyoma smooth muscle cells compared to corresponding untreated cells (Catherino et al., 2011; Wu et al., 2016). Resveratrol also reduced MMP-9 protein expression while increasing TIMP2 protein expression in ELT-3 cells and healthy uterine smooth muscle cells compared to corresponding untreated cells (Wu et al., 2016). Furthermore, resveratrol was reported to inhibit proliferation and induce apoptosis and cell cycle arrest in human uterine leiomyoma cells compared to untreated controls (Catherino et al., 2011).

Collagenase C. histolyticum

Collagenase C.histolyticum (CCH) is a bacterial enzyme that breaks down collagen. CCH has been approved by the US FDA for the treatment of Dupuytren's contracture (a thickening of the fibrous tissue layer underneath the skin of the palm and fingers) and Peyronie's disease (a connective tissue disorder of the penis). A recent study by Jayes et al. investigated whether CCH was effective in the digestion of interstitial collagen in uterine leiomyoma (Jayes et al., 2016). They found 37–77% fibrosis in untreated leiomyoma, indicating the collagen-rich nature of these tumors (Jayes et al., 2016). A reduced amount of fibrosis ranging from 5.3 to 2.4% was recorded after treatment with CCH. Furthermore, complete digestion of collagen fibrils was confirmed by transmission electron microscopy. Tissue stiffness was also reduced with CCH treatment (Jayes et al., 2016). The above results suggest that CCH may reduce leiomyoma size and bulk symptoms possibly through collagen digestion and by modulating mechanotransduction process. Clinical trials are necessary to evaluate the safety and efficacy of CCH.

Conclusions and future perspectives

Uterine leiomyoma expresses a wide variety of ECM components, including collagens, fibronectin, laminins, proteoglycans and integrins as well as MMPs and TIMPs. ECM proteins can induce mechanotransduction, thus activating pleiotropic intracellular signaling cascades such as the integrin-Rho/p38 MAPK/ERK pathways.

ECM accumulation is regulated by growth factors, cytokines and steroid hormones. Among growth factors and cytokines, TGF-β, activin-A, PDGF and TNF-α are able to increase the synthesis of ECM components through activation of multiple signaling pathways (e.g. Smad 2/3 and MEK/ERK). In addition, estrogen and progesterone increase ECM production by regulating the expression and activity of growth factors (TGF-β and IGF), again leading to stimulation of several signaling pathways (MEK/ERK, AKT and PLCγ).

ECM may act as a reservoir of growth factors and protect them from degradation in the ECM microenvironment. ECM proteins and their receptors (integrins) can interact with growth factors independently or synergistically. MMPs can degrade ECM proteins and GAGs of proteoglycans, thereby inducing local release of soluble growth factors from their insoluble state.

Uterine leiomyoma is thought to be an inflammatory/fibrotic disorder, and possibly myofibroblasts are thought to play key role in the process of fibrosis. Reproductive events (mainly menstruation), infection, mechanical stress, injury, oxidative stress or hypoxia may act as the initiator of inflammation in the uterus. In response to inflammation, myofibroblast cells produce ECM to promote necessary repair and tissue homeostasis, but deregulation of normal myofibroblast function may lead to fibrosis. The differentiation of myofibroblasts is regulated by TGF-β and activin-A and estrogen.

The above findings support the fact that ECM is a major molecular switch that can be considered to be a crucial therapeutic target to control abnormal growth of leiomyoma. Currently, several types of drugs are available for leiomyoma treatment but none of them has been introduced specifically as antifibrotic agents (Table II). Therefore, the introduction of effective antifibrotic drugs which target ECM components directly or interfere with their expression and deposition has the potential to be an effective solution for the management of uterine leiomyoma. The antifibrotic compounds can be introduced based on their ability to regulate ECM components and ECM receptors as well as growth factors, cytokines, steroid hormones and their corresponding receptors and intracellular signaling pathways, as well as miRNAs, involved in ECM production in leiomyoma. Since ECM is involved in both pathological and physiological processes, therefore, it will be an important challenge to identify compounds that specifically target unwanted fibrosis and optimize their use in interrupting the fibrotic process without disturbing the normal physiological environment.

Authors' roles

M.S.I. and P.C. conceived and designed the study. M.S.I. conducted the review of the literature and wrote the first draft of the manuscript. A.C., F.P., M.C. and P.C. supervised the work and corrected the manuscript. All authors contributed to analysis and interpretation of the data, critically revised and approved the manuscript.

Funding

The ‘Fondazione Cassa di Risparmio di Fabriano e Cupramontana’ (to M.C. and P.C.).

Conflict of interest

The authors have nothing to declare.

References

Abreu
JG
,
Coffinier
C
,
Larraı́n
J
,
Oelgeschläger
M
,
De Robertis
EM
.
Chordin-like CR domains and the regulation of evolutionarily conserved extracellular signaling systems
.
Gene
2002
;
1
:
39
47
.

Agarwal
R
,
Agarwal
P
.
Targeting extracellular matrix remodeling in disease: could resveratrol be a potential candidate?
Exp Biol Med
2017
;
4
:
374
383
.

Al-Hendy
A
,
Diamond
MP
,
Boyer
TG
,
Halder
SK
.
Vitamin D3 inhibits Wnt/β-catenin and mTOR signaling pathways in human uterine fibroid cells
.
J Clin Endocrinol Metab
2016
;
4
:
1542
1551
.

Alam
N
,
Goel
HL
,
Zarif
MJ
,
Butterfield
JE
,
Perkins
HM
,
Sansoucy
BG
,
Sawyer
TK
,
Languino
LR
.
The integrin-growth factor receptor duet
.
J Cell Physiol
2007
;
3
:
649
653
.

Albig
AR
,
Neil
JR
,
Schiemann
WP
.
Fibulins 3 and 5 antagonize tumor angiogenesis in vivo
.
Cancer Res
2006
;
5
:
2621
2629
.

Andersson-Sjöland
A
,
Hallgren
O
,
Rolandsson
S
,
Weitoft
M
,
Tykesson
E
,
Larsson-Callerfelt
A-K
,
Rydell-Törmänen
K
,
Bjermer
L
,
Malmström
A
,
Karlsson
JC
.
Versican in inflammation and tissue remodelling: the impact on lung disorders
.
Glycobiology
2014
;
25
:
243
251
.

Anuar
N
,
Di Gregoli
K
,
Newby
A
,
Johnson
J
.
IL-3 induced MMP-3 expression directs angiogensis and neovascularisation in atherosclerosis
.
Atherosclerosis
2016
;
244
:
e4
e5
.

Arici
A
,
Sozen
I
.
Transforming growth factor-β3 is expressed at high levels in leiomyoma where it stimulates fibronectin expression and cell proliferation
.
Fertil Steril
2000
;
5
:
1006
1011
.

Assoian
RK
,
Schwartz
MA
.
Coordinate signaling by integrins and receptor tyrosine kinases in the regulation of G1 phase cell-cycle progression
.
Curr Opin Genet Dev
2001
;
1
:
48
53
.

Baird
DD
,
Hill
MC
,
Schectman
JM
,
Hollis
BW
.
Vitamin D and the risk of uterine fibroids
.
Epidemiology
2013
;
3
:
447
453
.

Barbarisi
A
,
Petillo
O
,
Di Lieto
A
,
Melone
MA
,
Margarucci
S
,
Cannas
M
,
Peluso
G
.
17-beta estradiol elicits an autocrine leiomyoma cell proliferation: evidence for a stimulation of protein kinase-dependent pathway
.
J Cell Physiol
2001
;
3
:
414
424
.

Barker
NM
,
Carrino
DA
,
Caplan
AI
,
Hurd
WW
,
Liu
JH
,
Tan
H
,
Mesiano
S
.
Proteoglycans in leiomyoma and normal myometrium abundance, steroid hormone control, and implications for pathophysiology
.
Reprod Sci
2015
;
3
:
302
309
.

Benassayag
C
,
Leroy
MJ
,
Rigourd
V
,
Robert
B
,
Honore
JC
,
Mignot
TM
,
Vacher-Lavenu
MC
,
Chapron
C
,
Ferre
F
.
Estrogen receptors (ERα/ERβ) in normal and pathological growth of the human myometrium: pregnancy and leiomyoma
.
Am J Physiol
1999
;
6
:
E1112
E1118
.

Bergers
G
,
Brekken
R
,
McMahon
G
,
Vu
TH
,
Itoh
T
,
Tamaki
K
,
Tanzawa
K
,
Thorpe
P
,
Itohara
S
,
Werb
Z
.
Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis
.
Nat Cell Biol
2000
;
10
:
737
744
.

Berto
AGA
,
Oba
SM
,
Michelacci
YM
,
Sampaio
LO
.
Galactosaminoglycans from normal myometrium and leiomyoma
.
Braz J Med Biol Res
2001
;
5
:
633
637
.

Bill
HM
,
Knudsen
B
,
Moores
SL
,
Muthuswamy
SK
,
Rao
VR
,
Brugge
JS
,
Miranti
CK
.
Epidermal growth factor receptor-dependent regulation of integrin-mediated signaling and cell cycle entry in epithelial cells
.
Mol Cell Biol
2004
;
19
:
8586
8599
.

Bissell
MJ
,
Aggeler
J
.
Dynamic reciprocity: how do extracellular matrix and hormones direct gene expression?
Prog Clin Biol Res
1986
;
249
:
251
262
.

Böcher
WO
,
Wallasch
C
,
Höhler
T
,
Galle
PR
.
All-trans retinoic acid for treatment of chronic hepatitis C
.
Liver Int
2008
;
3
:
347
354
.

Boettger-Tong
H
,
Shipley
G
,
Hsu
CJ
,
Stancel
GM
.
Cultured human uterine smooth muscle cells are retinoid responsive
.
Proc Soc Exp Biol Med
1997
;
1
:
59
65
.

Bogusiewicz
M
,
Stryjecka-Zimmer
M
,
Postawski
K
,
Jakimiuk
AJ
,
Rechberger
T
.
Activity of matrix metalloproteinase-2 and-9 and contents of their tissue inhibitors in uterine leiomyoma and corresponding myometrium
.
Gynecol Endocrinol
2007
;
9
:
541
546
.

Border
WA
,
Okuda
S
,
Languino
LR
,
Ruoslahti
E
.
Transforming growth factor-β regulates production of proteoglycans by mesangial cells
.
Kidney Int
1990
;
37
:
689
695
.

Boulday
G
,
Fitau
J
,
Coupel
S
,
Soulillou
JP
,
Charreau
B
.
Exogenous tissue inhibitor of metalloproteinase‐1 promotes endothelial cell survival through activation of the phosphatidylinositol 3‐kinase/Akt pathway
.
Ann N Y Acad Sci
2004
;
1
:
28
36
.

Brauer
R
,
Beck
IM
,
Roderfeld
M
,
Roeb
E
,
Sedlacek
R
.
Matrix metalloproteinase-19 inhibits growth of endothelial cells by generating angiostatin-like fragments from plasminogen
.
BMC Biochem
2011
;
1
:
38
.

Britten
JL
,
Malik
M
,
Levy
G
,
Mendoza
M
,
Catherino
WH
.
Gonadotropin-releasing hormone (GnRH) agonist leuprolide acetate and GnRH antagonist cetrorelix acetate directly inhibit leiomyoma extracellular matrix production
.
Fertil Steril
2012
;
5
:
1299
1307
.

Brown
LF
,
Detmar
M
,
Tognazzi
K
,
Abu-Jawdeh
G
,
Iruela-Arispe
ML
.
Uterine smooth muscle cells express functional receptors (flt-1 and KDR) for vascular permeability factor/vascular endothelial growth factor
.
Lab Invest
1997
;
2
:
245
255
.

Buttram
VC
Jr,
Reiter
RC
.
Uterine leiomyomata: etiology, symptomatology, and management
.
Fertil Steril
1981
;
4
:
433
445
.

Bützow
R
,
Fukushima
D
,
Twardzik
DR
,
Ruoslahti
E
.
A 60-kD protein mediates the binding of transforming growth factor-β to cell surface and extracellular matrix proteoglycans
.
J Cell Biol
1993
;
3
:
721
727
.

