Sei sulla pagina 1di 14

2015. Published by The Company of Biologists Ltd | Development (2015) 142, 4191-4204 doi:10.1242/dev.

114777

REVIEW

Tendon development and musculoskeletal assembly: emerging


roles for the extracellular matrix
Arul Subramanian and Thomas F. Schilling*

ABSTRACT How are these differences established? The answer to this question
Tendons and ligaments are extracellular matrix (ECM)-rich structures has important implications for understanding how diseases or
that interconnect muscles and bones. Recent work has shown how damage to tendons and ligaments arise and for developing better
tendon fibroblasts (tenocytes) interact with muscles via the ECM to treatment strategies.
establish connectivity and strengthen attachments under tension. Despite their pivotal roles in musculoskeletal connectivity and
Similarly, ECM-dependent interactions between tenocytes and functional stability, the mechanisms that control tendon
cartilage/bone ensure that tendon-bone attachments form with the development have received much less attention than the processes
appropriate strength for the force required. Recent studies have also of myogenesis or skeletogenesis. Only a handful of factors are
established a close lineal relationship between tenocytes and skeletal known to help specify tenocyte progenitor cells (TPCs) at muscle
progenitors, highlighting the fact that defects in signals modulated by attachments, induce them to differentiate, and maintain and repair
the ECM can alter the balance between these fates, as occurs in them in the adult (Aslan et al., 2008; Huang et al., 2015; Liu et al.,
calcifying tendinopathies associated with aging. The dynamic fine- 2012, 2014; Schweitzer et al., 2010; Yang et al., 2013). Studies in
tuning of tendon ECM composition and assembly thus gives rise to animal models (e.g. fly, fish, chick and mouse embryos), in
the remarkable characteristics of this unique tissue type. Here, we particular those focusing on the formation of myotendinous
provide an overview of the functions of the ECM in tendon formation junctions (MTJs; the major sites of force transmission), have
and maturation that attempts to integrate findings from developmental revealed a crucial link between developing TPCs and the dynamic
genetics with those of matrix biology. ECM that surrounds these cells. Indeed, ECM proteins (e.g.
collagens, laminins, thrombospondins) initially guide myofibers to
KEY WORDS: Tendon, Ligament, Tenocyte, Extracellular matrix their sites of attachment, but also mediate signaling between TPCs
and muscles, regulate the maturation of MTJs, and maintain tendons
Introduction in response to mechanical force (Kjaer, 2004; Schwartz et al., 2013;
Tendons and ligaments are connective tissues that transmit Snow and Henry, 2009). Here, we review recent genetic studies that
mechanical forces between muscles and bones. Tendons attach have identified crucial roles for the ECM in tendon development,
muscle to skeleton, whereas ligaments attach skeletal elements to and we discuss the emerging nexus between the transcriptional
each other and stabilize skeletal joints. Vertebrates have evolved a control of tenocyte differentiation and the organization of the ECM
remarkable variety of tendons and ligaments to accommodate their associated with muscle fibers (myomatrix), MTJs and tendons
distinct modes of locomotion as well as their dramatic variations in (Fig. 1).
body size and strength. These range from broad sheets to highly
elastic cables, such as those of the Achilles tendon and the cruciate ECM production and regulation in developing tendons
ligaments of the knee. Because of their structural roles, injuries to Collagens ( predominantly Col1a) constitute the bulk of mature
tendons and ligaments are extremely common and often mammalian tendon ECM and MTJs, whereas laminins (Lams) and
debilitating. Thus, a fundamental question in musculoskeletal many other non-collagenous ECM components comprise the
biology is how these connective tissue structures develop in the remainder (Kannus et al., 1998; Kannus, 2000; Kjaer, 2004;
correct locations and acquire the strength necessary to translate Birch et al., 2013; Thorpe et al., 2013). Which cells secrete these
contractions of muscles into skeletal movements. ECM proteins, how are they produced in the correct proportions,
Both tendons and ligaments contain fibroblasts (termed tenocytes and how do they assemble?
and ligamentocytes, respectively) embedded in a unique
extracellular matrix (ECM) that is composed mainly of collagen The transcriptional regulation of ECM production by TPCs
fibril arrays capable of withstanding incredibly strong tensile forces. Strikingly, all of the key transcription factors known to function in
These fibrils are crosslinked to one another and wrapped in a tendon TPC development directly regulate the transcription of genes
sheath (Banos et al., 2008; Kannus et al., 1998; Ros et al., 1995). encoding ECM proteins (Fig. 2A,B). The best studied of these is
This basic structure is shared among tendons and ligaments, and the Scleraxis (Scx), a basic helix-loop-helix transcription factor, and the
DEVELOPMENT

fibroblasts that produce the ECM develop from common earliest known marker of TPCs. Scx is first induced through the
progenitors with similar gene expression profiles (Juneja and interplay of sonic hedgehog (Shh) and fibroblast growth factor
Veillette, 2013; Sugimoto et al., 2013; Tozer and Duprez, 2005; (FGF) signaling in the syndetome compartment of somites (the
Yang et al., 2013). However, each tendon or ligament differs in its region of the sclerotome adjacent to the myotome) and by
precise ECM composition, size and strength (Birch et al., 2013). transforming growth factor beta (TGF) signaling in the limbs of
mice at embryonic day (E) 10.5 (Schweitzer et al., 2001; Brent et al.,
Department of Developmental and Cell Biology, University of California, Irvine, 2003; Havis et al., 2014). In vitro, Scx overexpression is sufficient to
Irvine, CA 92697-2300, USA.
transform mesenchymal stem cells (MSCs) and human embryonic
*Author for correspondence (tschilli@uci.edu) stem cells (hESCs) into tenocytes (Fig. 2A) (Alberton et al., 2012;

4191
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

MTJ downregulated in Scx / mutants (Murchison et al., 2007;


Myomatrix Tendon matrix
Fn, Lam, Col, MMP,TIMP, Tsp SLRP, Col, Lam,Tsp, MMP, TIMP
Shukunami et al., 2006). At the ultrastructural level, the loss of
Scx disrupts the sheaths that surround each fascicle of collagen
fibrils as well as cellular processes, which normally encircle the
fibrils (Murchison et al., 2007). These results demonstrate that Scx
controls tendon ECM production, which is essential for effective
Tenocyte force transmission.
Muscle
fiber
It should be noted that TPCs still develop in Scx / mutant mice,
suggesting that other genes are required for the initial steps of TPC
specification. In Drosophila, the transcription factor Stripe (Sr)
specifies TPCs in epidermal segment border cells (Volk and
VijayRaghavan, 1994). Flies lacking Sr function fail to form TPCs
and display disrupted muscle patterning and attachments, whereas
Key Sr overexpression transforms ectodermal progenitors into TPCs
Laminin Fibronectin Collagen Thrombospondin
(Lam) (Fn) (Col) (Tsp) (Becker et al., 1997). Embryonic TPCs in mice express orthologs
of Sr Egr1 and Egr2 (Lejard et al., 2011) and Egr1 is sufficient
Fig. 1. Composition of the ECM surrounding muscle, tendon and to induce Scx expression and specify MSCs as tenocytes in vitro
myotendinous junctions. A muscle fiber (green) secretes ECM components (Guerquin et al., 2013). However, like Scx, both Egr1 and
into its surroundings (the myomatrix). Some of these components overlap with
Egr2 appear to be dispensable for tenocyte specification, as
those of the tendon ECM, which is secreted by tenocytes (red). Myomatrix is
primarily composed of Lam trimers and Fn. By contrast, the tendon
Egr1 //Egr2 / double mutant mice are viable. Instead they
matrix is rich in Col1a trimers and thrombospondin pentamers. The regulate the tendon ECM and MTJ, binding to tendon-specific
myotendinous junction (MTJ) is the narrow zone in which ECM components of enhancer elements of Col1a1 and Col1a2 that are also bound by Scx
tendon and muscle interact. (Fig. 2B) (Ljard et al., 2007, 2011; Guerquin et al., 2013). Egr1 /
mutant mice also downregulate Tnmd and are slow to heal tendon
Chen et al., 2012; Li et al., 2015). Thus, considerable effort has been injuries as adults (Guerquin et al., 2013). Thus, in contrast to flies,
made to elucidate the functions of Scx in the tenocyte lineage and to vertebrate Egrs function in tendon ECM production rather than TPC
identify its downstream targets. specification.
The loss of Scx (i.e. as in Scx / mutant mice) disrupts tenocyte Another potential TPC specifier is the TALE family atypical
differentiation leading to atrophy of force-transmitting tendons Iroquois-like homeodomain protein Mohawk (Mkx). Like Scx and
and a disorganized tendon ECM (Murchison et al., 2007). The Egr1, Mkx can drive bone marrow-derived MSCs towards a
expression of the major structural collagens, Col1a1, Col1a2, tenocyte fate in vitro (Liu et al., 2015; Otabe et al., 2015).
Col3a1 and Col14a1, is strongly reduced in Scx / mutants. Scx However, in mouse embryos the expression of Mkx begins in
directly controls Col1a1 and Col1a2 transcription (Fig. 2B) (Espira developing tenocytes later than that of Scx or Egr1/2 (at E13.5-
et al., 2009; Ljard et al., 2007). Numerous other tendon regulators, 14.5), becoming restricted to tendon sheath cells and collateral
such as the glycoprotein tenomodulin (Tnmd), are also ligaments (which stabilize joints) in the limbs by E16.5 (Anderson

A Tenocyte specification B Tendon-specific ECM gene regulation


Col1a1 Tgfb2
Col1a2 Col1a1
MSC Tnmd Col1a2
Col3a1 Tnmd
Shh, FGF,TGF Col14a1 Fmod
BMP (Axial) Smad3 Scx Tsp4 Smad3 Mkx Dcn
FGF (Mouse limb) Col1a1
Col1a2 Sox6
Tgfb2
SPC Egr1 Tnmd Smad3 Myod
TPC Bgn Mkx Runx
Sox5/6/9 Egr2
Scx Scx Tgfbr2
Sox9

C Skeletal-specific ECM gene regulation


ChM-1
Col2a1
Scx Mkx
Col9a2
Tnmd Col11a2
Runx2 Egr1 Mmp9
Agg
Comp Mmp13
Osteoblast Tenocyte Runx2 Tram2
Sox9 Runx2
DEVELOPMENT

Fig. 2. Transcriptional regulation of tenocyte specification and tendon ECM production. (A) A mesenchymal stem cell (MSC, purple), the common
progenitor for skeletal and tenocyte progenitors, becomes a skeletogenic progenitor cell (SPC, blue) if it is exposed to high levels of BMP signaling, then it
expresses Sox5/6/9 followed by Runx2 during its differentiation into an osteoblast. By contrast, an MSC becomes a tendon progenitor cell (TPC, pink) if it receives
high levels of Shh, FGF and TGF signaling, then expresses scleraxis (Scx) followed by Mkx, Egr1 and Tnmd during its transition into a tenocyte. The fate of the
progenitor cells is determined by the level of Sox9 and Scx. The plasticity of progenitor cell fate at this stage is represented by the double-headed gray arrow.
(B) The transcription factors involved in tenocyte specification also regulate the transcription of genes encoding ECM proteins. Direct (black) and indirect (gray)
transcriptional target genes regulated by Scx, Egr1, Egr2 and Mkx in TPCs are indicated. Note that Mkx also represses the expression of factors involved in
myogenic and skeletogenic progenitor formation. (C) Transcription factors expressed in skeletogenic progenitor cells (e.g. Sox9 and Runx2) directly regulate the
transcription of a distinct set of ECM target genes.

4192
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

A Tendon-independent phase Fig. 3. Myoblast-tenocyte interactions and ECM production. (A,B)


EC The formation of myotendinous junctions can be considered as a two-
E CM M step process. In the initial tendon-independent phase (A) in vertebrates
TPC (shown here for zebrafish trunk muscles), myoblasts (green) synthesize a
Myoblast pre-tendon ECM that includes the integrin ligands Tsp4 and Lama2. This

3
Scx, Mkx

Smad
ECM accumulates in the absence of TPCs (brown).
MyoD Sox6,
Runx2 Mechanotransduction coupled with TGF signaling (through Tgf2 and
Sox9
Tgfr2) leads to the Smad3-dependent expression of Scx and Mkx in
TPCs, which in turn leads to the expression of tendon-selective ECM
genes. Smad3 and Mkx also repress the activity of MyoD, Sox9 and
Runx2 to repress myogenic and skeletogenic fates during tenocyte
EC differentiation. A later tendon-dependent phase (B) relies on the
B Tendon-dependent phase M production of ECM, particularly Col1a1, Col1a2, Col12a1 and Col14a1,
by more mature TPCs, which extend processes into the ECM.
Myoblast TPC
Scx, Mkx
Egr1, Egr2

Key
FACIT Col Fn Col1a1 Tgfr2 Tsp4 Tgf2
Col12a1 Col1a2 Alk Lama2
Itg

et al., 2006). Mkx / mutant mice are viable, fertile, and form transcriptional regulators of tenocytes share functions in the
normal tendons at first with no defects in Scx expression but later production of tendon ECM and MTJ assembly and in the
exhibit reduced levels of Col1a1, Col1a2, Tnmd, fibromodulin repression of other mesenchymal fates. This is important not only
(Fmod) and decorin (Dcn) as well as thinning of collagen fibrils (Ito in the context of tenocyte development but also in the regulation of
et al., 2010; Kimura et al., 2011; Liu et al., 2010). Similar to Scx, MSCs, where the balance between these factors determines if a cell
Mkx can function as a transcriptional activator when complexed becomes a TPC or a skeletogenic (or myogenic) progenitor
with Smad2/3 to promote Col1a1, Col1a2, Tnmd and Dcn (Fig. 2A). An attractive model is one in which Scx (in concert
expression as well as TGF2 expression in murine MSCs (Liu with TGF signaling) shifts the fate of MSCs towards TPCs and
et al., 2015). However, at other promoters it interacts with the Sin3A/ initiates tendon ECM production, whereas factors expressed later in
histone deacetylase (HDAC) complex to repress gene expression, development, such as Mkx and Egr1, supplement the role of Scx by
including that of key myogenic factors such as MyoD (Myod1), inducing the expression of ECM proteins as well as by repressing
Sox6 and the cartilage determinant Sox9 (Fig. 2B) (Anderson et al., myogenic and skeletogenic fates. Furthermore, signaling mediated
2009; Anderson et al., 2012; Berthet et al., 2013; Chuang et al., by mechanical forces upregulates expression of Scx, Mkx and
2014). Thus, like Scx and Egr1/2, Mkx controls tendon maturation Smad3, stimulating more ECM production, thereby providing
and ECM production and might function, in part, to maintain positive feedback for fine-tuning tendon strength, as we discuss
tenocytes by preventing them from acquiring myogenic or further below (Eliasson et al., 2008; Maeda et al., 2011).
skeletogenic fates.
Both Scx and Mkx interact with Smad3, an essential ECM production and function during tenocyte morphogenesis and
transcriptional mediator of TGF signaling, to regulate tendon MTJ formation
ECM production (Berthet et al., 2013; Hosokawa et al., 2010; Although the migration of muscle progenitors has been well studied
Katzel et al., 2011; Oka et al., 2008; Pryce et al., 2009). in both invertebrates and vertebrates, very little is known about the
Accordingly, Tgfb2//Tgfb3/ conditional double mutant mice, role of ECM in morphogenesis of TPCs and the establishment of the
or conditional mutant mice lacking Tgfr2 receptors in limb MTJ. In Drosophila, myoblasts migrate to sites of attachment and
mesenchyme, initially form TPCs in the limbs but lose them by interact with tenocytes located at fixed sites at segment borders in the
E14.5, suggesting a role for TGF signaling in tendon maintenance epidermis (Volk and VijayRaghavan, 1994; Schweitzer et al., 2010).
(Pryce et al., 2009). Tendon defects in Smad3/ mutants are much These myoblasts recognize tenocytes through multiple signals
less severe, with transient reductions in Col1a1, Col1a2 and Tnmd including Thrombospondin (Tsp) in the ECM, which binds muscle
expression in their limbs. Like Mkx, Smad3 can also inhibit the integrins (Itgs) (Subramanian et al., 2007). Similarly, vertebrate
expression and activity of MyoD as well as that of skeletogenic myoblasts in the trunk and limbs elongate and attach via TPCs already
DEVELOPMENT

factors such as Runx2 in vitro (Fig. 2A, Fig. 3) (Alliston et al., 2001; localized to future muscle attachment sites. How do migrating
Kang et al., 2005; Liu et al., 2001). However, unlike Mkx, which vertebrate myofibers and TPCs interact, and how do these interactions
represses MyoD transcription, Smad3 acts post-translationally by differ between the trunk, limb and head? In the chick, early progenitor
binding E-box sites in MyoD and sequestering it away from its pools of limb TPCs condense and split to form individual tendons
targets (Chuang et al., 2014). These results hint at a dynamic (Kardon, 1998). By contrast, cranial TPCs that arise in the neural crest
network involving TGF signaling, Scx and Mkx to achieve and migrate to the locations of future MTJs (Grenier et al., 2009; Noden,
maintain the tenocyte fate. 1988; Noden and Trainor, 2005). Trunk TPCs arise from the
In summary, to date no single factor fits the bill as being syndetome of somites, whereas limb TPCs are thought to originate
both necessary and sufficient for TPC specification. Rather, from lateral plate mesoderm (Brent et al., 2003; Kardon, 1998). These

4193
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

distinct embryonic origins and modes of tendon morphogenesis raise 2000; Jelinsky et al., 2010). Thus, Tsp4 in the myomatrix helps
the question of where the patterning information for muscle muscles attach in the absence of TPCs and might also facilitate
connectivity lies, within the TPCs or within the muscles subsequent tendon maintenance and response to damage.
themselves? Chick-quail chimera studies suggests that, at least for
cranial and limb muscles, the TPCs and tendon matrix determine the ECM and collagen fibril assembly during tendon and MTJ maturation
pattern of attachments (Kardon, 1998; Kieny and Chevallier, 1979; As MTJs mature, tenocytes secrete the bulk of the MTJ/tendon
Noden, 1988). Do muscles and the myomatrix play any role in TPC ECM, particularly the many proteins and proteoglycans that make
formation or maintenance? In the limbs of chick embryos in which up the core functional units, the collagen fibrils (Fig. 3B). These
muscle progenitors have been surgically removed, tenocytes initially interact with one another and align into groups of fibrils or fascicles
develop in the correct locations but later degenerate (Kardon, 1998). during progressive phases of muscle attachment (Fig. 4). This
Limb tendons also degenerate in MyoD/ and Pax3/ mutant mice fibrillar organization is essential for tendons to bear the stress of
that lack the entire limb musculature (Bonnin et al., 2005; Brent et al., muscle contraction and prevent bone detachment (avulsion
2005). Cranial tendons in myod1//myf5/ double mutant zebrafish fractures) by controlling force distribution (Birch et al., 2013; Pan
show similar defects (Chen and Galloway, 2014). These results et al., 2013; Schwartz et al., 2013; Pingel et al., 2014). The fibrillar
suggest a dependence on muscles and the myomatrix for tenocyte network of proteins includes: (1) core force-transmitting, structural
maintenance but not specification. As discussed below, these collagens ( particularly Col1a1, Col1a2, Col2a1 and Col3a1); (2)
phenotypes could be due to a lack of mechanical forces transmitted scaffolding proteins [e.g. Tsp2 (Thbs2), Tsp4, Comp, Lama2]; and
via the ECM. (3) specialized crosslinking collagens (e.g. Col6a1, Col12a1,
What are the roles of specific ECM components of the myomatrix Col14a1 and Col22a1) and various crosslinking factors [e.g. Dcn,
or tendon ECM during the initial establishment of contact between Fmod, biglycan (Bgn)], which hold fibrils together to distribute
myoblasts and TPCs at attachment sites? Studies of axial muscles in forces efficiently and reduce friction (Figs 3, 4) (Wang et al., 2012;
the zebrafish trunk have provided insights into this process (Snow and Dunkman et al., 2013). Although the integration of structural
Henry, 2009). These muscles attach to intersegmental boundaries collagens into fibrils has been well documented, recent studies have
(ISBs) during embryogenesis before the appearance of TPCs. ISBs provided insight into the functions of scaffolding proteins and
are anatomically distinct from later tendons, but contain many of the specialized collagens during fibril assembly. These studies suggest
same ECM components and serve analogous functions in bearing the that similar to the early ECM at developing MTJs, ECM proteins of
forces of muscle contraction. During this tendon-independent phase the maturing tendon provide continuous feedback in response to
of development (Fig. 3A), fibronectin (Fn) and laminin-alpha2 mechanical force (Choi et al., 2014; Popov et al., 2015; Wall and
(Lama2) in the myomatrix are highly enriched at ISBs as the Banes, 2005; Zhang and Wang, 2010).
myoblasts elongate and are required for embryonic muscle Col12a1, Col14a1 and Col22a1 belong to the class of fibril-
attachments (Koshida et al., 2005; Snow et al., 2008). Mammalian associated collagens with interrupted triple helices (FACITs), which
muscles also require Fn and Lam for migration and attachment localize to muscle attachments in avian and mouse tendons, and are
(Turner et al., 1983; Bajanca et al., 2006; Vaz et al., 2012). Fn and also expressed in human tendon fibroblasts (Fig. 3B; Fig. 4) (Koch
Lam bind to Itg and dystrophin/dystroglycan complexes on muscle et al., 2004; Wlchli et al., 1994). Studies of FACITs at zebrafish ISBs
cell surfaces. In zebrafish, this leads to localized phosphorylation of have been informative for understanding their functions as MTJs
focal adhesion kinase (pFAK; Ptk2ab Zebrafish Information mature. In zebrafish, muscles first express Col12a1 and Col22a1 at
Network) at the ends of myofibers where they attach to ISBs, which larval stages and these progressively align into the orthogonal fibril
stabilizes myotome boundaries (Bassett et al., 2003; Henry et al., arrays of mature MTJ/tendon ECM (Charvet et al., 2011, 2013).
2005; Parsons et al., 2002; Snow et al., 2008). TPCs only appear later Col12a1 is expressed earlier than Col22a1 and colocalizes with
along the ISBs, where they contribute additional ECM to strengthen Lama2 throughout muscle fiber attachment (Bader et al., 2009).
existing attachments in what we refer to here as a tendon-dependent Col22a1 maintains attachments under tension, and its expression
phase (Fig. 3B) (Charvet et al., 2011, 2013; Chen and Galloway, increases in tendinopathies (see Box 1) in humans (Charvet et al.,
2014; Subramanian and Schilling, 2014). Thus, in zebrafish, somitic 2013; Jelinsky et al., 2011). Recently, mutations in the human
muscles attach via the myomatrix at ISBs prior to the appearance of COL12A1 gene that disrupt COL12A1 secretion have been linked to a
TPCs. This may help explain how other types of attachments, such as form of Bethlem myopathy (Bushby et al., 2014; Schessl et al., 2006);
fleshy insertions of mammalian muscles, develop. other forms of this myopathy are caused by mutations in COL6A1,
Another ECM protein recently shown to be crucial for tendon COL6A2 and COL6A3, which also show elevated expression in
development is thrombospondin 4 (Tsp4; also known as Thbs4). human tendinopathies (Bnnemann, 2011; Jelinsky et al., 2011). In
Like Fn and Lam, zebrafish Tsp4b is initially produced by mice, Col12a1 interacts with tenascin C (Tnc) and helps crosslink
myoblasts and accumulates at ISBs prior to muscle attachment other collagens during fibril maturation by interacting with Dcn (Veit
(Fig. 3A) (Subramanian and Schilling, 2014). Tsp4b maintains et al., 2006). Transmission electron microscopy studies also suggest
muscle attachments at the ISB, and its depletion leads to detachment that Col12a1 forms complexes with structural collagens (e.g. Col1a1,
upon contraction. The transplantation of wild-type myoblasts into Col1a2), as well as other scaffolding and crosslinking proteins (Dcn,
DEVELOPMENT