Cabrera
S
,
Gaxiola
M
,
Arreola
JL
,
Ramírez
R
,
Jara
P
,
D’Armiento
J
,
Richards
T
,
Selman
M
,
Pardo
A
.
Overexpression of MMP9 in macrophages attenuates pulmonary fibrosis induced by bleomycin
.
Int J Biochem Cell Biol
2007
;
12
:
2324
2338
.

Cardozo
ER
,
Clark
AD
,
Banks
NK
,
Henne
MB
,
Stegmann
BJ
,
Segars
JH
.
The estimated annual cost of uterine leiomyomata in the United States
.
Am J Obstet Gynecol
2012
;
3
:
211. e1
211. e9
.

Carrino
DA
,
Mesiano
S
,
Barker
NM
,
Hurd
WW
,
Caplan
AI
.
Proteoglycans of uterine fibroids and keloid scars: similarity in their proteoglycan composition
.
Biochem J
2012
;
2
:
361
368
.

Catherino
WH
,
Malik
M
,
Driggers
P
,
Chappel
S
,
Segars
J
,
Davis
J
.
Novel, orally active selective progesterone receptor modulator CP8947 inhibits leiomyoma cell proliferation without adversely affecting endometrium or myometrium
.
J Steroid Biochem Mol Biol
2010
;
4
:
279
286
.

Catherino
WH
,
Parrott
E
,
Segars
J
.
Proceedings from the national institute of child health and human development conference on the uterine fibroid research update workshop
.
Fertil Steril
2011
;
1
:
9
12
.

Cavin
S
,
Maric
D
,
Diviani
D
.
A-kinase anchoring protein-Lbc promotes pro-fibrotic signaling in cardiac fibroblasts
.
Biochim Biophys Acta
2014
;
2
:
335
345
.

Chakir
J
,
Shannon
J
,
Molet
S
,
Fukakusa
M
,
Elias
J
,
Laviolette
M
,
Boulet
L-P
,
Hamid
Q
.
Airway remodeling-associated mediators in moderate to severe asthma: effect of steroids on TGF-β, IL-11, IL-17, and type I and type III collagen expression
.
J Allergy Clin Immunol
2003
;
6
:
1293
1298
.

Chang
HL
,
Senaratne
TN
,
Zhang
LH
,
Szotek
PP
,
Stewart
E
,
Dombkowski
D
,
Preffer
F
,
Donahoe
PK
,
Teixeira
J
.
Uterine leiomyomas exhibit fewer stem/progenitor cell characteristics when compared with corresponding normal myometrium
.
Reprod Sci
2010
;
2
:
158
167
.

Chaudhuri
N
,
Duck
A
,
Frank
R
,
Holme
J
,
Leonard
C
.
Real world experiences: pirfenidone is well tolerated in patients with idiopathic pulmonary fibrosis
.
Respir Med
2014
;
1
:
224
226
.

Chegini
N
,
Luo
X
,
Ding
L
,
Ripley
D
.
The expression of Smads and transforming growth factor beta receptors in leiomyoma and myometrium and the effect of gonadotropin releasing hormone analogue therapy
.
Mol Cell Endocrinol
2003
;
1-2
:
9
16
.

Chegini
N
,
Ma
C
,
Tang
XM
,
Williams
RS
.
Effects of GnRH analogues, 'add-back' steroid therapy, antiestrogen and antiprogestins on leiomyoma and myometrial smooth muscle cell growth and transforming growth factor-β expression
.
Mol Hum Reprod
2002
;
12
:
1071
1078
.

Chegini
N
,
Tang
XM
,
Ma
C
.
Regulation of transforming growth factor-beta1 expression by granulocyte macrophage-colony-stimulating factor in leiomyoma and myometrial smooth muscle cells
.
J Clin Endocrinol Metab
1999
;
11
:
4138
4143
.

Chen
H-M
,
Lin
Y-H
,
Cheng
Y-M
,
Wing
L-YC
,
Tsai
S-J
.
Overexpression of integrin-β1 in leiomyoma promotes cell spreading and proliferation
.
J Clin Endocrinol Metab
2013
;
5
:
E837
E846
.

Chuang
T-D
,
Khorram
O
.
miR-200c regulates IL8 expression by targeting IKBKB: a potential mediator of inflammation in leiomyoma pathogenesis
.
PloS one
2014
;
4
:
e95370
.

Chuang
T-D
,
Khorram
O
.
Mechanisms underlying aberrant expression of miR-29c in uterine leiomyoma
.
Fertil Steril
2016
;
1
:
236
245. e1
.

Chuang
TD
,
Luo
X
,
Panda
H
,
Chegini
N
.
miR-93/106b and their host gene, MCM7, are differentially expressed in leiomyomas and functionally target F3 and IL-8
.
Mol Endocrinol
2012
a;
6
:
1028
1042
.

Chuang
TD
,
Panda
H
,
Luo
X
,
Chegini
N
.
miR-200c is aberrantly expressed in leiomyomas in an ethnic-dependent manner and targets ZEBs, VEGFA, TIMP2, and FBLN5
.
Endocr Relat Cancer
2012
b;
4
:
541
556
.

Chung
Y-J
,
Chae
B
,
Kwak
S-H
,
Song
J-Y
,
Lee
A-W
,
Jo
H-H
,
Lew
Y-O
,
Kim
J-H
,
Kim
M-R
.
Comparison of the inhibitory effect of gonadotropin releasing hormone (GnRH) agonist, selective estrogen receptor modulator (SERM), antiprogesterone on myoma cell proliferation in vitro
.
Int J Med Sci
2014
;
3
:
276
281
.

Chwalisz
K
,
Larsen
L
,
Mattia-Goldberg
C
,
Edmonds
A
,
Elger
W
,
Winkel
CA
.
A randomized, controlled trial of asoprisnil, a novel selective progesterone receptor modulator, in women with uterine leiomyomata
.
Fertil Steril
2007
;
6
:
1399
1412
.

Ciarmela
P
,
Bloise
E
,
Gray
PC
,
Carrarelli
P
,
Islam
MS
,
De Pascalis
F
,
Severi
FM
,
Vale
W
,
Castellucci
M
,
Petraglia
F
.
Activin-A and myostatin response and steroid regulation in human myometrium: disruption of their signalling in uterine fibroid
.
J Clin Endocrinol Metab
2011
a;
03
:
755
765
.

Ciarmela
P
,
Carrarelli
P
,
Islam
MS
,
Janjusevic
M
,
Zupi
E
,
Tosti
C
,
Castellucci
M
,
Petraglia
F
.
Ulipristal acetate modulates the expression and functions of activin A in leiomyoma cells
.
Reprod Sci
2014
;
9
:
1120
1125
.

Ciarmela
P
,
Islam
MS
,
Reis
FM
,
Gray
PC
,
Bloise
E
,
Petraglia
F
,
Vale
W
,
Castellucci
M
.
Growth factors and myometrium: biological effects in uterine fibroid and possible clinical implications
.
Hum Reprod Update
2011
b;
6
:
772
790
.

Ciavattini
A
,
Carpini
GD
,
Serri
M
,
Vignini
A
,
Sabbatinelli
J
,
Tozzi
A
,
Aggiusti
A
,
Clemente
N
.
Hypovitaminosis D and ‘small burden’ uterine fibroids: opportunity for a vitamin D supplementation
.
Medicine (Baltimore)
2016
;
52
:
e5698
.

Colnot
CL
,
Thompson
Z
,
Miclau
T
,
Werb
Z
,
Helms
JA
.
Altered fracture repair in the absence of MMP9
.
Development
2003
;
17
:
4123
4133
.

Courtoy
GE
,
Donnez
J
,
Marbaix
E
,
Dolmans
M-M
.
In vivo mechanisms of uterine myoma volume reduction with ulipristal acetate treatment
.
Fertil Steril
2015
;
2
:
426
434.e1
.

Cramer
SF
,
Patel
A
.
The frequency of uterine leiomyomas
.
Am J Clin Pathol
1990
;
4
:
435
438
.

Day Baird
D
,
Dunson
DB
,
Hill
MC
,
Cousins
D
,
Schectman
JM
.
High cumulative incidence of uterine leiomyoma in black and white women: ultrasound evidence
.
Am J Obstet Gynecol
2003
;
1
:
100
107
.

De Jonge
MJA
,
Dumez
H
,
Verweij
J
,
Yarkoni
S
,
Snyder
D
,
Lacombe
D
,
Marreaud
S
,
Yamaguchi
T
,
Punt
CJA
,
Van Oosterom
A
.
Phase I and pharmacokinetic study of halofuginone, an oral quinazolinone derivative in patients with advanced solid tumours
.
Eur J Cancer
2006
;
12
:
1768
1774
.

Denis
L
,
Debruyne
F
,
De Porre
P
,
Bruynseels
J
.
Early clinical experience with liarozole (Liazal™) in patients with progressive prostate cancer
.
Eur J Cancer
1998
;
4
:
469
475
.

Desmouliere
A
,
Chaponnier
C
,
Gabbiani
G
.
Tissue repair, contraction, and the myofibroblast
.
Wound Repair Regen
2005
;
1
:
7
12
.

Desmoulière
A
,
Geinoz
A
,
Gabbiani
F
,
Gabbiani
G
.
Transforming growth factor-β1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts
.
J Cell Biol
1993
;
1
:
103
111
.

Desmouliere
A
,
Redard
M
,
Darby
I
,
Gabbiani
G
.
Apoptosis mediates the decrease in cellularity during the transition between granulation tissue and scar
.
Am J Pathol
1995
;
1
:
56
66
.

Dimitrova
IK
,
Richer
JK
,
Rudolph
MC
,
Spoelstra
NS
,
Reno
EM
,
Medina
TM
,
Bradford
AP
.
Gene expression profiling of multiple leiomyomata uteri and matched normal tissue from a single patient
.
Fertil Steril
2009
;
6
:
2650
2663
.

Ding
L
,
Luo
X
,
Chegini
N
.
The expression of IL-13 and IL-15 in leiomyoma and myometrium and their influence on TGF-β and proteases expression in leiomyoma and myometrial smooth muscle cells and SKLM, leiomyosarcoma cell line
.
J Soc Gynecol Investig
2004
a;
11
:
319A
.

Ding
L
,
Xu
J
,
Luo
X
,
Chegini
N
.
Gonadotropin releasing hormone and transforming growth factor β activate mitogen-activated protein kinase/extracellularly regulated kinase and differentially regulate fibronectin, type I collagen, and plasminogen activator inhibitor-1 expression in leiomyoma and myometrial smooth muscle cells
.
J Clin Endocrinol Metab
2004
b;
11
:
5549
5557
.

Dixon
D
,
He
H
,
Haseman
JK
.
Immunohistochemical localization of growth factors and their receptors in uterine leiomyomas and matched myometrium
.
Environ Health Perspect
2000
;
108
:
795
802
.

Doerr
ME
,
Jones
JI
.
The roles of integrins and extracellular matrix proteins in the insulin-like growth factor I-stimulated chemotaxis of human breast cancer cells
.
J Biol Chem
1996
;
5
:
2443
2447
.

Donnez
J
,
Hudecek
R
,
Donnez
O
,
Matule
D
,
Arhendt
HJ
,
Zatik
J
,
Kasilovskiene
Z
,
Dumitrascu
MC
,
Fernandez
H
,
Barlow
DH
et al. .
Efficacy and safety of repeated use of ulipristal acetate in uterine fibroids
.
Fertil Steril
2015
;
2
:
519
527. e3
.

Donnez
J
,
Tatarchuk
TF
,
Bouchard
P
,
Puscasiu
L
,
Zakharenko
NF
,
Ivanova
T
,
Ugocsai
G
,
Mara
M
,
Jilla
MP
,
Bestel
E
.
Ulipristal acetate versus placebo for fibroid treatment before surgery
.
N Engl J Med
2012
a;
5
:
409
420
.