Tsp4b-deficient embryos locally rescues muscle attachments at Fmod, thrombospondins) (Font et al., 1996). Thus the FACIT
ISBs, consistent with a role for Tsp4b in the myomatrix during the proteins are highly conserved regulators of tendon ultrastructure and
tendon-independent phase of attachment (Subramanian and elasticity.
Schilling, 2014). Interestingly, tsp4b mRNA abruptly disappears Other proteins involved in fibril assembly include the small
from differentiating myofibers as they attach, suggesting a feedback leucine-rich proteoglycans (SLRPs) such as Dcn (Fig. 4), Fmod,
mechanism that regulates tsp4b transcription. Mammalian biglycan and lumican. These are found in relatively small amounts
myoblasts also express Tsp4, and human TSP4 (THBS4) in tendons, yet loss-of-function mutations in the genes encoding
expression increases in pathological conditions such as Duchenne these proteins disrupt collagen fibrillogenesis (Chakravarti, 2002;
muscular dystrophy and alpha-sarcoglycanopathies (Chen et al., Corsi et al., 2002; Zhang et al., 2006). SLRPs mainly crosslink

4194
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

A Early attachment phase

Myyo
M tubbee
otu
Cartilage cells

B Mid-attachment phase

Myotube
Tenocyte

Cartilage
Cartilage Cells
cells

C Late attachment phase


tube
Myo

Tenocyte

Key
Tsp4
Lam Col1a1
Dcn FACIT Col1a2
collagen trimer
DMD
Itg Fn
complex
DEVELOPMENT

Fig. 4. Maturation and assembly of the tendon ECM. Diagram illustrating progressive changes in the ECM at an MTJ as it matures. (A) In the early attachment
phase, myoblasts (green) first extend towards a cartilage condensation (blue) and reorganize the local ECM by secreting Tsp4 (red), which interacts with Fn and
Lam. A magnified view (right) of the boxed area illustrates how Tsp4 pentamers assemble Fn, Lam and Dcn and facilitate binding to Itgs on both muscle and
cartilage cell surfaces, thereby promoting adhesion. (B) Following this, in the mid-attachment phase, linear collagen fibrils (Col1a1 trimers, dark blue) form,
tenocytes (red) invade, and Sox9+/Scx+ progenitors (dark blue and orange) become detected at the future attachment site on the cartilage, the enthesis. The
magnified view illustrates how Col1a1 trimers begin to align perpendicular to skeletal cells (enthesis, dark blue and orange). Dystrophin (DMD) complexes appear
on muscle surfaces. (C) In the final late attachment phase, collagen fibrils become crosslinked into a lattice, with tenocytes (red cells) extending processes to
surround fibrils, and entheses chondrifying ( purple). The magnified view shows Col1a1 trimers becoming crosslinked by FACIT collagens and surrounded by
tenocyte (red) processes, stabilizing the ECM and its interactions with Itgs on muscle and cartilage cells.

4195
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

Integrin signaling
Box 1. Tendinopathies Many collagens and laminins at developing MTJs directly bind Itg
The term tendinopathy refers to a diverse set of tendon disorders complexes that are present in the membranes of muscle cells and
(overlapping in some cases both in genetic and molecular terms with tenocytes (Fig. 5) (Docheva et al., 2014; Mayer et al., 1997; Pan
myopathies) that are caused either by genetic mutations in ECM et al., 2013; Rooney et al., 2006, 2012). In muscle, these continuous
components of MTJs or by mechanical stress that leads to tendon
ECM damage. Hallmarks of tendinopathies include COL1A1, COL1A2,
structural links between ECM proteins, the sarcolemma and the
COL4A1 and COL4A2 overexpression, fibril disorganization, increased cytoskeleton, maintain fiber integrity and modulate adhesion and
collagen crosslinking, reduced tissue inhibition of MMPs (i.e. TIMP gene expression. Not surprisingly, several types of human
activity), and elevated expression of MMP2, MMP14 and MMP19 as well tendinopathies (see Box 1) are associated with changes in the
as of versican, biglycan and Dcn (Jelinsky et al., 2011; Parkinson et al., expression of Itgs and their ligands (Bnnemann, 2011; Jelinsky
2011; Dunkman et al., 2013; Zhou et al., 2014). Recent studies have also et al., 2011; Schessl et al., 2006). It is thus important to determine
identified COL6A1/2/3 and COL12A1 as genes underlying one form of
the specific roles of Itg signaling during MTJ maturation and tendon
human myotendinopathy, which affects both myomatrix structure and
tendon matrix structure (Bnnemann, 2011; Pan et al., 2013). repair after injury.
Different Itg heterodimer combinations lend specificity for
different ligands. For example, laminins (Lama2, Lama4) bind
collagen fibrils, but they also appear to cross-regulate one anothers Itga7/b1 in the muscle basement membrane (Fig. 5) (Yurchenco et al.,
transcription through as yet unknown mechanisms (Yoon and 2004; Durbeej, 2010; Carmignac and Durbeej, 2012), and defects in
Halper, 2005; Zhang et al., 2006; Dunkman et al., 2013). This might LAMA2 have been associated with merosin-deficient muscular
involve feedback regulation of Scx and Mkx expression through the dystrophy in humans (Tom et al., 1994; Rooney et al., 2012).
tendon ECM, and recent studies have suggested that Dcn and Fmod Furthermore, mutations that disrupt genes encoding crosslinking
are regulated by Mkx (Fig. 2B) (Ito et al., 2010; Liu et al., 2010; collagens (e.g. COL12A1 in the case of EhlersDanlos syndrome),
Alberton et al., 2012). These results point to a system by which the which bind Itga1/b1 or Itga2/b1, cause widespread defects in skin,
dynamics of fibril ultrastructure feedback on ECM production to bones and tendons (Mayer et al., 1997; Zou et al., 2014).
modify tendon strength continuously. Which other Itg ligands control MTJ formation and maturation? In
Tendon ECM and collagen fibrils are also continuously Drosophila, Sr promotes the transcription of the single fly
remodeled in response to mechanical forces, at least around the thrombospondin (Tsp) gene in tenocytes. Tsp binds PS2 (If)/PS
circumference of a tendon fascicle (Frolova et al., 2014; (Mys) Itg heterodimers on fly muscle and tendon cell surfaces and
Heinemeier et al., 2012; Herchenhan et al., 2013; Kjaer, 2004; patterns muscle attachments (Chanana et al., 2007; Subramanian
Pingel et al., 2014). TPCs in culture subjected to moderate et al., 2007). Vertebrates have at least five thrombospondins, with
mechanical forces show an increase in collagen fibril diameter, diverse functions in cell migration, vasculogenesis, wound healing
though diameter decreases with excessive mechanical force (De and cancer (Adams and Lawler, 2004; Bornstein et al., 2004;
Almeida et al., 2010; Pingel et al., 2014). This remodeling occurs Kyriakides et al., 1999; Mustonen et al., 2012). Among these,
primarily through the activities of matrix metalloproteinases zebrafish Tsp4b is first secreted by myoblasts (Fig. 3A) and later
(MMPs) and their corresponding tissue inhibitors (TIMPs), as by TPCs (Fig. 3B), and is essential for muscle attachment
well as disintegrin and metalloprotease with thrombospondin (Subramanian and Schilling, 2014). Accordingly, zebrafish
repeats (ADAMTS) proteases (Bedi et al., 2010; Gotoh et al., embryos depleted of Tsp4b have defects in laminin localization and
2013; Jones et al., 2006; Maeda et al., 2013). Almost all of the 23 FAK phosphorylation (indicating reduced Itg signaling) at developing
MMPs and 19 ADAMTS proteins known to be expressed in ISBs, and their muscles detach under tension. These findings suggest
vertebrates are detectable in adult tendon tissue and play a variety that, in zebrafish, Tsp4 plays key roles in organizing the ECM of both
of both positive and negative roles in establishing a functional MTJ muscle and tendon, particularly those components essential for Itg
and tendon ECM (Davis et al., 2013; Spanoudes et al., 2014). signaling. This requirement for Tsp4 is at least partially conserved, as
MMPs are zinc-dependent endopeptidases that bind and unwind Tsp4 / mutant mice show defects in ECM deposition in developing
the triple helix of collagen monomers. Collagenases such as Mmp1 tendons (Fig. 4) (Frolova et al., 2014; Subramanian and Schilling,
target the structural collagens for degradation, whereas gelatinases 2014). Tsp4 and other subtype B thrombospondins form pentamers
such as Mmp2 and Mmp9 and membrane-bound MMPs, such as that directly bind to collagens, laminins and other ECM proteins
Mmp14, degrade smaller network collagens. Importantly, recent (Hauser et al., 1995). Surprisingly, human TSP4 protein
studies have shown that the expression and activity of MMPs are microinjected into the ECM surrounding Tsp4b-deficient myofibers
regulated by signals activated in response to mechanical forces, in zebrafish localizes to ISBs and locally rescues laminin localization,
such as Itg and Tgf (Yu and Stamenkovic, 2000; Farhat et al., Itg signaling and muscle attachments, suggesting that Tsp4 could
2015). MMP misregulation also occurs upon tendon inflammation, function as a scaffold for other ECM proteins during their assembly at
and recent studies suggest that MMP inhibition can improve tendon muscle attachments. Consistent with this model, the ability of
repair (Bedi et al., 2010; Jelinsky et al., 2011; Farhat et al., 2012; microinjected zebrafish tsp4b mRNA to rescue Tsp4b-deficient
Davis et al., 2013). attachments requires Itg binding and pentamerization; mutation of
DEVELOPMENT

either the Itg-binding (KGD) domain or the pentamerization (CQAC)


ECM-mediated signaling during tendon and MTJ formation, domain of Tsp4b disrupts its localization to tendons and eliminates its
maturation and repair ability to rescue muscle attachments in Tsp4b-deficient larvae
Multiple signaling pathways involving the ECM influence both the (Subramanian and Schilling, 2014). These results, along with a
formation of muscle attachments and the maturation of tenocytes. potentially conserved requirement for Tsp4 in mice, suggest that
Important players in muscle cells include Itgs and dystrophin, which TSP4 defects could contribute to human tendinopathies, highlighting
interact with ECM components at the MTJ. Furthermore, in both TSP4 as an attractive therapeutic target for strengthening the tendon
muscles and tendons, mechanical forces are thought to have a role in ECM. They also highlight the close association between the structural
dynamic remodeling of the ECM. and signaling roles of the ECM.

4196
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

Muscle Mechanical stress


contraction ECM Col22a1 ECM

ad3
Col1a1

Sm
Col1a2
Col3a1
Actin
Col6a1
filaments
Smad3 Col12a1
Col14a1
Muscle Tenocyte Tgfb2
Egr1
Tnmd

Smad3
Egr2
Scx Fmod
Mkx Tsp4

Actin
filaments

Key
Lama2, TGF FACIT
Tsp4b TGF Tgfr Itg Col1a
Lama4 LLC Col

Fig. 5. Model for ECM-mediated feedback from mechanical force and its effects on tenocyte gene expression. A tenocyte (orange) synthesizes the tendon
ECM, including Lama2, Lama4 (brown), Tsp4b (red pentagons), Col1a (blue) and FACIT Col ( purple), all of which signal through Itg receptors (dark blue) on
muscle and tenocyte cell surfaces in response to mechanical stress (gray arrows). In addition, stress causes the ECM to release TGF (yellow) from the TGF
large latent complex (LLC) (gray dotted arrows). Itg and TGF signaling in tenocytes feedback to regulate Scx-, Egr1/2- and Mkx-induced transcription (dashed
arrows) of the same Itg ligands as well as of other ECM components to modulate tendon stiffness. Smad3 also interacts with Scx and Mkx to activate target ECM
genes. The muscle fiber also contributes to the tendon matrix by secreting FACIT Col22a1.

TGF signaling Mmp9 and Bmp1 proteases (Munger and Sheppard, 2011). Few
TGF signaling provides another striking example of the relationship studies have addressed these mechanisms of TGF activation
between ECM structure and signaling in tendons. Genetic studies in specifically for the ECM of tendons or MTJs. A recent
mice have revealed crucial roles for TGF at multiple steps in tendon transcriptomic analysis of Scx-expressing tenocytes from mouse
development, maturation, maintenance and repair (Figs 2, 3, 5). limbs reveals that both TGF and MAPK signaling are strongly
Removing the functions of both TGF2 and TGF3 ligands, or of upregulated, but that only TGF upregulates Tsp2, Tsp4 and LTBP
TGFr2, eliminates most if not all differentiated tendons, whereas components of the ECM and promotes the tenocyte cell fate (Havis
exogenous TGF is sufficient to induce the expression of Scx et al., 2014).
and Col1a1 (Fig. 2A) (Chuang et al., 2014; Pryce et al., 2009). The correct regulation of TGF activity is crucial not only for
Tgfb2//Tgfb3/ mutant mice lose Scx expression in TPCs tendon development but also for healing injured tendons. Injuries
between E11.5 and E12.5, suggesting that TGF is required for lead to excessive release of TGF owing to mechanical force-
TPC maintenance (Pryce et al., 2009). Signaling through TGFr2 mediated activation of TGF (discussed below) and can cause
phosphorylates Smad2 and Smad3, which translocate to the nucleus fibrotic scarring of the tendon, thereby disrupting its function
and activate target genes, thereby maintaining differentiated (Farhat et al., 2015). In one model, elevated TGF might
tenocytes (Figs 3 and 5). Mouse Smad3 / mutants have reduced overactivate MMPs, which in turn promotes further release of
tendon tensile strength and increased spacing between collagen active TGF from the ECM as well as activating expression of
fascicles as well as reduced Mkx and increased Mmp9 expression Scx and Mkx and driving further ECM production.
(Berthet et al., 2013; Katzel et al., 2011).
Interestingly, the most likely source of TGF ligand at muscle Mechanical forces and signaling
attachments is the ECM. A recent in vitro study has shown that As alluded to throughout this Review, tenocytes actively sense
during tenocyte differentiation, Mkx activates the expression of mechanical force, leading to changes in gene expression,
TGF in differentiating MSCs (Fig. 5) (Liu et al., 2015). TGFs are cytoskeletal organization and ECM secretion (Fig. 5) (Banos
secreted bound to latent TGF-binding proteins (LTBPs), which et al., 2008; Maeda et al., 2009, 2013). Such feedback must be
form part of the large latency complex (LLC) in the ECM (Wipff extremely important for a tissue that constantly adjusts its stiffness
et al., 2007; Maeda et al., 2011). They are also secreted along with to changing loads. It depends, at least in part, on signaling through
latency-associated peptides (LAPs), which block association with gap junctional complexes localized to tenocyte processes (Kruegel
DEVELOPMENT

TGF receptors, and along with other proteins of the LLC, they and Miosge, 2010; Maeda et al., 2012). Indeed, rats subjected to
become incorporated into ECM via interactions between LTBPs and running on treadmills have increased Tnmd and Col1a1 expression
Fn, fibrillin or Dcn (Isogai et al., 2003; Rifkin, 2005; Farhat et al., as well as TPC proliferation (Eliasson et al., 2009; Zhang and Wang,
2012). In this manner, TGFs are stored in the ECM and must be 2013). Elevated secretion of both Col4a1 and Col6a1 is also seen in
released from the LLC and LAPs in order to be activated and developing chick tendons under stress, and this alters the
available to interact with cognate receptors (Horiguchi et al., 2012). crosslinking of fibrils (Marturano et al., 2014), thereby fine-
Activation may occur by release of TGF stimulated by shearing tuning tendon strength and promoting repair (Bailey et al., 1998;
forces, LLC degradation by proteases, interactions with Itgs through Willett et al., 2010). What are the molecular mechanisms underlying
RGD motifs on LTBPs themselves, or by the activity of Mmp2, these cellular responses?

4197
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

TGF signaling is one such mechanosensitive pathway that ECM functions at tendon-bone attachments
could control the response of tendons to force. Mechanical force So far, we have emphasized the ECM associated with tendon-
causes release of TGF1 from LTBPs in the ECM (Fig. 5) muscle attachments. But tendons also attach, at their other ends, to
(Maeda et al., 2011; Wipff et al., 2007). Under normal loads, cartilage/bone under a unique set of mechanical constraints. Many
TGF signaling through Smad2/3 maintains Scx expression in tendons insert into bony protrusions known as osteotendinous
tenocytes, whereas excessive loading disrupts TGF signaling, junctions or entheses, such as the deltoid tuberosity on the
damages the ECM and leads to tenocyte cell death in mice humerus (Fig. 6A). A characteristic structural feature of entheses is
(Maeda et al., 2009, 2010, 2011). TGF signaling in response to the presence of fibrocartilage, a tissue with physical properties
force also upregulates ITGA1 and ITGA2 expression in human somewhere in between cartilage and tendon. Within an enthesis, a
TPC cultures (Popov et al., 2015). Tenocytes elevate expression rapid transition from a more bone-like cellular and ECM structure to
of Tgfb1, Tgfbr2 and Smad7 in response to injury in mice a more cartilage-like (less rigid) structure in regions closer to the
(Guerquin et al., 2013). Thus, one attractive model is that force tendon is observed. In addition, a unique mineralization gradient
triggers TGF signaling leading to increased expression of Scx forms from the bony front to the point of tendon insertion, the width
and Mkx, which in turn activates TGF expression, creating a of which is constant with corresponding changes in cellular density
positive-feedback loop that leads secondarily to remodeling/ (Schwartz et al., 2012). The enthesis ECM also shows more Dcn
strengthening of ECM (Fig. 5) (Liu et al., 2015). Notably, TGF and Bgn localized to the tendon side, whereas Col2a1, Col9, Col10
signaling in response to mechanical forces also controls MMP and aggrecan localize to the bony side (Thomopoulos et al., 2003).
expression during tendon repair (Katzel et al., 2011; Farhat et al., These gradual transitions in ECM and rigidity are essential for the
2015). Interestingly, injured tendons in Egr1 / mutant mice fail proper transmission of contractile forces to the bone to prevent
to upregulate TGF and Scx or to repair their tendons efficiently avulsion fractures (Zelzer et al., 2014).
(Guerquin et al., 2013). In the appendicular skeleton, entheses are established through
The unique collagen fibril organization of tendons allows them specialized contours and protuberances on bones called eminences.
to bear the stress of muscle contraction and prevents bone Recent genetic studies in mice have begun to elucidate the signals
fractures (Davis et al., 2013; Pan et al., 2013; Schwartz et al., that control the formation of these structures (Blitz et al., 2009,
2013; Zhang et al., 2006). How fibrils physically anchor to cells 2013; Zelzer et al., 2014). In mouse embryos, eminence
and how the dynamics of these anchors are regulated under load development coincides with the formation of muscle/tendon
remain unclear. Biopsies of human Achilles tendons have shown attachments, suggesting that attachments impose physical changes
that fibrils buckle in overloaded tendons (Pingel et al., 2014). In on the bone. However, mouse mutants that lack muscles, such as
tenocytes cultured in collagen gels, mechanical force translates splotch delayed (spd) or muscular dysgenesis (mdg) mutants, still
into cytoskeletal force through non-muscle myosin II bound to form eminences like the deltoid tuberosity on the humerus, despite
actin fibrils associated with focal adhesion complexes, which in severe joint fusions (Fig. 6B) (Blitz et al., 2009).
turn associate with Itgs and other ECM receptors. The chemical Perhaps not surprisingly, TGF signaling also plays important
inhibition of myosin II function (using blebbistatin) reduces roles in the formation of entheses. Loss of TGF signaling through
cytoskeletal traction forces and leads to ECM remodeling (Maeda ablation of Tgfr2 in limb mesenchyme eliminates eminences in the
et al., 2013). Thus, in addition to inducing transcriptional limb, though this might be due to a broader or earlier role for TGF
changes, mechanical force can stimulate changes in cytoskeletal signaling in skeletal/tendon progenitors (Fig. 6B) (Blitz et al., 2009,
tension that reverberate back to the ECM to alter its ultrastructure. 2013). Instead, evidence is building to suggest that the more crucial
MMPs have also been shown to modulate ECM structure in inducers of eminences are other members of the TGF superfamily,
response to mechanical cues during MTJ maturation and after namely bone morphogenetic proteins (BMPs). The conditional
injury. For example, tenocytes in silicone micropillar gels elevate deletion in mice of Bmp4 specifically in tenocytes using an Scx:Cre
MMP expression levels in response to gel deformation (Maeda driver completely eliminates eminences (Blitz et al., 2009). Pulse-
et al., 2013). In addition, humans show dramatic increases in the chase labeling studies show that eminences form from secondary
levels of MMP2, MMP9 and MMP14 in adult tendons following fields of Scx/Sox9 co-expressing cells that lie immediately adjacent
endurance exercise, suggesting that these proteins aid MTJ repair in to major skeletal condensations (Fig. 6B) (Blitz et al., 2013); these
response to mechanotransduction (Rullman et al., 2009). This cells form in mutants lacking BMP signaling, but do not
effect depends on the timing of loading as, in cultured tendon differentiate. Early Sox9 expression is observed in both the
fascicles, cells upregulate MMP2 and MMP13 after very short skeletal condensation and the eminence, but the subset of these
cycles of loading, but downregulate MMP1 after longer cycles cells that express Scx are delayed in expression of Col2a1, thereby
(Maeda et al., 2009, 2013). Similarly, in vitro studies of mechanical restricting chondrogenesis to the developing enthesis. Scx-
loading on mouse tenocytes have shown that, whereas low levels of expressing cells in developing entheses express Bmp4 and this is
shear force lead to upregulation of Col1a and Tmnd, increasing the lost in tendons of Scx/ mutant mice, which lack entheses. Thus, an
force leads to upregulation of Runx2 and Sox9 (Zhang and Wang, attractive hypothesis is that Bmp4 secreted by tenocytes induces
2015). Understanding these dynamic responses to mechanical enthesis formation in adjacent skeletogenic mesenchyme (Fig. 6B).
DEVELOPMENT