Donnez
J
,
Tomaszewski
J
,
Vázquez
F
,
Bouchard
P
,
Lemieszczuk
B
,
Baró
F
,
Nouri
K
,
Selvaggi
L
,
Sodowski
K
,
Bestel
E
.
Ulipristal acetate versus .euprolide acetate for uterine fibroids
.
N Engl J Med
2012
b;
5
:
421
432
.

Dou
Q
,
Tarnuzzer
RW
,
Williams
RS
,
Schultz
GS
,
Chegini
N
.
Differential expression of matrix metalloproteinases and their tissue inhibitors in leiomyomata: a mechanism for gonadotrophin releasing hormone agonist-induced tumour regression
.
Mol Hum Reprod
1997
;
11
:
1005
1014
.

Dou
Q
,
Zhao
Y
,
Tarnuzzer
RW
,
Rong
H
,
Williams
RS
,
Schultz
GS
,
Chegini
N
.
Suppression of transforming growth factor-beta (TGF beta) and TGF beta receptor messenger ribonucleic acid and protein expression in leiomyomata in women receiving gonadotropin-releasing hormone agonist therapy
.
J Clin Endocrinol Metab
1996
;
9
:
3222
3230
.

Droguett
R
,
Cabello-Verrugio
C
,
Riquelme
C
,
Brandan
E
.
Extracellular proteoglycans modify TGF-β bio-availability attenuating its signaling during skeletal muscle differentiation
.
Matrix Biol
2006
;
6
:
332
341
.

Duscher
D
,
Maan
ZN
,
Wong
VW
,
Rennert
RC
,
Januszyk
M
,
Rodrigues
M
,
Hu
M
,
Whitmore
AJ
,
Whittam
AJ
,
Longaker
MT
.
Mechanotransduction and fibrosis
.
J Biomech
2014
;
9
:
1997
2005
.

Eichenlaub-Ritter
U
,
Winterscheidt
U
,
Vogt
E
,
Shen
Y
,
Tinneberg
H-R
,
Sorensen
R
.
2-Methoxyestradiol induces spindle aberrations, chromosome congression failure, and nondisjunction in mouse oocytes
.
Biol Reprod
2007
;
5
:
784
793
.

Engel
JB
,
Audebert
A
,
Frydman
R
,
Zivny
J
,
Diedrich
K
.
Presurgical short term treatment of uterine fibroids with different doses of cetrorelix acetate: a double-blind, placebo-controlled multicenter study
.
Eur J Obstet Gynecol Reprod Biol
2007
;
2
:
225
232
.

Esteve
JLC
,
Acosta
R
,
Pérez
Y
,
Rodriguez
B
,
Seigler
I
,
Sanchez
C
,
Tomasi
G
.
Mifepristone versus placebo to treat uterine myoma: a double-blind, randomized clinical trial
.
Int J Womens Health
2013
;
5
:
361
369
.

Esteve
JLC
,
Acosta
R
,
Perez
Y
,
Campos
R
,
Hernandez
AV
,
Texido
CS
.
Treatment of uterine myoma with 5 or 10mg mifepristone daily during 6 months, post-treatment evolution over 12 months: double-blind randomised clinical trial
.
Eur J Obstet Gynecol Reprod Biol
2012
;
2
:
202
208
.

Ettinger
B
,
Black
DM
,
Mitlak
BH
,
Knickerbocker
RK
,
Nickelsen
T
,
Genant
HK
,
Christiansen
C
,
Delmas
PD
,
Zanchetta
JR
,
Stakkestad
J
.
Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial
.
JAMA
1999
;
7
:
637
645
.

Eyden
BP
,
Hale
RJ
,
Richmond
I
,
Buckley
CH
.
Cytoskeletal filaments in the smooth muscle cells of uterine leiomyomata and myometrium: an ultrastructural and immunohistochemical analysis
.
Virchows Arch A Pathol Anat Histopathol
1992
;
1
:
51
58
.

Felberbaum
RE
,
Küpker
W
,
Krapp
M
,
Gehl
B
,
Ludwig
M
,
Diedrich
K
.
Preoperative reduction of uterine fibroids in only 16 days by administration of a gonadotrophin-releasing hormone antagonist (Cetrotide)
.
Reprod Biomed Online
2001
;
1
:
14
18
.

Feng
L
,
Jayes
FL
,
Johnson
LN
,
Schomberg
DW
,
Leppert
PC
.
Biochemical pathways and myometrial cell differentiation leading to nodule formation containing collagen and fibronectin
.
Curr Protein Pept Sci
2016
;
2
:
155
166
.

Fichtner-Feigl
S
,
Strober
W
,
Kawakami
K
,
Puri
RK
,
Kitani
A
.
IL-13 signaling through the IL-13α2 receptor is involved in induction of TGF-β1 production and fibrosis
.
Nat Med
2005
;
1
:
99
106
.

Fiedler
LR
,
Schönherr
E
,
Waddington
R
,
Niland
S
,
Seidler
DG
,
Aeschlimann
D
,
Eble
JA
.
Decorin regulates endothelial cell motility on collagen I through activation of insulin-like growth factor I receptor and modulation of α2β1 integrin activity
.
J Biol Chem
2008
;
25
:
17406
17415
.

Fletcher
NM
,
Saed
MG
,
Abu-Soud
HM
,
Al-Hendy
A
,
Diamond
MP
,
Saed
GM
.
Uterine fibroids are characterized by an impaired antioxidant cellular system: potential role of hypoxia in the pathophysiology of uterine fibroids
.
J Assist Reprod Genet
2013
;
7
:
969
974
.

Font
B
,
Eichenberger
D
,
Goldschmidt
D
,
Boutillon
MM
,
Hulmes
DJS
.
Structural requirements for fibromodulin binding to collagen and the control of type I collagen fibrillogenesis
.
Eur J Biochem
1998
;
3
:
580
587
.

Gabbiani
G
.
The myofibroblast in wound healing and fibrocontractive diseases
.
J Pathol
2003
;
4
:
500
503
.

Gallagher
CS
,
Morton
CC
.
Genetic association studies in uterine fibroids: risk alleles presage the path to personalized therapies
.
Semin Reprod Med
2016
;
34
:
235
241
.

Gallagher
WM
,
Currid
CA
,
Whelan
LC
.
Fibulins and cancer: friend or foe?
Trends Mol Med
2005
;
7
:
336
340
.

Geiger
B
,
Bershadsky
A
,
Pankov
R
,
Yamada
KM
.
Transmembrane crosstalk between the extracellular matrix and the cytoskeleton
.
Nat Rev Mol Cell Biol
2001
;
11
:
793
805
.

Gentry
CC
,
Okolo
SO
,
Fong
LF
,
Crow
JC
,
Maclean
AB
,
Perrett
CW
.
Quantification of vascular endothelial growth factor-A in leiomyomas and adjacent myometrium
.
Clin Sci (Lond)
2001
;
6
:
691
695
.

Giannandrea
M
,
Parks
WC
.
Diverse functions of matrix metalloproteinases during fibrosis
.
Dis Model Mech
2014
;
2
:
193
203
.

Gilden
M
,
Malik
M
,
Britten
J
,
Delgado
T
,
Levy
G
,
Catherino
WH
.
Leiomyoma fibrosis inhibited by liarozole, a retinoic acid metabolic blocking agent
.
Fertil Steril
2012
;
4
:
1557
1562
.

Giri
SN
,
Hyde
DM
,
Braun
RK
,
Gaarde
W
,
Harper
JR
,
Pierschbacher
MD
.
Antifibrotic effect of decorin in a bleomycin hamster model of lung fibrosis
.
Biochem Pharmacol
1997
;
11
:
1205
1216
.

Gonzalez-Barcena
D
,
Alvarez
RB
,
Ochoa
EP
,
Cornejo
IC
,
Comaru-Schally
AM
,
Schally
AV
,
Engel
J
,
Reissmann
T
,
Riethmuller-Winzen
H
.
Treatment of uterine leiomyomas with luteinizing hormone-releasing hormone antagonist Cetrorelix
.
Hum Reprod
1997
;
9
:
2028
2035
.

Goss
PE
,
Strasser
K
,
Marques
R
,
Clemons
M
,
Oza
A
,
Goel
R
,
Blackstein
M
,
Kaizer
L
,
Sterns
EE
,
Nabholtz
J-M
.
Liarozole fumarate (R85246): in the treatment of ER negative, tamoxifen refractory or chemotherapy resistant postmenopausal metastatic breast cancer
.
Breast Cancer Res Treat
2000
;
2
:
177
188
.

Grudzien
MM
,
Low
PS
,
Manning
PC
,
Arredondo
M
,
Belton
RJ
,
Nowak
RA
.
The antifibrotic drug halofuginone inhibits proliferation and collagen production by human leiomyoma and myometrial smooth muscle cells
.
Fertil Steril
2010
;
4
:
1290
1298
.

Guo
J
,
Gu
N
,
Chen
J
,
Shi
T
,
Zhou
Y
,
Rong
Y
,
Zhou
T
,
Yang
W
,
Cui
X
,
Chen
W
.
Neutralization of interleukin-1 beta attenuates silica-induced lung inflammation and fibrosis in C57BL/6 mice
.
Arch Toxicol
2013
;
11
:
1963
1973
.

Guo
XC
,
Segars
JH
.
The impact and management of fibroids for fertility: an evidence-based approach
.
Obstet Gynecol Clin North Am
2012
;
4
:
521
533
.

Gupta
SC
,
Patchva
S
,
Aggarwal
BB
.
Therapeutic roles of curcumin: lessons learned from clinical trials
.
AAPS J
2013
;
1
:
195
218
.

Halder
SK
,
Goodwin
JS
,
Al-Hendy
A
.
1,25-Dihydroxyvitamin D reduces TGF-β3-induced fibrosis-related gene expression in human uterine leiomyoma cells
.
J Clin Endocrinol Metab
2011
;
4
:
E754
E762
.

Halder
SK
,
Osteen
KG
,
Al-Hendy
A
.
1, 25-dihydroxyvitamin d3 reduces extracellular matrix-associated protein expression in human uterine fibroid cells
.
Biol Reprod
2013
;
6
:
150
.

Halder
SK
,
Sharan
C
,
Al-Hendy
A
.
1,25-dihydroxyvitamin D3 treatment shrinks uterine leiomyoma tumors in the Eker rat model
.
Biol Reprod
2012
;
4
:
116
.

Hanks
SK
,
Calalb
MB
,
Harper
MC
,
Patel
SK
.
Focal adhesion protein-tyrosine kinase phosphorylated in response to cell attachment to fibronectin
.
Proc Natl Acad Sci USA
1992
;
18
:
8487
8491
.

Harrison-Woolrych
ML
,
Sharkey
AM
,
Charnock-Jones
DS
,
Smith
SK
.
Localization and quantification of vascular endothelial growth factor messenger ribonucleic acid in human myometrium and leiomyomata
.
J Clin Endocrinol Metab
1995
;
6
:
1853
1858
.

Harrison
MR
,
Hahn
NM
,
Pili
R
,
Oh
WK
,
Hammers
H
,
Sweeney
C
,
Kim
K
,
Perlman
S
,
Arnott
J
,
Sidor
C
.
A phase II study of 2-methoxyestradiol (2ME2) NanoCrystal® dispersion (NCD) in patients with taxane-refractory, metastatic castrate-resistant prostate cancer (CRPC)
.
Invest New Drugs
2011
;
6
:
1465
1474
.

Hassan
MH
,
Eyzaguirre
E
,
Arafa
HMM
,
Hamada
FMA
,
Salama
SA
,
Al-Hendy
A
.
Memy I: a novel murine model for uterine leiomyoma using adenovirus-enhanced human fibroid explants in severe combined immune deficiency mice
.
Am J Obstet Gynecol
2008
;
2
:
156. e1
156. e8
.

Hassan
MH
,
Salama
SA
,
Arafa
HM
,
Hamada
FM
,
Al-Hendy
A
.
Adenovirus-mediated delivery of a dominant-negative estrogen receptor gene in uterine leiomyoma cells abrogates estrogen- and progesterone-regulated gene expression
.
J Clin Endocrinol Metab
2007
;
10
:
3949
3957
.

Hedbom
E
,
Heinegård
D
.
Binding of fibromodulin and decorin to separate sites on fibrillar collagens
.
J Biol Chem
1993
;
36
:
27307
27312
.