stimulation might lead to improved therapeutic interventions; the Interestingly, Sox9-expressing secondary fields of eminence
systemic inhibition of MMPs, for example, can reduce fibrotic progenitors still form in Bmpr1a/ mutant mice but never
scarring of muscle ECM (Farhat et al., 2015). MMP expression is differentiate, indicating a role for BMP signaling in maturation
also regulated by TGFs (Yu and Stamenkovic, 2000; Ge and rather than specification. Taken together, these studies establish an
Greenspan, 2006; Farhat et al., 2015). In turn, MMP2, MMP9 and early phase of eminence/enthesis development, which is
BMP1 proteases might specifically digest LLC and release active independent of muscle development, and suggest that Scx-driven
TGF, establishing a positive-feedback loop that could help fine- Bmp4 signaling non-autonomously regulates their formation
tune MMP levels, both during normal tendon function and in (Fig. 6B) (Blitz et al., 2013; Murchison et al., 2007; Pryce et al.,
response to tendon injury or exercise (Fig. 5). 2009). Such coordinated expression of Scx and Sox9 in tenocytes

4198
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

A Wild type spd mutant

Col2b
Gdf5
Col2a1 Col2b Col2a1

Col2a1 Col2a1

Gdf5-expressing Col2b-expressing Col2a-expressing


joint cells cartilage cells at cartilage cells
articular surface

B Tgfr2 lof in limb and Bmp4 lof in


Wild type Sox9 lof in tenocytes limb mesenchyme

Enthesis

Secondary
field

Primary
field

Sox9
Col2a

Tendon

Muscle

Scx+/Bmp+ Sox9high,Scxlow Scxhigh,Sox9low Sox9+ cells that do


TPCs Tenocytes
enthesis cells enthesis cells not express Col2a

Fig. 6. ECM functions at tendon-bone attachments. (A) Diagrams illustrating changes in cartilage at the developing humero-ulnar joint of the mouse
forelimb in wild-type embryos (left) and in splotch delayed (spd, Pax3) mutants (right), which lack muscles. Proliferating chondrocytes express Col2a1 (light blue),
whereas cells forming at the edges of the joint express Col2b (dark blue), and cells in the joint interzone secrete Gdf5 (green) into the joint region. The loss
of muscles in spd mutants leads to loss of Gdf5 expression, disorganized Col2b+ interzone cells and joint fusion. (B) Diagrams illustrating changes in cartilage
and tenocytes at a developing eminence. The primary field contains cells that form chondrocytes within the developing bone, whereas the secondary field
consists of Sox9-positive progenitor cells that lie outside of the primary field. In wild-type embryos, three different subsets of Scx-expressing cells at a muscle
insertion site of a developing long bone are found: Sox9+/Scx+, Scx+ or Scx+/Bmp4+. Loss of Tgfr2 in limb mesenchyme or of Sox9 in tenocytes leads to a loss of
the Sox9/Scx co-expressing and Sox9-expressing population in the secondary field, but not other tenocytes. Loss of Bmp4 signaling leads to a loss of both Sox9+/
DEVELOPMENT

Scx+ and Scx+ populations in the secondary field. Dotted lines outline primary field. Dashed lines outline secondary field. lof, loss of function.

and chondrocytes is an emerging theme both during tendon as during aging and disease (Magne and Bougault, 2015; Zhang and
development and in MSCs (Fig. 2A) (Asou et al., 2002; Soeda Wang, 2015).
et al., 2010; Sugimoto et al., 2013). The close lineal relationship Finally, and not surprisingly, entheses are also extremely
between tenocytes and chondrocytes/osteocytes (Fig. 2A) and their sensitive to mechanical forces. Mechanical forces have long been
regulation at entheses also may help explain the ectopic ossification known to be important for the development of the bones to which
of tendons (see Box 2) that occurs normally in some species as well muscles attach, as well as for the maintenance of skeletogenic cell

4199
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

mechanical load. This occurs regionally within each tendon from


Box 2. Ectopic tendon calcification its muscle origin to its bony insertion. Understanding the tendon/
Ectopic ossification of tendons occurs normally in some species and is ligament gene regulatory program thus requires knowledge of
also observed during aging and in disease (Magne and Bougault, 2015; how this dynamic network of ECM proteins self-assembles and
Zhang and Wang, 2015). Spondyloarthritis, for example, is an feeds back upon transcriptional regulators in response to
inflammatory enthesitis (Weinreb et al., 2014) that can lead to ectopic
ossification spreading from the bone to the tendon or ligament. Ectopic
mechanical forces.
ossification is also seen in calcifying tendinopathies, a common The musculoskeletal system and its associated ECM have also
consequence of aging affecting as many as 1 in 5 adults over 50 years evolved to suit dramatically different modes of locomotion, feeding
of age. Tendon ossification appears to be caused, in part, by tenocyte- strategies and body sizes of different vertebrates. Changes in the
dependent degradation of the tendon ECM (Magne and Bougault, 2015) interconnections between individual muscles and bones underlie
as well as altered responses to mechanical forces and BMP/Smad many of the evolutionary differences between species. For example,
signaling (Rui et al., 2013). The genetic inactivation of two small
mandibles of fish (Malawi cichlids), birds (Darwins finches) and
proteoglycans (Bgn and Fmod) of the ECM of cultured mouse TPCs
leads to ectopic activation of BMP signaling and tendon ossification (Bi dogs can have very different functional morphologies depending on
et al., 2007). Mechanical stretching of TPCs in vitro can also lead to BMP their feeding strategies, which notably have all been linked to
upregulation and abnormal ossification (Zhang and Wang, 2013, 2015). changes in BMP signaling (Abzhanov et al., 2004; Albertson et al.,
Surprisingly, BMP signaling oscillates in a circadian manner, and these 2005; Schoenebeck et al., 2012). These reflect coordinated changes
cycles are deregulated in arrhythmic mutant mice, which correlates with in the regulation of cranial neural crest cells that form not only the
increased tendon ossification (Yeung et al., 2014) Thus, tendons and
craniofacial skeleton, but also cranial TPCs during evolution.
ligaments are really tissues living on the edge with respect to their ECM
composition and cellular constituents, presumably owing to their extreme
Indeed, recent studies grafting neural crest cells between quail and
responsiveness to feedback through mechanical force. duck embryos reveal that some distinct craniofacial morphologies
have evolved through changes in cell-intrinsic mechanisms. Jaw
cartilage differentiation, marked by Runx2 expression, occurs
populations (Shwartz et al., 2013). The paralysis of specific muscles earlier in the duck embryo, whereas tendon differentiation, marked
leads to bone defects at sites of attachment and loss of joint by Scx expression, occurs earlier in the quail (Tokita and Schneider,
progenitors (Kahn et al., 2009). The tendon ECM plays vital roles in 2009). How these species-specific differences in timing of skeletal
these adaptations of muscles and bones to mechanical loading and tendon differentiation reflect changes in ECM organization and
(Kjaer, 2004). in mechanical stress to suit their adaptive functions is an important
area for future investigation.
Conclusions and perspectives Understanding the genetic control of the development of tendons,
Vertebrate tendons begin life similar to skeletal progenitors in the their integration into the musculoskeletal system and their ECM
embryo, but rapidly establish unique identities and tissue organization also has important implications for regenerative
organization, in large part through interactions with and assembly medicine. Current efforts to treat tendon injuries or diseases focus
of the tendon ECM. This involves: (1) essential transcription factors either on ameliorating inflammation or driving MSCs towards the
expressed in tenocytes, such as Scx, Mkx and Egr1, that drive the tenocyte fate with hopes of stem cell therapy (Yang et al., 2013).
expression of ECM proteins; (2) ECM components such as laminins Both approaches leave out the crucial role of establishing the
that help establish muscle attachments in the absence of tenocytes appropriate ECM for a specific tendon type and the loads that it
and control tenocyte morphogenesis; (3) ECM components such as needs to bear. Attaining the correct strength also relies on correct
Tsp4 that drive the assembly of collagen fibrils at MTJs through cellular responses to mechanical forces, which are transduced
their interactions with other ECM proteins and Itgs on muscle cell through the ECM. Thus, it is important to consider the ECM and its
surfaces; and (4) the maintenance and repair of these ECM functions going forward in efforts to improve diagnosis and therapy
components in response to mechanical forces. In this Review, we design for tendon disorders.
have highlighted recent genetic studies that have provided insights
into the molecular mechanisms underlying these different Acknowledgements
processes. We thank members of the Schilling lab and anonymous reviewers for critical reading
of the manuscript.
The discovery of Scx and Mkx has provided inroads into
understanding the tendon/ligament gene regulatory program and
Competing interests
revealed the close relationship between tenocytes and skeletal The authors declare no competing or financial interests.
lineages. Scx is the earliest known marker of TPCs, and its
overexpression transforms MSCs into tenocytes (Alberton et al., Funding
2012), whereas Sox9 overexpression converts MSCs into The authors were supported by National Institutes of Health awards [R21 AR62792
cartilage (Fig. 2A) (Takimoto et al., 2012). However, unlike and R01 DE13828 to T.F.S.]. Deposited in PMC for release after 12 months.
Sox9, which when eliminated leads to a failure to form cartilage,
loss-of-function Scx or Mkx mutations in mice do not eliminate References
Abzhanov, A., Protas, M., Grant, B. R., Grant, P. R. and Tabin, C. J. (2004). Bmp4
DEVELOPMENT

tenocytes, and mutants are viable although they have severe and morphological variation of beaks in Darwins finches. Science 305,
musculoskeletal deformities. This implies that tendon 1462-1465.
development involves multiple, partially redundant transcription Adams, J. C. and Lawler, J. (2004). The thrombospondins. Int. J. Biochem. Cell
Biol. 36, 961-968.
factors and cell-cell signals, each playing a unique role in Alberton, P., Popov, C., Pra gert, M., Kohler, J., Shukunami, C., Schieker, M. and
building and maintaining the complex network of ECM proteins Docheva, D. (2012). Conversion of human bone marrow-derived mesenchymal
at muscle attachments. stem cells into tendon progenitor cells by ectopic expression of scleraxis. Stem
These studies also highlight the fact that the stability of muscle Cells Dev. 21, 846-858.
Albertson, R. C., Streelman, J. T., Kocher, T. D. and Yelick, P. C. (2005).
attachments is not pre-established during development but Integration and evolution of the cichlid mandible: the molecular basis of alternate
evolves through constant adaptation of the ECM to changing feeding strategies. Proc. Natl. Acad. Sci. USA 102, 16287-16292.