Herndon
CN
,
Aghajanova
L
,
Bayalan
S
,
Erikson
D
,
Barragan
F
,
Goldfien
G
,
Vo
KC
,
Hawkins
S
,
Giudice
LC
.
Global transcriptome abnormalities of the eutopic endometrium from women with adenomyosis
.
Reprod Sci
2016
;
10
:
1289
1303
.

Higgins
DF
,
Kimura
K
,
Bernhardt
WM
,
Shrimanker
N
,
Akai
Y
,
Hohenstein
B
,
Saito
Y
,
Johnson
RS
,
Kretzler
M
,
Cohen
CD
.
Hypoxia promotes fibrogenesis in vivo via HIF-1 stimulation of epithelial-to-mesenchymal transition
.
J Clin Invest
2007
;
12
:
3810
3820
.

Hinz
B
,
Phan
SH
,
Thannickal
VJ
,
Galli
A
,
Bochaton-Piallat
ML
,
Gabbiani
G
.
The myofibroblast: one function, multiple origins
.
Am J Pathol
2007
;
6
:
1807
1816
.

Ho
YY
,
Lagares
D
,
Tager
AM
,
Kapoor
M
.
Fibrosis-a lethal component of systemic sclerosis
.
Nat Rev Rheumatol
2014
;
7
:
390
402
.

Hoekstra
AV
,
Sefton
EC
,
Berry
E
,
Lu
Z
,
Hardt
J
,
Marsh
E
,
Yin
P
,
Clardy
J
,
Chakravarti
D
,
Bulun
S
.
Progestins activate the AKT pathway in leiomyoma cells and promote survival
.
J Clin Endocrinol Metab
2009
;
5
:
1768
1774
.

Holdsworth-Carson
SJ
,
Zaitseva
M
,
Vollenhoven
BJ
,
Rogers
PAW
.
Clonality of smooth muscle and fibroblast cell populations isolated from human fibroid and myometrial tissues
.
Mol Hum Reprod
2014
;
3
:
250
259
.

Huang
H
,
Kamm
RD
,
Lee
RT
.
Cell mechanics and mechanotransduction: pathways, probes, and physiology
.
Am J Physiol Cell Physiol
2004
;
1
:
C1
C11
.

Hwu
YM
,
Li
SH
,
Lee
RK
,
Tsai
YH
,
Yeh
TS
,
Lin
SY
.
Increased expression of platelet-derived growth factor C messenger ribonucleic acid in uterine leiomyomata
.
Fertil Steril
2008
;
2
:
468
471
.

Iimuro
Y
,
Nishio
T
,
Morimoto
T
,
Nitta
T
,
Stefanovic
B
,
Choi
SK
,
Brenner
DA
,
Yamaoka
Y
.
Delivery of matrix metalloproteinase-1 attenuates established liver fibrosis in the rat
.
Gastroenterology
2003
;
2
:
445
458
.

Ikenaka
Y
,
Yoshiji
H
,
Kuriyama
S
,
Yoshii
J
,
Noguchi
R
,
Tsujinoue
H
,
Yanase
K
,
Namisaki
T
,
Imazu
H
,
Masaki
T
.
Tissue inhibitor of metalloproteinases‐1 (TIMP‐1) inhibits tumor growth and angiogenesis in the TIMP‐1 transgenic mouse model
.
Int J Cancer
2003
;
3
:
340
346
.

Islam
MS
,
Catherino
WH
,
Protic
O
,
Janjusevic
M
,
Gray
PC
,
Giannubilo
SR
,
Ciavattini
A
,
Lamanna
P
,
Tranquilli
AL
,
Petraglia
F
et al. .
Role of activin-A and myostatin and their signaling pathway in human myometrial and leiomyoma cell function
.
J Clin Endocrinol Metab
2014
a;
5
:
E775
E785
.

Islam
MS
,
Greco
S
,
Janjusevic
M
,
Ciavattini
A
,
Giannubilo
SR
,
D’Adderio
A
,
Biagini
A
,
Fiorini
R
,
Castellucci
M
,
Ciarmela
P
.
Growth factors and pathogenesis
.
Best Pract Res Clin Obstet Gynaecol
2016
;
34
:
25
36
.

Islam
MS
,
Protic
O
,
Ciavattini
A
,
Giannubilo
SR
,
Tranquilli
AL
,
Catherino
WH
,
Castellucci
M
,
Ciarmela
P
.
Tranilast, an orally active antiallergic compound, inhibits extracellular matrix production in human uterine leiomyoma and myometrial cells
.
Fertil Steril
2014
b;
2
:
597
606
.

Islam
MS
,
Protic
O
,
Giannubilo
S
,
Stortoni
P
,
Ciavattini
A
,
Lamanna
P
,
Tranquilli
AL
,
Castellucci
M
,
Ciarmela
P
.
Antiproliferative effect of tranilast on human myometrial and leiomyoma cells
.
Biol Biomed Rep
2012
;
5
:
321
327
.

Islam
MS
,
Protic
O
,
Stortoni
P
,
Grechi
G
,
Lamanna
P
,
Petraglia
F
,
Castellucci
M
,
Ciarmela
P
.
Complex networks of multiple factors in the pathogenesis of uterine leiomyoma
.
Fertil Steril
2013
a;
1
:
178
193
.

Islam
MS
,
Protic
O
,
Toti
P
,
Giannubilo
SR
,
Tranquilli
AL
,
Petraglia
F
,
Castellucci
M
,
Ciarmela
P
.
Uterine leiomyoma: available medical treatments and new possible therapeutic options
.
J Clin Endocrinol Metab
2013
b;
3
:
921
934
.

Itoh
Y
.
MT1‐MMP: A key regulator of cell migration in tissue
.
IUBMB life
2006
;
10
:
589
596
.

Iwahashi
M
,
Muragaki
Y
.
Increased type I and V collagen expression in uterine leiomyomas during the menstrual cycle
.
Fertil Steril
2011
;
6
:
2137
2139
.

Iwahashi
M
,
Muragaki
Y
,
Ikoma
M
,
Mabuchi
Y
,
Kobayashi
AYA
,
Tanizaki
Y
,
Ino
K
.
Immunohistochemical analysis of collagen expression in uterine leiomyomata during the menstrual cycle
.
Exp Ther Med
2010
;
2
:
287
290
.

Jakubowska
K
,
Pryczynicz
A
,
Iwanowicz
P
,
Niewiński
A
,
Maciorkowska
E
,
Hapanowicz
J
,
Jagodzińska
D
,
Kemona
A
,
Guzińska-Ustymowicz
K
.
Expressions of matrix metalloproteinases (MMP-2, MMP-7, and MMP-9) and their inhibitors (TIMP-1, TIMP-2) in inflammatory bowel diseases
.
Gastroenterol Res Pract
2016
;
2016
:
2456179
.

Jayes
FL
,
Liu
B
,
Moutos
FT
,
Kuchibhatla
M
,
Guilak
F
,
Leppert
PC
.
Loss of stiffness in collagen-rich uterine fibroids after digestion with purified collagenase Clostridium histolyticum
.
Am J Obstet Gynecol
2016
;
5
:
596.e1
596.e8
.

Jirecek
S
,
Lee
A
,
Pavo
I
,
Crans
G
,
Eppel
W
,
Wenzl
R
.
Raloxifene prevents the growth of uterine leiomyomas in premenopausal women
.
Fertil Steril
2004
;
1
:
132
136
.

Jirouskova
M
,
Zbodakova
O
,
Gregor
M
,
Chalupsky
K
,
Sarnova
L
,
Hajduch
M
,
Ehrmann
J
,
Jirkovska
M
,
Sedlacek
R
.
Hepatoprotective effect of MMP-19 deficiency in a mouse model of chronic liver fibrosis
.
PloS one
2012
;
10
:
e46271
.

Jones
JL
,
Walker
RA
.
Integrins: a role as cell signalling molecules
.
Mol Pathol
1999
;
4
:
208
.

Joseph
DS
,
Malik
M
,
Nurudeen
S
,
Catherino
WH
.
Myometrial cells undergo fibrotic transformation under the influence of transforming growth factor β-3
.
Fertil Steril
2010
;
5
:
1500
1508
.

Kashani
BN
,
Centini
G
,
Morelli
SS
,
Weiss
G
,
Petraglia
F
.
Role of medical management for uterine leiomyomas
.
Best Pract Res Clin Obstet Gynaecol
2016
;
34
:
85
103
.

Kastner
P
,
Krust
A
,
Turcotte
B
,
Stropp
U
,
Tora
L
,
Gronemeyer
H
,
Chambon
P
.
Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B
.
EMBO J
1990
;
5
:
1603
1614
.

Kawaguchi
K
,
Fujii
S
,
Konishi
I
,
Nanbu
Y
,
Nonogaki
H
,
Mori
T
.
Mitotic activity in uterine leiomyomas during the menstrual cycle
.
Am J Obstet Gynecol
1989
;
3
:
637
641
.

Ke
X
,
Dou
F
,
Cheng
Z
,
Dai
H
,
Zhang
W
,
Qu
X
,
Ding
P
,
Zuo
X
.
High expression of cyclooxygenase-2 in uterine fibroids and its correlation with cell proliferation
.
Eur J Obstet Gynecol Reprod Biol
2013
;
2
:
199
203
.

Kino
T
,
Takatori
H
,
Manoli
I
,
Wang
Y
,
Tiulpakov
A
,
Blackman
MR
,
Su
YA
,
Chrousos
GP
,
DeCherney
AH
,
Segars
JH
.
Brx mediates the response of lymphocytes to osmotic stress through the activation of NFAT5
.
Sci Signal
2009
;
57
:
ra5
.

Kjerulff
KH
,
Langenberg
P
,
Seidman
JD
,
Stolley
PD
,
Guzinski
GM
.
Uterine leiomyomas. Racial differences in severity, symptoms and age at diagnosis
.
J Reprod Med
1996
;
7
:
483
490
.

Klein
T
,
Bischoff
R
.
Physiology and pathophysiology of matrix metalloproteases
.
Amino Acids
2011
;
2
:
271
290
.

Kleinman
HK
,
Cannon
FB
,
Laurie
GW
,
Hassell
JR
,
Aumailley
M
,
Terranova
VP
,
Martin
GR
,
DuBois-Dalcq
M
.
Biological activities of laminin
.
J Cell Biochem
1985
;
4
:
317
325
.

Konneh
M
.
Tranilast Kissei Pharmaceutical
.
IDrugs
1998
;
1
:
141
146
.

Koohestani
F
,
Braundmeier
AG
,
Mahdian
A
,
Seo
J
,
Bi
J
,
Nowak
RA
.
Extracellular matrix collagen alters cell proliferation and cell cycle progression of human uterine leiomyoma smooth muscle cells
.
PloS one
2013
;
9
:
e75844
.

Koohestani
F
,
Qiang
W
,
MacNeill
AL
,
Druschitz
SA
,
Serna
VA
,
Adur
M
,
Kurita
T
,
Nowak
RA
.
Halofuginone suppresses growth of human uterine leiomyoma cells in a mouse xenograft model
.
Hum Reprod
2016
;
7
:
1540
1551
.

Korompelis
P
,
Piperi
C
,
Adamopoulos
C
,
Dalagiorgou
G
,
Korkolopoulou
P
,
Sepsa
A
,
Antsaklis
A
,
Papavassiliou
AG
.
Expression of vascular endothelial factor-A, gelatinases (MMP-2, MMP-9) and TIMP-1 in uterine leiomyomas
.
Clin Chem Lab Med
2015
;
9
:
1415
1424
.

Kovacs
KA
,
Oszter
A
,
Gocze
PM
,
Kornyei
JL
,
Szabo
I
.
Comparative analysis of cyclin D1 and oestrogen receptor (α and β) levels in human leiomyoma and adjacent myometrium
.
Mol Hum Reprod
2001
;
11
:
1085
1091
.

Kurachi
O
,
Matsuo
H
,
Samoto
T
,
Maruo
T
.
Tumor necrosis factor-α expression in human uterine leiomyoma and its down-regulation by progesterone
.
J Clin Endocrinol Metab
2001
;
5
:
2275
2280
.