4200
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

Alliston, T., Choy, L., Ducy, P., Karsenty, G. and Derynck, R. (2001). TGF-beta- Chanana, B., Graf, R., Koledachkina, T., Pflanz, R. and Vorbru ggen, G. (2007).
induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin AlphaPS2 integrin-mediated muscle attachment in Drosophila requires the ECM
expression and inhibits osteoblast differentiation. EMBO J. 20, 2254-2272. protein Thrombospondin. Mech. Dev. 124, 463-475.
Anderson, D. M., Arredondo, J., Hahn, K., Valente, G., Martin, J. F., Wilson- Charvet, B., Malbouyres, M., Pagnon-Minot, A., Ruggiero, F. and Le Guellec, D.
Rawls, J. and Rawls, A. (2006). Mohawk is a novel homeobox gene expressed in (2011). Development of the zebrafish myoseptum with emphasis on the
the developing mouse embryo. Dev. Dyn. 235, 792-801. myotendinous junction. Cell Tissue Res. 346, 439-449.
Anderson, D. M., Beres, B. J., Wilson-Rawls, J. and Rawls, A. (2009). The Charvet, B., Guiraud, A., Malbouyres, M., Zwolanek, D., Guillon, E., Bretaud, S.,
homeobox gene Mohawk represses transcription by recruiting the sin3A/HDAC Monnot, C., Schulze, J., Bader, H. L., Allard, B. et al. (2013). Knockdown of
co-repressor complex. Dev. Dyn. 238, 572-580. col22a1 gene in zebrafish induces a muscular dystrophy by disruption of the
Anderson, D. M., George, R., Noyes, M. B., Rowton, M., Liu, W., Jiang, R., Wolfe, myotendinous junction. Development 140, 4602-4613.
S. A., Wilson-Rawls, J. and Rawls, A. (2012). Characterization of the DNA- Chen, J. W. and Galloway, J. L. (2014). The development of zebrafish tendon and
binding properties of the Mohawk homeobox transcription factor. J. Biol. Chem. ligament progenitors. Development 141, 2035-2045.
287, 35351-35359. Chen, Y.-W., Zhao, P., Borup, R. and Hoffman, E. P. (2000). Expression profiling
Aslan, H., Kimelman-Bleich, N., Pelled, G. and Gazit, D. (2008). Molecular targets in the muscular dystrophies: identification of novel aspects of molecular
for tendon neoformation. J. Clin. Invest. 118, 439-444. pathophysiology. J. Cell Biol. 151, 1321-1336.
Asou, Y., Nifuji, A., Tsuji, K., Shinomiya, K., Olson, E. N., Koopman, P. and Chen, X., Yin, Z., Chen, J. I., Shen, W. l., Liu, H. H., Tang, Q., Fang, Z., Lu, L. R.,
Noda, M. (2002). Coordinated expression of scleraxis and Sox9 genes Ji, J. and Ouyang, H. W. (2012). Force and scleraxis synergistically promote the
during embryonic development of tendons and cartilage. J. Orthop. Res. 20, commitment of human ES cells derived MSCs to tenocytes. Sci. Rep. 2, 977.
827-833. Choi, W. J., Park, M. S., Park, K. H., Courneya, J.-P., Cho, J. S., Schon, L. C. and
Bader, H. L., Keene, D. R., Charvet, B., Veit, G., Driever, W., Koch, M. and Lee, J. W. (2014). Comparative analysis of gene expression in normal and
Ruggiero, F. (2009). Zebrafish collagen XII is present in embryonic connective degenerative human tendon cells: effects of cyclic strain. Foot Ankle Int. 35,
tissue sheaths (fascia) and basement membranes. Matrix Biol. 28, 32-43. 1045-1056.
Bailey, A. J., Paul, R. G. and Knott, L. (1998). Mechanisms of maturation and Chuang, H.-N., Hsiao, K.-M., Chang, H.-Y., Wu, C.-C. and Pan, H. (2014). The
ageing of collagen. Mech. Ageing Dev. 106, 1-56. homeobox transcription factor Irxl1 negatively regulates MyoD expression and
Bajanca, F., Luz, M., Raymond, K., Martins, G. G., Sonnenberg, A., Tajbakhsh, myoblast differentiation. FEBS J. 281, 2990-3003.
S., Buckingham, M. and Thorsteinsdo ttir, S. (2006). Integrin alpha6beta1- Corsi, A., Xu, T., Chen, X.-D., Boyde, A., Liang, J., Mankani, M., Sommer, B.,
laminin interactions regulate early myotome formation in the mouse embryo. Iozzo, R. V., Eichstetter, I., Robey, P. G., et al. (2002). Phenotypic effects of
Development 133, 1635-1644. biglycan deficiency are linked to collagen fibril abnormalities, are synergized by
Banos, C. C., Thomas, A. H. and Kuo, C. K. (2008). Collagen fibrillogenesis in decorin deficiency, and mimic Ehlers-Danlos-like changes in bone and other
tendon development: current models and regulation of fibril assembly. Birth connective tissues. J. Bone Miner. Res. 17, 1180-1189.
Davis, M. E., Gumucio, J. P., Sugg, K. B., Bedi, A. and Mendias, C. L. (2013).
Defects Res. C. Embryo Today 84, 228-244.
Bassett, D. I., Bryson-Richardson, R. J., Daggett, D. F., Gautier, P., Keenan, MMP inhibition as a potential method to augment the healing of skeletal muscle
and tendon extracellular matrix. J. Appl. Physiol. 115, 884-891.
D. G. and Currie, P. D. (2003). Dystrophin is required for the formation of stable
De Almeida, F. M., Tomiosso, T. C., Biancalana, A., Mattiello-Rosa, S. M., Vidal,
muscle attachments in the zebrafish embryo. Development 130, 5851-5860.
B. C., Gomes, L. and Pimentel, E. R. (2010). Effects of stretching on
Becker, S., Pasca, G., Strumpf, D., Min, L. and Volk, T. (1997). Reciprocal
morphological and biochemical aspects of the extracellular matrix of the rat
signaling between Drosophila epidermal muscle attachment cells and their
calcaneal tendon. Cell Tissue Res. 342, 97-105.
corresponding muscles. Development 124, 2615-2622.
Docheva, D., Popov, C., Alberton, P. and Aszodi, A. (2014). Integrin signaling in
Bedi, A., Kovacevic, D., Hettrich, C., Gulotta, L. V., Ehteshami, J. R., Warren,
skeletal development and function. Birth Defects Res. C Embryo Today Rev. 102,
R. F. and Rodeo, S. A. (2010). The effect of matrix metalloproteinase inhibition on
13-36.
tendon-to-bone healing in a rotator cuff repair model. J. Shoulder Elbow Surg. 19,
Dunkman, A. A., Buckley, M. R., Mienaltowski, M. J., Adams, S. M., Thomas,
384-391.
S. J., Satchell, L., Kumar, A., Pathmanathan, L., Beason, D. P., Iozzo, R. V.
Berthet, E., Chen, C., Butcher, K., Schneider, R. A., Alliston, T. and
et al. (2013). Decorin expression is important for age-related changes in tendon
Amirtharajah, M. (2013). Smad3 binds Scleraxis and Mohawk and regulates
structure and mechanical properties. Matrix Biol. 32, 3-13.
tendon matrix organization. J. Orthop. Res. 31, 1475-1483.
Durbeej, M. (2010). Laminins. Cell Tissue Res. 339, 259-268.
Bi, Y., Ehirchiou, D., Kilts, T. M., Inkson, C. A., Embree, M. C., Sonoyama, W.,
Eliasson, P., Fahlgren, A. and Aspenberg, P. (2008). Mechanical load and BMP
Li, L., Leet, A. I., Seo, B.-M., Zhang, L. et al. (2007). Identification of tendon stem/ signaling during tendon repair: a role for follistatin? Clin. Orthop. Relat. Res. 466,
progenitor cells and the role of the extracellular matrix in their niche. Nat. Med. 13, 1592-1597.
1219-1227. Eliasson, P., Andersson, T. and Aspenberg, P. (2009). Rat Achilles tendon
Birch, H. L., Thorpe, C. T. and Rumian, A. P. (2013). Specialization of extracellular healing: mechanical loading and gene expression. J. Appl. Physiol. 107, 399-407.
matrix for function in tendons and ligaments. Muscles. Ligaments Tendons J. 3, Espira, L., Lamoureux, L., Jones, S. C., Gerard, R. D., Dixon, I. M. C. and
12-22. Czubryt, M. P. (2009). The basic helixloophelix transcription factor scleraxis
Blitz, E., Viukov, S., Sharir, A., Shwartz, Y., Galloway, J. L., Pryce, B. A., regulates fibroblast collagen synthesis. J. Mol. Cell. Cardiol. 47, 188-195.
Johnson, R. L., Tabin, C. J., Schweitzer, R. and Zelzer, E. (2009). Bone ridge Farhat, Y. M., Al-Maliki, A. A., Chen, T., Juneja, S. C., Schwarz, E. M., OKeefe,
patterning during musculoskeletal assembly is mediated through SCX regulation R. J. and Awad, H. A. (2012). Gene expression analysis of the pleiotropic effects
of Bmp4 at the tendon-skeleton junction. Dev. Cell 17, 861-873. of TGF-1 in an in vitro model of flexor tendon healing. PLoS ONE 7, e51411.
Blitz, E., Sharir, A., Akiyama, H. and Zelzer, E. (2013). Tendon-bone attachment Farhat, Y. M., Al-Maliki, A. A., Easa, A., OKeefe, R. J., Schwarz, E. M. and Awad,
unit is formed modularly by a distinct pool of Scx- and Sox9-positive progenitors. H. A. (2015). TGF-1 suppresses plasmin and MMP activity in flexor tendon cells
Development 140, 2680-2690. via PAI-1: implications for scarless flexor tendon repair. J. Cell Physiol. 230,
Bo nnemann, C. G. (2011). The collagen VI-related myopathies: muscle meets its 318-326.
matrix. Nat. Rev. Neurol. 7, 379-390. Font, B., Eichenberger, D., Rosenberg, L. M. and Van Der Rest, M. (1996).
Bonnin, M.-A., Laclef, C., Blaise, R., Eloy-Trinquet, S., Relaix, F., Maire, P. and Characterization of the interactions of type XII collagen with two small
Duprez, D. (2005). Six1 is not involved in limb tendon development, but is proteoglycans from fetal bovine tendon, decorin and fibromodulin. Matrix Biol.
expressed in limb connective tissue under Shh regulation. Mech. Dev. 122, 15, 341-348.
573-585. Frolova, E. G., Drazba, J., Krukovets, I., Kostenko, V., Blech, L., Harry, C.,
Bornstein, P., Agah, A. and Kyriakides, T. R. (2004). The role of thrombospondins Vasanji, A., Drumm, C., Sul, P., Jenniskens, G. J. et al. (2014). Control of
1 and 2 in the regulation of cellmatrix interactions, collagen fibril formation, and organization and function of muscle and tendon by thrombospondin-4. Matrix Biol.
the response to injury. Int. J. Biochem. Cell Biol. 36, 1115-1125. 37, 35-48.
DEVELOPMENT