Kurzrock
R
,
Estey
E
,
Talpaz
M
.
All-trans retinoic acid: tolerance and biologic effects in myelodysplastic syndrome
.
J Clin Oncol
1993
;
8
:
1489
1495
.

Lee
BS
,
Margolin
SB
,
Nowak
RA
.
Pirfenidone: a novel pharmacological agent that inhibits leiomyoma cell proliferation and collagen production
.
J Clin Endocrinol Metab
1998
;
1
:
219
223
.

Lee
CG
,
Homer
RJ
,
Zhu
Z
,
Lanone
S
,
Wang
X
,
Koteliansky
V
,
Shipley
JM
,
Gotwals
P
,
Noble
P
,
Chen
Q
.
Interleukin-13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor β1
.
J Exp Med
2001
;
6
:
809
822
.

Lee
S
,
Jilani
SM
,
Nikolova
GV
,
Carpizo
D
,
Iruela-Arispe
ML
.
Processing of VEGF-A by matrix metalloproteinases regulates bioavailability and vascular patterning in tumors
.
J Cell Biol
2005
;
4
:
681
691
.

Leibovich
SJ
,
Ross
R
.
The role of the macrophage in wound repair. A study with hydrocortisone and antimacrophage serum
.
Am J Pathol
1975
;
1
:
71
100
.

Leppert
P
,
Fouany
M
,
Segars
JH
. Understanding uterine fibroids. In:
Segars
JH
(ed).
Fibroids
.
Oxford
:
John Wiley & Sons, Ltd
,
2013
;
1
10
.

Leppert
PC
,
Baginski
T
,
Prupas
C
,
Catherino
WH
,
Pletcher
S
,
Segars
JH
.
Comparative ultrastructure of collagen fibrils in uterine leiomyomas and normal myometrium
.
Fertil Steril
2004
;
1182
1187
.

Leppert
PC
,
Catherino
WH
,
Segars
JH
.
A new hypothesis about the origin of uterine fibroids based on gene expression profiling with microarrays
.
Am J Obstet Gynecol
2006
;
2
:
415
420
.

Levens
E
,
Luo
X
,
Ding
L
,
Williams
RS
,
Chegini
N
.
Fibromodulin is expressed in leiomyoma and myometrium and regulated by gonadotropin-releasing hormone analogue therapy and TGF-β through Smad and MAPK-mediated signalling
.
Mol Hum Reprod
2005
;
7
:
489
494
.

Levy
G
,
Malik
M
,
Britten
J
,
Gilden
M
,
Segars
J
,
Catherino
WH
.
Liarozole inhibits transforming growth factor-β3-mediated extracellular matrix formation in human three-dimensional leiomyoma cultures
.
Fertil Steril
2014
;
1
:
272
281. e2
.

Li
G
,
Fridman
R
,
Kim
H-RC
.
Tissue inhibitor of metalloproteinase-1 inhibits apoptosis of human breast epithelial cells
.
Cancer Res
1999
;
24
:
6267
6275
.

Liang
M
,
Wang
H
,
Zhang
Y
,
Lu
S
,
Wang
Z
.
Expression and functional analysis of platelet-derived growth factor in uterine leiomyomata
.
Cancer Biol Ther
2006
;
1
:
28
33
.

Lindahl
GE
,
Chambers
RC
,
Papakrivopoulou
J
,
Dawson
SJ
,
Jacobsen
MC
,
Bishop
JE
,
Laurent
GJ
.
Activation of fibroblast procollagen α1(I) transcription by mechanical strain is transforming growth factor-β-dependent and involves increased binding of CCAAT-binding factor (CBF/NF-Y) at the proximal promoter
.
J Biol Chem
2002
;
8
:
6153
6161
.

Löffek
S
,
Schilling
O
,
Franzke
C-W
.
Biological role of matrix metalloproteinases: a critical balance
.
Eur Respir J
2011
;
38
:
191
208
.

Logan
A
,
Baird
A
,
Berry
M
.
Decorin attenuates gliotic scar formation in the rat cerebral hemisphere
.
Exp Neurol
1999
;
2
:
504
510
.

Luo
N
,
Guan
Q
,
Zheng
L
,
Qu
X
,
Dai
H
,
Cheng
Z
.
Estrogen-mediated activation of fibroblasts and its effects on the fibroid cell proliferation
.
Transl Res
2014
;
3
:
232
241
.

Luo
X
,
Chegini
N
.
The expression and potential regulatory function of microRNAs in the pathogenesis of leiomyoma
.
Semin Reprod Med
2008
;
6
:
500
514
.

Luo
X
,
Ding
L
,
Chegini
N
.
CCNs, fibulin-1C and S100A4 expression in leiomyoma and myometrium: inverse association with TGF-β and regulation by TGF-β in leiomyoma and myometrial smooth muscle cells
.
Mol Hum Reprod
2006
;
4
:
245
256
.

Luo
X
,
Ding
L
,
Xu
J
,
Chegini
N
.
Gene expression profiling of leiomyoma and myometrial smooth muscle cells in response to transforming growth factor-β
.
Endocrinology
2005
;
3
:
1097
1118
.

Luo
X
,
Yin
P
,
Reierstad
S
,
Ishikawa
H
,
Lin
Z
,
Pavone
ME
,
Zhao
H
,
Marsh
EE
,
Bulun
SE
.
Progesterone and mifepristone regulate L-type amino acid transporter 2 and 4F2 heavy chain expression in uterine leiomyoma cells
.
J Clin Endocrinol Metab
2009
;
11
:
4533
4539
.

Mahabeleshwar
GH
,
Feng
W
,
Reddy
K
,
Plow
EF
,
Byzova
TV
.
Mechanisms of integrin-vascular endothelial growth factor receptor cross-activation in angiogenesis
.
Circ Res
2007
;
6
:
570
580
.

Malik
M
,
Britten
J
,
Cox
J
,
Patel
A
,
Catherino
WH
.
Gonadotropin-releasing hormone analogues inhibit leiomyoma extracellular matrix despite presence of gonadal hormones
.
Fertil Steril
2016
;
1
:
214
224
.

Malik
M
,
Britten
J
,
Segars
J
,
Catherino
WH
.
Leiomyoma cells in 3-dimensional cultures demonstrate an attenuated response to fasudil, a rho-kinase inhibitor, when compared to 2-dimensional cultures
.
Reprod Sci
2014
;
9
:
1126
1138
.

Malik
M
,
Catherino
WH
.
Novel method to characterize primary cultures of leiomyoma and myometrium with the use of confirmatory biomarker gene arrays
.
Fertil Steril
2007
;
5
:
1166
1172
.

Malik
M
,
Mendoza
M
,
Payson
M
,
Catherino
WH
.
Curcumin, a nutritional supplement with antineoplastic activity, enhances leiomyoma cell apoptosis and decreases fibronectin expression
.
Fertil Steril
2009
;
5
Suppl:
2177
2184
.

Malik
M
,
Norian
J
,
McCarthy-Keith
D
,
Britten
J
,
Catherino
WH
.
Why leiomyomas are called fibroids: the central role of extracellular matrix in symptomatic women
.
Semin Reprod Med
2010
;
3
:
169
179
.

Malik
M
,
Segars
J
,
Catherino
WH
.
Integrin β1 regulates leiomyoma cytoskeletal integrity and growth
.
Matrix Biol
2012
;
7-8
:
389
397
.

Malik
M
,
Webb
J
,
Catherino
WH
.
Retinoic acid treatment of human leiomyoma cells transformed the cell phenotype to one strongly resembling myometrial cells
.
Clin Endocrinol (Oxf)
2008
;
3
:
462
470
.

Mangrulkar
RS
,
Ono
M
,
Ishikawa
M
,
Takashima
S
,
Klagsbrun
M
,
Nowak
RA
.
Isolation and characterization of heparin-binding growth factors in human leiomyomas and normal myometrium
.
Biol Reprod
1995
;
3
:
636
646
.

Manicone
AM
,
Huizar
I
,
McGuire
JK
.
Matrilysin (matrix metalloproteinase-7) regulates anti-inflammatory and antifibrotic pulmonary dendritic cells that express CD103 (α E β 7-integrin)
.
Am J Pathol
2009
;
6
:
2319
2331
.

Maniotis
AJ
,
Chen
CS
,
Ingber
DE
.
Demonstration of mechanical connections between integrins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure
.
Proc Natl Acad Sci USA
1997
;
3
:
849
854
.

Mannello
F
,
Luchetti
F
,
Falcieri
E
,
Papa
S
.
Multiple roles of matrix metalloproteinases during apoptosis
.
Apoptosis
2005
;
1
:
19
24
.

Marsh
EE
,
Chibber
S
,
Wu
J
,
Siegersma
K
,
Kim
J
,
Bulun
S
.
Epidermal growth factor-containing fibulin-like extracellular matrix protein 1 expression and regulation in uterine leiomyoma
.
Fertil Steril
2016
a;
4
:
1070
1075
.

Marsh
EE
,
Steinberg
ML
,
Parker
JB
,
Wu
J
,
Chakravarti
D
,
Bulun
SE
.
Decreased expression of microRNA-29 family in leiomyoma contributes to increased major fibrillar collagen production
.
Fertil Steril
2016
b;
3
:
766
772
.

Marshall
LM
,
Spiegelman
D
,
Barbieri
RL
,
Goldman
MB
,
Manson
JE
,
Colditz
GA
,
Willett
WC
,
Hunter
DJ
.
Variation in the incidence of uterine leiomyoma among premenopausal women by age and race
.
Obstet Gynecol
1997
;
6
:
967
973
.

Martino
S
,
Disch
D
,
Dowsett
SA
,
Keech
CA
,
Mershon
JL
.
Safety assessment of raloxifene over eight years in a clinical trial setting
.
Curr Med Res Opin
2005
;
9
:
1441
1452
.

Maruo
T
,
Ohara
N
,
Wang
J
,
Matsuo
H
.
Sex steroidal regulation of uterine leiomyoma growth and apoptosis
.
Hum Reprod Update
2004
;
3
:
207
220
.

Mas
A
,
Nair
S
,
Laknaur
A
,
Simon
C
,
Diamond
MP
,
Al-Hendy
A
.
Stro-1/CD44 as putative human myometrial and fibroid stem cell markers
.
Fertil Steril
2015
;
1
:
225
234.e3
.

Matrisian
LM
.
The matrix-degrading metalloproteinases
.
Bioessays
1992
;
7
:
455
463
.

Matsumoto
S-i
,
Katoh
M
,
Saito
S
,
Watanabe
T
,
Masuho
Y
.
Identification of soluble type of membrane-type matrix metalloproteinase-3 formed by alternatively spliced mRNA
.
Biochim Biophys Acta
1997
;
2
:
159
170
.

McCarthy-Keith
DM
,
Malik
M
,
Britten
J
,
Segars
J
,
Catherino
WH
.
Gonadotropin-releasing hormone agonist increases expression of osmotic response genes in leiomyoma cells
.
Fertil Steril
2011
;
7
:
2383
2387
.

McCawley
LJ
,
Matrisian
LM
.
Matrix metalloproteinases: they’re not just for matrix anymore!
Curr Opin Cell Biol
2001
;
5
:
534
540
.

Melis
GB
,
Piras
B
,
Marotto
MF
,
Orru
MM
,
Maricosu
G
,
Pilloni
M
,
Guerriero
S
,
Angiolucci
M
,
Lello
S
,
Paoletti
AM
.
Pharmacokinetic evaluation of ulipristal acetate for uterine leiomyoma treatment
.
Expert Opin Drug Metab Toxicol
2012
;
7
:
901
908
.

Mesquita
FS
,
Dyer
SN
,
Heinrich
DA
,
Bulun
SE
,
Marsh
EE
,
Nowak
RA
.
Reactive oxygen species mediate mitogenic growth factor signaling pathways in human leiomyoma smooth muscle cells
.
Biol Reprod
2010
;
2
:
341
351
.