Brent, A. E., Schweitzer, R. and Tabin, C. J. (2003). A somitic compartment of Ge, G. and Greenspan, D. S. (2006). BMP1 controls TGF1 activation via cleavage
tendon progenitors. Cell 113, 235-248. of latent TGF-binding protein. J. Cell Biol. 175, 111-120.
Brent, A. E., Braun, T. and Tabin, C. J. (2005). Genetic analysis of interactions Gotoh, M., Mitsui, Y., Shibata, H., Yamada, T., Shirachi, I., Nakama, K., Okawa,
between the somitic muscle, cartilage and tendon cell lineages during mouse T., Higuchi, F. and Nagata, K. (2013). Increased matrix metalloprotease-3 gene
development. Development 132, 515-528. expression in ruptured rotator cuff tendons is associated with postoperative
Bushby, K. M. D., Collins, J. and Hicks, D. (2014). Collagen type VI myopathies. tendon retear. Knee Surg. Sports Traumatol. Arthrosc. 21, 1807-1812.
Adv. Exp. Med. Biol. 802, 185-199. Grenier, J., Teillet, M.-A., Grifone, R., Kelly, R. G. and Duprez, D. (2009).
Carmignac, V. and Durbeej, M. (2012). Cell-matrix interactions in muscle disease. Relationship between neural crest cells and cranial mesoderm during head
J. Pathol. 226, 200-218. muscle development. PLoS ONE 4, e4381.
Chakravarti, S. (2002). Functions of lumican and fibromodulin: lessons from Guerquin, M.-J., Charvet, B., Nourissat, G., Havis, E., Ronsin, O., Bonnin, M.-A.,
knockout mice. Glycoconj. J. 19, 287-293. Ruggiu, M., Olivera-Martinez, I., Robert, N., Lu, Y. et al. (2013). Transcription

4201
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

factor EGR1 directs tendon differentiation and promotes tendon repair. J. Clin. accumulation for maintenance of somite boundaries in zebrafish embryos. Dev.
Invest. 123, 3564-3576. Cell 8, 587-598.
Hauser, N., Paulsson, M., Kale, A. and DiCesare, P. (1995). Tendon Kruegel, J. and Miosge, N. (2010). Basement membrane components are
extracellular matrix contains pentameric thrombospondin-4 (TSP-4). FEBS Lett. key players in specialized extracellular matrices. Cell. Mol. Life Sci. 67,
368, 307-310. 2879-2895.
Havis, E., Bonnin, M.-A., Olivera-Martinez, I., Nazaret, N., Ruggiu, M., Weibel, J., Kyriakides, T. R., Leach, K. J., Hoffman, A. S., Ratner, B. D. and Bornstein, P.
Durand, C., Guerquin, M.-J., Bonod-Bidaud, C., Ruggiero, F. et al. (2014). (1999). Mice that lack the angiogenesis inhibitor, thrombospondin 2, mount an
Transcriptomic analysis of mouse limb tendon cells during development. altered foreign body reaction characterized by increased vascularity. Proc. Natl.
Development 141, 3683-3696. Acad. Sci. USA 96, 4449-4454.
Heinemeier, K. M., Skovgaard, D., Bayer, M. L., Qvortrup, K., Kjaer, A., Kjaer, Le jard, V., Brideau, G., Blais, F., Salingcarnboriboon, R., Wagner, G., Roehrl,
M., Magnusson, S. P. and Kongsgaard, M. (2012). Uphill running improves rat M. H. A., Noda, M., Duprez, D., Houillier, P. and Rossert, J. (2007). Scleraxis
Achilles tendon tissue mechanical properties and alters gene expression without and NFATc regulate the expression of the pro-alpha1(I) collagen gene in tendon
inducing pathological changes. J. Appl. Physiol. 113, 827-836. fibroblasts. J. Biol. Chem. 282, 17665-17675.
Henry, C. A., McNulty, I. M., Durst, W. A., Munchel, S. E. and Amacher, S. L. Lejard, V., Blais, F., Guerquin, M.-J., Bonnet, A., Bonnin, M.-A., Havis, E.,
(2005). Interactions between muscle fibers and segment boundaries in zebrafish. Malbouyres, M., Bidaud, C. B., Maro, G., Gilardi-Hebenstreit, P. et al. (2011).
Dev. Biol. 287, 346-360. EGR1 and EGR2 involvement in vertebrate tendon differentiation. J. Biol. Chem.
Herchenhan, A., Bayer, M. L., Svensson, R. B., Magnusson, S. P. and Kjaer, M. 286, 5855-5867.
(2013). In vitro tendon tissue development from human fibroblasts demonstrates Li, Y., Ramcharan, M., Zhou, Z., Leong, D. J., Akinbiyi, T., Majeska, R. J. and
collagen fibril diameter growth associated with a rise in mechanical strength. Dev. Sun, H. B. (2015). The role of scleraxis in fate determination of mesenchymal
Dyn. 242, 2-8. stem cells for tenocyte differentiation. Sci. Rep. 5, 13149.
Horiguchi, M., Ota, M. and Rifkin, D. B. (2012). Matrix control of transforming Liu, D., Black, B. L. and Derynck, R. (2001). TGF- inhibits muscle differentiation
growth factor- function. J. Biochem. 152, 321-329. through functional repression of myogenic transcription factors by Smad3. Genes
Hosokawa, R., Oka, K., Yamaza, T., Iwata, J., Urata, M., Xu, X., Bringas, P., Dev. 15, 2950-2966.
Nonaka, K. and Chai, Y. (2010). TGF-beta mediated FGF10 signaling in Liu, W., Watson, S. S., Lan, Y., Keene, D. R., Ovitt, C. E., Liu, H., Schweitzer, R.
cranial neural crest cells controls development of myogenic progenitor cells and Jiang, R. (2010). The atypical homeodomain transcription factor Mohawk
through tissuetissue interactions during tongue morphogenesis. Dev. Biol. 341, controls tendon morphogenesis. Mol. Cell. Biol. 30, 4797-4807.
186-195. Liu, C.-F., Aschbacher-Smith, L., Barthelery, N. J., Dyment, N., Butler, D. and
Huang, A. H., Lu, H. H. and Schweitzer, R. (2015). Molecular regulation of tendon Wylie, C. (2012). Spatial and temporal expression of molecular markers and cell
cell fate during development. J. Orthop. Res. 33, 800-812. signals during normal development of the mouse patellar tendon. Tissue Eng. A
Isogai, Z., Ono, R. N., Ushiro, S., Keene, D. R., Chen, Y., Mazzieri, R., 18, 598-608.
Charbonneau, N. L., Reinhardt, D. P., Rifkin, D. B. and Sakai, L. Y. (2003). Liu, H., Zhu, S., Zhang, C., Lu, P., Hu, J., Yin, Z., Ma, Y., Chen, X. and OuYang, H.
Latent transforming growth factor beta-binding protein 1 interacts with fibrillin and (2014). Crucial transcription factors in tendon development and differentiation:
is a microfibril-associated protein. J. Biol. Chem. 278, 2750-2757. their potential for tendon regeneration. Cell Tissue Res. 356, 287-298.
Ito, Y., Toriuchi, N., Yoshitaka, T., Ueno-Kudoh, H., Sato, T., Yokoyama, S., Liu, H., Zhang, C., Zhu, S., Lu, P., Zhu, T., Gong, X., Zhang, Z., Hu, J., Yin, Z.,
Nishida, K., Akimoto, T., Takahashi, M., Miyaki, S. et al. (2010). The Mohawk Heng, B. C. et al. (2015). Mohawk promotes the tenogenesis of mesenchymal
homeobox gene is a critical regulator of tendon differentiation. Proc. Natl. Acad. stem cells through activation of the TGF signaling pathway. Stem Cells 33,
Sci. USA 107, 10538-10542. 443-455.
Jelinsky, S. A., Archambault, J., Li, L. and Seeherman, H. (2010). Tendon- Maeda, E., Shelton, J. C., Bader, D. L. and Lee, D. A. (2009). Differential regulation
selective genes identified from rat and human musculoskeletal tissues. J. Orthop. of gene expression in isolated tendon fascicles exposed to cyclic tensile strain in
Res. 28, 289-297. vitro. J. Appl. Physiol. 106, 506-512.
Jelinsky, S. A., Rodeo, S. A., Li, J., Gulotta, L. V., Archambault, J. M. and Maeda, E., Fleischmann, C., Mein, C. A., Shelton, J. C., Bader, D. L. and Lee,
Seeherman, H. J. (2011). Regulation of gene expression in human tendinopathy. D. A. (2010). Functional analysis of tenocytes gene expression in tendon fascicles
BMC Musculoskelet. Disord. 12, 86. subjected to cyclic tensile strain. Connect. Tissue Res. 51, 434-444.
Jones, G. C., Corps, A. N., Pennington, C. J., Clark, I. M., Edwards, D. R., Maeda, T., Sakabe, T., Sunaga, A., Sakai, K., Rivera, A. L., Keene, D. R., Sasaki,
Bradley, M. M., Hazleman, B. L. and Riley, G. P. (2006). Expression profiling of T., Stavnezer, E., Iannotti, J., Schweitzer, R. et al. (2011). Conversion of
metalloproteinases and tissue inhibitors of metalloproteinases in normal and mechanical force into TGF--mediated biochemical signals. Curr. Biol. 21,
degenerate human achilles tendon. Arthritis Rheum. 54, 832-842. 933-941.
Juneja, S. C. and Veillette, C. (2013). Defects in tendon, ligament, and enthesis in Maeda, E., Ye, S., Wang, W., Bader, D. L., Knight, M. M. and Lee, D. A. (2012).
response to genetic alterations in key proteoglycans and glycoproteins: a review. Gap junction permeability between tenocytes within tendon fascicles is
Arthritis 2013, 154812. suppressed by tensile loading. Biomech. Model. Mechanobiol. 11, 439-447.
Kahn, J., Shwartz, Y., Blitz, E., Krief, S., Sharir, A., Breitel, D. A., Rattenbach, R., Maeda, E., Sugimoto, M. and Ohashi, T. (2013). Cytoskeletal tension modulates
Relaix, F., Maire, P., Rountree, R. B. et al. (2009). Muscle contraction is MMP-1 gene expression from tenocytes on micropillar substrates. J. Biomech. 46,
necessary to maintain joint progenitor cell fate. Dev. Cell 16, 734-743. 991-997.
Kang, J. S., Alliston, T., Delston, R. and Derynck, R. (2005). Repression of Runx2 Magne, D. and Bougault, C. (2015). What understanding tendon cell differentiation
function by TGF-beta through recruitment of class II histone deacetylases by can teach us about pathological tendon ossification. Histol. Histopathol. 30,
Smad3. EMBO J. 24, 2543-2555. 901-910.
Kannus, P. (2000). Structure of the tendon connective tissue. Scand. J. Med. Sci. Marturano, J. E., Xylas, J. F., Sridharan, G. V., Georgakoudi, I. and Kuo, C. K.
Sports 10, 312-320. (2014). Lysyl oxidase-mediated collagen crosslinks may be assessed as markers
Kannus, P., Jozsa, L., Ja rvinen, T. A. H., Ja rvinen, T. L. N., Kvist, M., Natri, A. of functional properties of tendon tissue formation. Acta Biomater. 10, 1370-1379.
and Ja rvinen, M. (1998). Location and distribution of non-collagenous matrix Mayer, U., Saher, G., Fa ssler, R., Bornemann, A., Echtermeyer, F., von der
proteins in musculoskeletal tissues of rat. Histochem. J. 30, 799-810. Mark, H., Miosge, N., Po schl, E. and von der Mark, K. (1997). Absence of
Kardon, G. (1998). Muscle and tendon morphogenesis in the avian hind limb. integrin alpha 7 causes a novel form of muscular dystrophy. Nat. Genet. 17,
Development 125, 4019-4032. 318-323.
Katzel, E. B., Wolenski, M., Loiselle, A. E., Basile, P., Flick, L. M., Langstein, Munger, J. S. and Sheppard, D. (2011). Cross talk among TGF- signaling
H. N., Hilton, M. J., Awad, H. A., Hammert, W. C. and OKeefe, R. J. (2011). pathways, integrins, and the extracellular matrix. Cold Spring Harb. Perspect. Biol.
Impact of Smad3 loss of function on scarring and adhesion formation during 3, a005017.
tendon healing. J. Orthop. Res. 29, 684-693. Murchison, N. D., Price, B. A., Conner, D. A., Keene, D. R., Olson, E. N., Tabin,
Kieny, M. and Chevallier, A. (1979). Autonomy of tendon development in the C. J. and Schweitzer, R. (2007). Regulation of tendon differentiation by scleraxis
embryonic chick wing. J. Embryol. Exp. Morphol. 49, 153-165. distinguishes force-transmitting tendons from muscle-anchoring tendons.
DEVELOPMENT