Mitropoulou
TN
,
Theocharis
AD
,
Stagiannis
KD
,
Karamanos
NK
.
Identification, quantification and fine structural characterization of glycosaminoglycans from uterine leiomyoma and normal myometrium
.
Biochimie
2001
;
6
:
529
536
.

Mitsiades
N
,
Yu
W-H
,
Poulaki
V
,
Tsokos
M
,
Stamenkovic
I
.
Matrix metalloproteinase-7-mediated cleavage of Fas ligand protects tumor cells from chemotherapeutic drug cytotoxicity
.
Cancer Res
2001
;
2
:
577
581
.

Moore
AB
,
Yu
L
,
Swartz
CD
,
Zheng
X
,
Wang
L
,
Castro
L
,
Kissling
GE
,
Walmer
DK
,
Robboy
SJ
,
Dixon
D
.
Human uterine leiomyoma-derived fibroblasts stimulate uterine leiomyoma cell proliferation and collagen type I production, and activate RTKs and TGF beta receptor signaling in coculture
.
Cell Commun Signal
2010
;
8
:
10
.

Morikawa
A
,
Ohara
N
,
Xu
Q
,
Nakabayashi
K
,
DeManno
DA
,
Chwalisz
K
,
Yoshida
S
,
Maruo
T
.
Selective progesterone receptor modulator asoprisnil down-regulates collagen synthesis in cultured human uterine leiomyoma cells through up-regulating extracellular matrix metalloproteinase inducer
.
Hum Reprod
2008
;
4
:
944
951
.

Mui
KL
,
Chen
CS
,
Assoian
RK
.
The mechanical regulation of integrin–cadherin crosstalk organizes cells, signaling and forces
.
J Cell Sci
2016
;
6
:
1093
1100
.

Murphy
FR
,
Issa
R
,
Zhou
X
,
Ratnarajah
S
,
Nagase
H
,
Arthur
MJ
,
Benyon
C
,
Iredale
JP
.
Inhibition of apoptosis of activated hepatic stellate cells by tissue inhibitor of metalloproteinase-1 is mediated via effects on matrix metalloproteinase inhibition implications for reversibility of liver fibrosis
.
J Biol Chem
2002
;
13
:
11069
11076
.

Nagase
H
,
Visse
R
,
Murphy
G
.
Structure and function of matrix metalloproteinases and TIMPs
.
Cardiovasc Res
2006
;
3
:
562
573
.

Nair
S
,
Al-Hendy
A
.
Adipocytes enhance the proliferation of human leiomyoma cells via TNF-α proinflammatory cytokine
.
Reprod Sci
2011
;
12
:
1186
1192
.

Nakamura
T
,
Sugino
K
,
Titani
K
,
Sugino
H
.
Follistatin, an activin-binding protein, associates with heparan sulfate chains of proteoglycans on follicular granulosa cells
.
J Biol Chem
1991
;
29
:
19432
19437
.

Nelissen
I
,
Martens
E
,
Van den Steen
PE
,
Proost
P
,
Ronsse
I
,
Opdenakker
G
.
Gelatinase B/matrix metalloproteinase-9 cleaves interferonon-β and is a target for immunotherapy
.
Brain
2003
;
6
:
1371
1381
.

Nierth-Simpson
EN
,
Martin
MM
,
Chiang
TC
,
Melnik
LI
,
Rhodes
LV
,
Muir
SE
,
Burow
ME
,
McLachlan
JA
.
Human uterine smooth muscle and leiomyoma cells differ in their rapid 17β-estradiol signaling: implications for proliferation
.
Endocrinology
2009
;
5
:
2436
2445
.

Nilsson
S
,
Makela
S
,
Treuter
E
,
Tujague
M
,
Thomsen
J
,
Andersson
G
,
Enmark
E
,
Pettersson
K
,
Warner
M
,
Gustafsson
.
Mechanisms of estrogen action
.
Physiol Rev
2001
;
4
:
1535
1565
.

Nishizuka
I
,
Ichikawa
Y
,
Ishikawa
T
,
Kamiyama
M
,
Hasegawa
S
,
Momiyama
N
,
Miyazaki
K
,
Shimada
H
.
Matrilysin stimulates DNA synthesis of cultured vascular endothelial cells and induces angiogenesis in vivo
.
Cancer Lett
2001
;
2
:
175
182
.

Norian
JM
,
Malik
M
,
Parker
CY
,
Joseph
D
,
Leppert
PC
,
Segars
JH
,
Catherino
WH
.
Transforming growth factor β3 regulates the versican variants in the extracellular matrix-rich uterine leiomyomas
.
Reprod Sci
2009
;
12
:
1153
1164
.

Norian
JM
,
Owen
CM
,
Taboas
J
,
Korecki
C
,
Tuan
R
,
Malik
M
,
Catherino
WH
,
Segars
JH
.
Characterization of tissue biomechanics and mechanical signaling in uterine leiomyoma
.
Matrix Biol
2012
;
1
:
57
65
.

Ockleford
C
,
Bright
N
,
Hubbard
A
,
D’Lacey
C
,
Smith
J
,
Gardiner
L
,
Sheikh
T
,
Albentosa
M
,
Turtle
K
.
Micro-trabeculae, macro-plaques or mini-basement membranes in human term fetal membranes?
Philos Trans R Soc Lond B Biol Sci
1993
;
1300
:
121
136
.

Oh
J
,
Seo
D-W
,
Diaz
T
,
Wei
B
,
Ward
Y
,
Ray
JM
,
Morioka
Y
,
Shi
S
,
Kitayama
H
,
Takahashi
C
.
Tissue inhibitors of metalloproteinase 2 inhibits endothelial cell migration through increased expression of RECK
.
Cancer Res
2004
;
24
:
9062
9069
.

Okolo
S
.
Incidence, aetiology and epidemiology of uterine fibroids
.
Best Pract Res Clin Obstet Gynaecol
2008
;
4
:
571
588
.

Ono
M
,
Maruyama
T
,
Masuda
H
,
Kajitani
T
,
Nagashima
T
,
Arase
T
,
Ito
M
,
Ohta
K
,
Uchida
H
,
Asada
H
.
Side population in human uterine myometrium displays phenotypic and functional characteristics of myometrial stem cells
.
Proc Natl Acad Sci USA
2007
;
47
:
18700
18705
.

Onozuka
I
,
Kakinuma
S
,
Kamiya
A
,
Miyoshi
M
,
Sakamoto
N
,
Kiyohashi
K
,
Watanabe
T
,
Funaoka
Y
,
Ueyama
M
,
Nakagawa
M
.
Cholestatic liver fibrosis and toxin-induced fibrosis are exacerbated in matrix metalloproteinase-2 deficient mice
.
Biochem Biophys Res Commun
2011
;
1
:
134
140
.

Ortéga
N
,
L’Faqihi
F-E
,
Plouët
J
.
Control of vascular endothelial growth factor angiogenic activity by the extracellular matrix
.
Biol Cell
1998
;
5
:
381
390
.

Paffoni
A
,
Somigliana
E
,
Vigano
P
,
Benaglia
L
,
Cardellicchio
L
,
Pagliardini
L
,
Papaleo
E
,
Candiani
M
,
Fedele
L
.
Vitamin D status in women with uterine leiomyomas
.
J Clin Endocrinol Metab
2013
;
8
:
E1374
E1378
.

Palmer
SS
,
Haynes-Johnson
D
,
Diehl
T
,
Nowak
RA
.
Increased expression of stromelysin 3 mRNA in leiomyomas (uterine fibroids) compared with myometrium
.
J Soc Gynecol Investig
1998
;
4
:
203
209
.

Pankov
R
,
Yamada
KM
.
Fibronectin at a glance
.
J Cell Sci
2002
;
20
:
3861
3863
.

Park
JE
,
Keller
G-A
,
Ferrara
N
.
The vascular endothelial growth factor (VEGF) isoforms: differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF
.
Mol Biol Cell
1993
;
12
:
1317
1326
.

Park
SB
,
Jee
BC
,
Kim
SH
,
Cho
YJ
,
Han
M
.
Cyclooxygenase-2 inhibitor, celecoxib, inhibits leiomyoma cell proliferation through the nuclear factor κB pathway
.
Reprod Sci
2014
;
9
:
1187
1195
.

Parsons
JT
.
Focal adhesion kinase: the first ten years
.
J Cell Sci
2003
;
8
:
1409
1416
.

PaszeK
MJ
,
Zahir
N
,
Johnson
KR
,
Lakins
JN
,
Rozenberg
GI
,
Gefen
A
,
Reinhart-King
CA
,
Margulies
SS
,
Dembo
M
,
Boettiger
D
.
Tensional homeostasis and the malignant phenotype
.
Cancer cell
2005
;
3
:
241
254
.

Patel
A
,
Malik
M
,
Britten
J
,
Cox
J
,
Catherino
WH
.
Mifepristone inhibits extracellular matrix formation in uterine leiomyoma
.
Fertil Steril
2016
;
4
:
1102
1110
.

Pei
D
.
Identification and characterization of the fifth membrane-type matrix metalloproteinase MT5-MMP
.
J Biol Chem
1999
;
13
:
8925
8932
.

Pickering
JG
.
Regulation of vascular cell behavior by collagen form is function
.
Circ Res
2001
;
5
:
458
459
.

Plewka
A
,
Madej
P
,
Plewka
D
,
Kowalczyk
A
,
Miskiewicz
A
,
Wittek
P
,
Leks
T
,
Bilski
R
.
Immunohistochemical localization of selected pro-inflammatory factors in uterine myomas and myometrium in women of various ages
.
Folia Histochem Cytobiol
2013
;
1
:
73
83
.

Polacheck
WJ
,
German
AE
,
Mammoto
A
,
Ingber
DE
,
Kamm
RD
.
Mechanotransduction of fluid stresses governs 3D cell migration
.
Proc Natl Acad Sci USA
2014
;
7
:
2447
2452
.

Poli
G
.
Pathogenesis of liver fibrosis: role of oxidative stress
.
Mol Aspects Med
2000
;
3
:
49
98
.

Porter
KE
,
Turner
NA
,
O'Regan
DJ
,
Balmforth
AJ
,
Ball
SG
.
Simvastatin reduces human atrial myofibroblast proliferation independently of cholesterol lowering via inhibition of RhoA
.
Cardiovasc Res
2004
;
4
:
745
755
.

Powell
DW
,
Mifflin
RC
,
Valentich
JD
,
Crowe
SE
,
Saada
JI
,
West
AB
.
Myofibroblasts. I. Paracrine cells important in health and disease
.
Am J Physiol Cell Physiol
1999
a;
1
:
C1
C19
.

Powell
WC
,
Fingleton
B
,
Wilson
CL
,
Boothby
M
,
Matrisian
LM
.
The metalloproteinase matrilysin proteolytically generates active soluble Fas ligand and potentiates epithelial cell apoptosis
.
Curr Biol
1999
b;
24
:
1441
1447
.

Protic
O
,
Toti
P
,
Islam
MS
,
Occhini
R
,
Giannubilo
SR
,
Catherino
WH
,
Janjusevic
M
,
Lamanna
P
,
Cinti
S
,
Petraglia
F
et al. .
Possible involvement of inflammatory/reparative processes in the development of uterine fibroids
.
Cell Tissue Res
2016
;
2
:
415
427
.

Qiang
W
,
Liu
Z
,
Serna
VA
,
Druschitz
SA
,
Liu
Y
,
Espona-Fiedler
M
,
Wei
J-J
,
Kurita
T
.
Down-regulation of miR-29b is essential for pathogenesis of uterine leiomyoma
.
Endocrinology
2014
;
3
:
663
669
.

Raines
EW
,
Ross
R
.
Compartmentalization of PDGF on extracellular binding sites dependent on exon-6-encoded sequences
.
J Cell Biol
1992
;
2
:
533
543
.

Rajkumar
SV
,
Richardson
PG
,
Lacy
MQ
,
Dispenzieri
A
,
Greipp
PR
,
Witzig
TE
,
Schlossman
R
,
Sidor
CF
,
Anderson
KC
,
Gertz
MA
.
Novel therapy with 2-methoxyestradiol for the treatment of relapsed and plateau phase multiple myeloma
.
Clin Cancer Res
2007
;
20
:
6162
6167
.

Rapraeger
AC
,
Krufka
A
,
Olwin
BB
.
Requirement of heparan sulfate for bFGF-mediated fibroblast growth and myoblast differentiation
.
Science
1991
;
5013
:
1705
1708
.

Richards
PA
,
Richards
PD
,
Tiltman
AJ
.
The ultrastructure of fibromyomatous myometrium and its relationship to infertility
.
Hum Reprod Update
1998
;
5
:
520
525
.

Rodríguez
D
,
Morrison
CJ
,
Overall
CM
.
Matrix metalloproteinases: what do they not do? New substrates and biological roles identified by murine models and proteomics
.
Biochim Biophys Acta
2010
;
1
:
39
54
.

Rogers
R
,
Norian
J
,
Malik
M
,
Christman
G
,
Abu-Asab
M
,
Chen
F
,
Korecki
C
,
Iatridis
J
,
Catherino
WH
,
Tuan
RS
.
Mechanical homeostasis is altered in uterine leiomyoma
.
Am J Obstet Gynecol
2008
;
4
:
474. e1
474. e11
.

Rohani
MG
,
Parks
WC
.
Matrix remodeling by MMPs during wound repair
.
Matrix Biol
2015
;
44-46
:
113
121
.

Ruwhof
C
,
van der Laarse
A
.
Mechanical stress-induced cardiac hypertrophy: mechanisms and signal transduction pathways
.
Cardiovasc Res
2000
;
1
:
23
37
.

Saksela
O
,
Moscatelli
D
,
Sommer
A
,
Rifkin
DB
.
Endothelial cell-derived heparan sulfate binds basic fibroblast growth factor and protects it from proteolytic degradation
.
J Cell Biol
1988
;
2
:
743
751
.

Salama
SA
,
Diaz-Arrastia
CR
,
Kilic
GS
,
Kamel
MW
.
2-Methoxyestradiol causes functional repression of transforming growth factor β3 signaling by ameliorating Smad and non-Smad signaling pathways in immortalized uterine fibroid cells
.
Fertil Steril
2012
;
1
:
178
184. e1
.

Salama
SA
,
Nasr
AB
,
Dubey
RK
,
Al-Hendy
A
.
Estrogen metabolite 2-methoxyestradiol induces apoptosis and inhibits cell proliferation and collagen production in rat and human leiomyoma cells: a potential medicinal treatment for uterine fibroids
.
J Soc Gynecol Investig
2006
;
8
:
542
550
.

Sanci
M
,
Dikis
C
,
Inan
S
,
Turkoz
E
,
Dicle
N
,
Ispahi
C
.
Immunolocalization of VEGF, VEGF receptors, EGF-R and Ki-67 in leiomyoma, cellular leiomyoma and leiomyosarcoma
.
Acta Histochem
2011
;
3
:
317
325
.

Santulli
P
,
Borghese
B
,
Lemaréchal
H
,
Leconte
M
,
Millischer
A-E
,
Batteux
F
,
Chapron
C
,
Borderie
D
.
Increased serum oxidative stress markers in women with uterine leiomyoma
.
PloS one
2013
;
8
:
e72069
.

Scaffidi
AK
,
Petrovic
N
,
Moodley
YP
,
Fogel-Petrovic
M
,
Kroeger
KM
,
Seeber
RM
,
Eidne
KA
,
Thompson
PJ
,
Knight
DA
.
αvβ3 integrin interacts with the transforming growth factor β (TGFβ) type II receptor to potentiate the proliferative effects of TGFβ1 in living human lung fibroblasts
.
J Biol Chem
2004
;
36
:
37726
37733
.

Schenk
S
,
Hintermann
E
,
Bilban
M
,
Koshikawa
N
,
Hojilla
C
,
Khokha
R
,
Quaranta
V
.
Binding to EGF receptor of a laminin-5 EGF-like fragment liberated during MMP-dependent mammary gland involution
.
J Cell Biol
2003
;
1
:
197
209
.

Schlaepfer
DD
,
Mitra
SK
.
Multiple connections link FAK to cell motility and invasion
.
Curr Opin Genet Dev
2004
;
1
:
92
101
.

Schmidt
G
,
Hausser
H
,
Kresse
H
.
Interaction of the small proteoglycan decorin with fibronectin. Involvement of the sequence NKISK of the core protein
.
Biochem J
1991
;
280
:
411
414
.

Schneller
M
,
Vuori
K
,
Ruoslahti
E
.
αvβ3 integrin associates with activated insulin and PDGFβ receptors and potentiates the biological activity of PDGF
.
EMBO J
1997
;
18
:
5600
5607
.

Seo
D-W
,
Li
H
,
Guedez
L
,
Wingfield
PT
,
Diaz
T
,
Salloum
R
,
Wei
B-Y
,
Stetler-Stevenson
WG
.
TIMP-2 mediated inhibition of angiogenesis: an MMP-independent mechanism
.
Cell
2003
;
2
:
171
180
.

Seong
J
,
Wang
N
,
Wang
Y
.
Mechanotransduction at focal adhesions: from physiology to cancer development
.
J Cell Mol Med
2013
;
5
:
597
604
.

Serini
G
,
Bochaton-Piallat
M-L
,
Ropraz
P
,
Geinoz
A
,
Borsi
L
,
Zardi
L
,
Gabbiani
G
.
The fibronectin domain ED-A is crucial for myofibroblastic phenotype induction by transforming growth factor-β1
.
J Cell Biol
1998
;
3
:
873
881
.

Shen
Q
,
Hua
Y
,
Jiang
W
,
Zhang
W
,
Chen
M
,
Zhu
X
.
Effects of mifepristone on uterine leiomyoma in premenopausal women: a meta-analysis
.
Fertil Steril
2013
;
6
:
1722
1726. e10
.

Shi
Y
,
Massagué
J
.
Mechanisms of TGF-β signaling from cell membrane to the nucleus
.
Cell
2003
;
6
:
685
700
.

Shime
H
,
Kariya
M
,
Orii
A
,
Momma
C
,
Kanamori
T
,
Fukuhara
K
,
Kusakari
T
,
Tsuruta
Y
,
Takakura
K
,
Nikaido
T
.
Tranilast inhibits the proliferation of uterine leiomyoma cells in vitro through G1 arrest associated with the induction of p21waf1 and p53
.
J Clin Endocrinol Metab
2002
;
12
:
5610
5617
.

Shull
MM
,
Ormsby
I
,
Kier
AB
,
Pawlowskr
S
,
Diebold
RJ
,
Yin
M
,
Allen
R
,
Sidman
C
,
Proetzel
G
,
Calvint
D
.
Targeted disruption of the mouse transforming growth factor-β1 gene results in multifocal inflammatory disease
.
Nature
1992
;
6397
:
693
699
.

Smith
J
,
Ockleford
CD
.
Laser scanning confocal examination and comparison of nidogen (entactin) with laminin in term human amniochorion
.
Placenta
1994
;
1
:
95
106
.

Solomon
SD
,
McMurray
JJV
,
Pfeffer
MA
,
Wittes
J
,
Fowler
R
,
Finn
P
,
Anderson
WF
,
Zauber
A
,
Hawk
E
,
Bertagnolli
M
.
Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention
.
N Engl J Med
2005
;
11
:
1071
1080
.

Sounni
NE
,
Devy
L
,
Hajitou
A
,
Frankenne
F
,
Munaut
C
,
Gilles
C
,
Deroanne
C
,
Thompson
EW
,
Foidart
J-M
,
Noël
A
.
MT1-MMP expression promotes tumor growth and angiogenesis through an up-regulation of vascular endothelial growth factor expression
.
FASEB J
2002
;
6
:
555
564
.

Sounni
NE
,
Noël
A
.
Membrane type-matrix metalloproteinases and tumor progression
.
Biochimie
2005
;
3
:
329
342
.

Steinauer
J
,
Pritts
EA
,
Jackson
R
,
Jacoby
AF
.
Systematic review of mifepristone for the treatment of uterine leiomyomata
.
Obstet Gynecol
2004
;
6
:
1331
.

Stewart
EA
,
Friedman
AJ
,
Peck
K
,
Nowak
RA
.
Relative overexpression of collagen type I and collagen type III messenger ribonucleic acids by uterine leiomyomas during the proliferative phase of the menstrual cycle
.
J Clin Endocrinol Metab
1994
;
3
:
900
906
.

Stovall
TG
,
Ling
FW
,
Henry
LC
,
Woodruff
MR
.
A randomized trial evaluating leuprolide acetate before hysterectomy as treatment for leiomyomas
.
Am J Obstet Gynecol
1991
;
6 Pt 1
:
1420
1423
. ; discussion 1423-5.

Stovall
TG
,
Muneyyirci-Delale
O
,
Summitt
RL
Jr
,
Scialli
AR
.
GnRH agonist and iron versus placebo and iron in the anemic patient before surgery for leiomyomas: a randomized controlled trial. The Leuprolide Study Group
.
Obstet Gynecol
1995
;
1
:
65
71
.

Stracke
JO
,
Hutton
M
,
Stewart
M
,
Pendás
AM
,
Smith
B
,
López-Otin
C
,
Murphy
G
,
Knäuper
V
.
Biochemical characterization of the catalytic domain of human matrix metalloproteinase 19. Evidence for a role as a potent basement membrane degrading enzyme
.
J Biol Chem
2000
;
20
:
14809
14816
.

Strand
S
,
Vollmer
P
,
van den Abeelen
L
,
Gottfried
D
,
Alla
V
,
Heid
H
,
Kuball
J
,
Theobald
M
,
Galle
PR
,
Strand
D
.
Cleavage of CD95 by matrix metalloproteinase-7 induces apoptosis resistance in tumour cells
.
Oncogene
2004
;
20
:
3732
3736
.

Sun
Q
,
Weber
CR
,
Sohail
A
,
Bernardo
MM
,
Toth
M
,
Zhao
H
,
Turner
JR
,
Fridman
R
.
MMP25 (MT6-MMP) is highly expressed in human colon cancer, promotes tumor growth, and exhibits unique biochemical properties
.
J Biol Chem
2007
;
30
:
21998
22010
.

Suo
G
,
Jiang
Y
,
Cowan
B
,
Wang
JY
.
Platelet-derived growth factor C is upregulated in human uterine fibroids and regulates uterine smooth muscle cell growth
.
Biol Reprod
2009
;
4
:
749
758
.

Suzuki
M
,
Raab
G
,
Moses
MA
,
Fernandez
CA
,
Klagsbrun
M
.
Matrix metalloproteinase-3 releases active heparin-binding EGF-like growth factor by cleavage at a specific juxtamembrane site
.
J Biol Chem
1997
;
50
:
31730
31737
.

Syssoev
KA
,
Kulagina
NV
,
Chukhlovin
AB
,
Morozova
EB
,
Totolian
AA
.
Expression of mRNA for chemokines and chemokine receptors in tissues of the myometrium and uterine leiomyoma
.
Bull Exp Biol Med
2008
;
1
:
84
89
.

Tang
XM
,
Dou
Q
,
Zhao
Y
,
McLean
F
,
Davis
J
,
Chegini
N
.
The expression of transforming growth factor-beta s and TGF-β receptor mRNA and protein and the effect of TGF-βs on human myometrial smooth muscle cells in vitro
.
Mol Hum Reprod
1997
;
3
:
233
240
.

Taniguchi
Y
,
Morita
I
,
Kubota
T
,
Murota
S
,
Aso
T
.
Human uterine myometrial smooth muscle cell proliferation and vascular endothelial growth-factor production in response to platelet-derived growth factor
.
J Endocrinol
2001
;
1
:
79
86
.

Thorne
JT
,
Segal
TR
,
Chang
S
,
Jorge
S
,
Segars
JH
,
Leppert
PC
.
Dynamic reciprocity between cells and their microenvironment in reproduction
.
Biol Reprod
2015
;
1
:
25
.

Tomasek
JJ
,
Gabbiani
G
,
Hinz
B
,
Chaponnier
C
,
Brown
RA
.
Myofibroblasts and mechano-regulation of connective tissue remodelling
.
Nat Rev Mol Cell Biol
2002
;
5
:
349
363
.

Toullec
A
,
Gerald
D
,
Despouy
G
,
Bourachot
B
,
Cardon
M
,
Lefort
S
,
Richardson
M
,
Rigaill
G
,
Parrini
MC
,
Lucchesi
C
.
Oxidative stress promotes myofibroblast differentiation and tumour spreading
.
EMBO Mol Med
2010
;
6
:
211
230
.

Tsigkou
A
,
Reis
FM
,
Ciarmela
P
,
Lee
MH
,
Jiang
B
,
Tosti
C
,
Shen
FR
,
Shi
Z
,
Chen
YG
,
Petraglia
F
.
Expression levels of myostatin and matrix metalloproteinase 14 mRNAs in uterine leiomyoma are correlated with dysmenorrhea
.
Reprod Sci
2015
;
12
:
1597
1602
.

Turner
RS
,
Thomas
RG
,
Craft
S
,
Van Dyck
CH
,
Mintzer
J
,
Reynolds
BA
,
Brewer
JB
,
Rissman
RA
,
Raman
R
,
Aisen
PS
.
A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease
.
Neurology
2015
;
16
:
1383
1391
.

Velasco
G
,
Cal
S
,
Merlos-Suárez
A
,
Ferrando
AA
,
Alvarez
S
,
Nakano
A
,
Arribas
J
,
López-Otín
C
.
Human MT6-matrix metalloproteinase: identification, progelatinase A activation, and expression in brain tumors
.
Cancer Res
2000
;
4
:
877
882
.

Wang
H
,
Lafdil
F
,
Wang
L
,
Yin
S
,
Feng
D
,
Gao
B
.
Tissue inhibitor of metalloproteinase 1 (TIMP-1) deficiency exacerbates carbon tetrachloride-induced liver injury and fibrosis in mice: involvement of hepatocyte STAT3 in TIMP-1 production
.
Cell Biosci
2011
;
1
:
14
.

Wang
J
,
Ohara
N
,
Wang
Z
,
Chen
W
,
Morikawa
A
,
Sasaki
H
,
DeManno
DA
,
Chwalisz
K
,
Maruo
T
.
A novel selective progesterone receptor modulator asoprisnil (J867) down-regulates the expression of EGF, IGF-I, TGFβ3 and their receptors in cultured uterine leiomyoma cells
.
Hum Reprod
2006
;
7
:
1869
1877
.

Wang
T
,
Zhang
X
,
Obijuru
L
,
Laser
J
,
Aris
V
,
Lee
P
,
Mittal
K
,
Soteropoulos
P
,
Wei
JJ
.
A micro-RNA signature associated with race, tumor size, and target gene activity in human uterine leiomyomas
.
Genes Chromosomes Cancer
2007
;
4
:
336
347
.

Wang
Y
,
Feng
G
,
Wang
J
,
Zhou
Y
,
Liu
Y
,
Shi
Y
,
Zhu
Y
,
Lin
W
,
Xu
Y
,
Li
Z
.
Differential effects of tumor necrosis factor-α on matrix metalloproteinase-2 expression in human myometrial and uterine leiomyoma smooth muscle cells
.
Hum Reprod
2015
;
1
:
61
70
.

Webb
AH
,
Gao
BT
,
Goldsmith
ZK
,
Irvine
AS
,
Saleh
N
,
Lee
RP
,
Lendermon
JB
,
Bheemreddy
R
,
Zhang
Q
,
Brennan
RC
.
Inhibition of MMP-2 and MMP-9 decreases cellular migration, and angiogenesis in in vitro models of retinoblastoma
.
BMC cancer
2017
;
1
:
434
.

Wegienka
G
.
Are uterine leiomyoma a consequence of a chronically inflammatory immune system?
Med Hypotheses
2012
;
2
:
226
231
.

Werner
S
,
Hubner
G
,
Alzheimer
C
.
Activin: a novel player in tissue repair processes
.
Histol Histopathol
1999
;
1
:
295
304
.

Wight
TN
.
Versican: a versatile extracellular matrix proteoglycan in cell biology
.
Curr Opin Cell Biol
2002
;
5
:
617
623
.

Wijelath
ES
,
Rahman
S
,
Namekata
M
,
Murray
J
,
Nishimura
T
,
Mostafavi-Pour
Z
,
Patel
Y
,
Suda
Y
,
Humphries
MJ
,
Sobel
M
.
Heparin-II domain of fibronectin is a vascular endothelial growth factor-binding domain enhancement of VEGF biological activity by a singular growth factor/matrix protein synergism
.
Circ Res
2006
;
8
:
853
860
.

Williams
AR
,
Critchley
HO
,
Osei
J
,
Ingamells
S
,
Cameron
IT
,
Han
C
,
Chwalisz
K
.
The effects of the selective progesterone receptor modulator asoprisnil on the morphology of uterine tissues after 3 months treatment in patients with symptomatic uterine leiomyomata
.
Hum Reprod
2007
;
6
:
1696
1704
.

Wipff
P-J
,
Rifkin
DB
,
Meister
J-J
,
Hinz
B
.
Myofibroblast contraction activates latent TGF-β1 from the extracellular matrix
.
J Cell Biol
2007
;
6
:
1311
1323
.

Wolanska
M
,
Bankowski
E
.
Fibroblast growth factors (FGF) in human myometrium and uterine leiomyomas in various stages of tumour growth
.
Biochimie
2006
;
2
:
141
146
.

Wolanska
M
,
Sobolewski
K
,
Bańkowski
E
,
Jaworski
S
.
Matrix metalloproteinases of human leiomyoma in various stages of tumor growth
.
Gynecol Obstet Invest
2004
;
1
:
14
18
.

Wolanska
M
,
Sobolewski
K
,
Drożdżewicz
M
,
Bańkowski
E
.
Extracellular matrix components in uterine leiomyoma and their alteration during the tumour growth
.
Mol Cell Biochem
1998
;
1
:
145
152
.

Wortham
NC
,
Alam
NA
,
Barclay
E
,
Pollard
PJ
,
Wagner
BE
,
Manek
S
,
Elia
G
,
Tomlinson
IPM
.
Aberrant expression of apoptosis proteins and ultrastructural aberrations in uterine leiomyomas from patients with hereditary leiomyomatosis and renal cell carcinoma
.
Fertil Steril
2006
;
4
:
961
971
.

Wu
C-H
,
Shieh
T-M
,
Wei
L-H
,
Cheng
T-F
,
Chen
H-Y
,
Huang
T-C
,
Wang
K-L
,
Hsia
S-M
.
Resveratrol inhibits proliferation of myometrial and leiomyoma cells and decreases extracellular matrix-associated protein expression
.
J Funct Foods
2016
;
23
:
241
252
.

Wynn
TA
.
Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases
.
J Clin Invest
2007
;
3
:
524
529
.

Wynn
TA
.
Cellular and molecular mechanisms of fibrosis
.
J Pathol
2008
;
2
:
199
210
.

Xing
Z
,
Tremblay
GM
,
Sime
PJ
,
Gauldie
J
.
Overexpression of granulocyte-macrophage colony-stimulating factor induces pulmonary granulation tissue formation and fibrosis by induction of transforming growth factor-β1 and myofibroblast accumulation
.
Am J Pathol
1997
;
1
:
59
66
.

Xu
J
,
Luo
X
,
Chegini
N
.
Differential expression, regulation, and induction of Smads, transforming growth factor-β signal transduction pathway in leiomyoma, and myometrial smooth muscle cells and alteration by gonadotropin-releasing hormone analog
.
J Clin Endocrinol Metab
2003
;
3
:
1350
1361
.

Xu
Q
,
Ohara
N
,
Liu
J
,
Amano
M
,
Sitruk-Ware
R
,
Yoshida
S
,
Maruo
T
.
Progesterone receptor modulator CDB-2914 induces extracellular matrix metalloproteinase inducer in cultured human uterine leiomyoma cells
.
Mol Hum Reprod
2008
;
3
:
181
191
.

Yamaguchi
Y
,
Mann
DM
,
Ruoslahti
E
.
Negative regulation of transforming growth factor-β by the proteoglycan decorin
.
Nature
1990
;
6281
:
281
284
.

Yamashita
CM
,
Dolgonos
L
,
Zemans
RL
,
Young
SK
,
Robertson
J
,
Briones
N
,
Suzuki
T
,
Campbell
MN
,
Gauldie
J
,
Radisky
DC
.
Matrix metalloproteinase 3 is a mediator of pulmonary fibrosis
.
Am J Pathol
2011
;
4
:
1733
1745
.

Yang
Q
,
Mas
A
,
Diamond
MP
,
Al-Hendy
A
.
The mechanism and function of epigenetics in uterine leiomyoma development
.
Reprod Sci
2016
;
2
:
163
175
.

Yayon
A
,
Klagsbrun
M
,
Esko
JD
,
Leder
P
,
Ornitz
DM
.
Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor
.
Cell
1991
;
4
:
841
848
.

Yerushalmi
GM
,
Gilboa
Y
,
Jakobson-Setton
A
,
Tadir
Y
,
Goldchmit
C
,
Katz
D
,
Seidman
DS
.
Vaginal mifepristone for the treatment of symptomatic uterine leiomyomata: an open-label study
.
Fertil Steril
2014
;
2
:
496
500
.

Yin
P
,
Lin
Z
,
Reierstad
S
,
Wu
J
,
Ishikawa
H
,
Marsh
EE
,
Innes
J
,
Cheng
Y
,
Pearson
K
,
Coon
JS
.
Transcription factor KLF11 integrates progesterone receptor signaling and proliferation in uterine leiomyoma cells
.
Cancer Res
2010
;
4
:
1722
1730
.

Yin
P
,
Ono
M
,
Moravek
MB
,
Coon
JS
,
Navarro
A
,
Monsivais
D
,
Dyson
MT
,
Druschitz
SA
,
Malpani
SS
,
Serna
VA
.
Human uterine leiomyoma stem/progenitor cells expressing CD34 and CD49b initiate tumors in vivo
.
J Clin Endocrinol Metab
2015
;
4
:
E601
E606
.

Yoshiji
H
,
Kuriyama
S
,
Miyamoto
Y
,
Thorgeirsson
UP
,
Gomez
DE
,
Kawata
M
,
Yoshii
J
,
Ikenaka
Y
,
Noguchi
R
,
Tsujinoue
H
.
Tissue inhibitor of metalloproteinases‐1 promotes liver fibrosis development in a transgenic mouse model
.
Hepatology
2000
;
6
:
1248
1254
.

Yu
L
,
Saile
K
,
Swartz
CD
,
He
H
,
Zheng
X
,
Kissling
GE
,
Di
X
,
Lucas
S
,
Robboy
SJ
,
Dixon
D
.
Differential expression of receptor tyrosine kinases (RTKs) and IGF-I pathway activation in human uterine leiomyomas
.
Mol Med
2008
;
5-6
:
264
275
.

Zbucka
M
,
Miltyk
W
,
Bielawski
T
,
Surazynski
A
,
Palka
J
,
Wolczynski
S
.
Mechanism of collagen biosynthesis up-regulation in cultured leiomyoma cells
.
Folia Histochem Cytobiol
2008
;
45
:
S181
S185
.

Zhang
X
,
Huang
S
,
Guo
J
,
Zhou
L
,
You
L
,
Zhang
T
,
Zhao
Y
.
Insights into the distinct roles of MMP-11 in tumor biology and future therapeutics
.
Int J Oncol
2016
;
5
:
1783
1793
.

Zhao
X-H
,
Laschinger
C
,
Arora
P
,
Szászi
K
,
Kapus
A
,
McCulloch
CA
.
Force activates smooth muscle α-actin promoter activity through the Rho signaling pathway
.
J Cell Sci
2007
;
10
:
1801
1809
.

Zheng
L-H
,
Cai
F-F
,
Ge
I
,
Biskup
E
,
Cheng
Z-P
.
Stromal fibroblast activation and their potential association with uterine fibroids (Review)
.
Oncol Lett
2014
;
2
:
479
486
.