Kimura, W., Machii, M., Xue, X., Sultana, N., Hikosaka, K., Sharkar, M. T. K., Development 134, 2697-2708.
Uezato, T., Matsuda, M., Koseki, H. and Miura, N. (2011). Irxl1 mutant mice Mustonen, E., Ruskoaho, H. and Rysa , J. (2012). Thrombospondin-4, tumour
show reduced tendon differentiation and no patterning defects in musculoskeletal necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor Fn14:
system development. Genesis 49, 2-9. Novel extracellular matrix modulating factors in cardiac remodelling. Ann. Med. 44,
Kjaer, M. (2004). Role of extracellular matrix in adaptation of tendon and skeletal 793-804.
muscle to mechanical loading. Physiol. Rev. 84, 649-698. Noden, D. M. (1988). Interactions and fates of avian craniofacial mesenchyme.
Koch, M., Schulze, J., Hansen, U., Ashwodt, T., Keene, D. R., Brunken, W. J., Development 103 Suppl., 121-140.
Burgeson, R. E., Bruckner, P. and Bruckner-Tuderman, L. (2004). A novel Noden, D. M. and Trainor, P. A. (2005). Relations and interactions between cranial
marker of tissue junctions, collagen XXII. J. Biol. Chem. 279, 22514-22521. mesoderm and neural crest populations. J. Anat. 207, 575-601.
Koshida, S., Kishimoto, Y., Ustumi, H., Shimizu, T., Furutani-Seiki, M., Oka, K., Oka, S., Hosokawa, R., Bringas, P., Brockhoff, H. C., Nonaka, K. and
Kondoh, H. and Takada, S. (2005). Integrin alpha5-dependent fibronectin Chai, Y. (2008). TGF-beta mediated Dlx5 signaling plays a crucial role in osteo-

4202
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

chondroprogenitor cell lineage determination during mandible development. Dev. Soeda, T., Deng, J. M., de Crombrugghe, B., Behringer, R. R., Nakamura, T.
Biol. 321, 303-309. and Akiyama, H. (2010). Sox9-expressing precursors are the cellular origin of
Otabe, K., Nakahara, H., Hasegawa, A., Matsukawa, T., Ayabe, F., Onizuka, N., the cruciate ligament of the knee joint and the limb tendons. Genesis 48,
Inui, M., Takada, S., Ito, Y., Sekiya, I. et al. (2015). Transcription factor Mohawk 635-644.
controls tenogenic differentiation of bone marrow mesenchymal stem cells in vitro Spanoudes, K., Gaspar, D., Pandit, A. and Zeugolis, D. I. (2014). The biophysical,
and in vivo. J. Orthop. Res. 33, 1-8. biochemical, and biological toolbox for tenogenic phenotype maintenance in vitro.
Pan, T.-C., Zhang, R.-Z., Markova, D., Arita, M., Zhang, Y., Bogdanovich, S., Trends Biotechnol. 32, 474-482.
Khurana, T. S., Bo nnemann, C. G., Birk, D. E. and Chu, M.-L. (2013). Subramanian, A. and Schilling, T. F. (2014). Thrombospondin-4 controls matrix
COL6A3 protein deficiency in mice leads to muscle and tendon defects similar to assembly during development and repair of myotendinous junctions. eLife 2014,
human collagen VI congenital muscular dystrophy. J. Biol. Chem. 288, e02372.
14320-14331. Subramanian, A., Wayburn, B., Bunch, T. and Volk, T. (2007). Thrombospondin-
Parkinson, J., Samiric, T., Ilic, M. Z., Cook, J. and Handley, C. J. (2011). mediated adhesion is essential for the formation of the myotendinous junction in
Involvement of proteoglycans in tendinopathy. J. Musculoskelet. Neuronal Drosophila. Development 134, 1269-1278.
Interact. 11, 86-93. Sugimoto, Y., Takimoto, A., Akiyama, H., Kist, R., Scherer, G., Nakamura, T.,
Parsons, M. J., Pollard, S. M., Sau de, L., Feldman, B., Coutinho, P., Hirst, Hiraki, Y. and Shukunami, C. (2013). Scx+/Sox9+ progenitors contribute to
E. M. A. and Stemple, D. L. (2002). Zebrafish mutants identify an essential role for the establishment of the junction between cartilage and tendon/ligament.
laminins in notochord formation. Development 129, 3137-3146. Development 140, 2280-2288.
Pingel, J., Lu, Y., Starborg, T., Fredberg, U., Langberg, H., Nedergaard, A., Takimoto, A., Oro, M., Hiraki, Y. and Shukunami, C. (2012). Direct conversion of
Weis, M., Eyre, D., Kjaer, M. and Kadler, K. E. (2014). 3-D ultrastructure and tenocytes into chondrocytes by Sox9. Exp. Cell Res. 318, 1492-1507.
collagen composition of healthy and overloaded human tendon: evidence of Thomopoulos, S., Williams, G. R., Gimbel, J. A., Favata, M. and Soslowsky, L. J.
tenocyte and matrix buckling. J. Anat. 224, 548-555. (2003). Variation of biomechanical, structural, and compositional properties along
Popov, C., Burggraf, M., Kreja, L., Ignatius, A., Schieker, M. and Docheva, D. the tendon to bone insertion site. J. Orthop. Res. 21, 413-419.
(2015). Mechanical stimulation of human tendon stem/progenitor cells results in Thorpe, C. T., Birch, H. L., Clegg, P. D. and Screen, H. R. C. (2013). The role of the
upregulation of matrix proteins, integrins and MMPs, and activation of p38 and non-collagenous matrix in tendon function. Int. J. Exp. Pathol. 94, 248-259.
ERK1/2 kinases. BMC Mol. Biol. 16, 1219. Tokita, M. and Schneider, R. A. (2009). Developmental origins of species-specific
Pryce, B. A., Watson, S. S., Murchison, N. D., Staverosky, J. A., Du nker, N. and muscle pattern. Dev. Biol. 331, 311-325.
Schweitzer, R. (2009). Recruitment and maintenance of tendon progenitors by Tome , F. M., Evangelista, T., Leclerc, A., Sunada, Y., Manole, E., Estournet, B.,
TGFbeta signaling are essential for tendon formation. Development 136, Barois, A., Campbell, K. P. and Fardeau, M. (1994). Congenital muscular
1351-1361. dystrophy with merosin deficiency. C. R. Acad. Sci. III. 317, 351-357.
Rifkin, D. B. (2005). Latent transforming growth factor-beta (TGF-beta) Tozer, S. and Duprez, D. (2005). Tendon and ligament: development, repair and
binding proteins: orchestrators of TGF-beta availability. J. Biol. Chem. 280, disease. Birth Defects Res. C. Embryo Today 75, 226-236.
7409-7412. Turner, D. C., Lawton, J., Dollenmeier, P., Ehrismann, R. and Chiquet, M.
Rooney, J. E., Welser, J. V., Dechert, M. A., Flintoff-Dye, N. L., Kaufman, S. J. (1983). Guidance of myogenic cell migration by oriented deposits of fibronectin.
and Burkin, D. J. (2006). Severe muscular dystrophy in mice that lack dystrophin
Dev. Biol. 95, 497-504.
and alpha7 integrin. J. Cell Sci. 119, 2185-2195. Vaz, R., Martins, G. G., Thorsteinsdo ttir, S. and Rodrigues, G. (2012).
Rooney, J. E., Knapp, J. R., Hodges, B. L., Wuebbles, R. D. and Burkin, D. J.
Fibronectin promotes migration, alignment and fusion in an in vitro myoblast cell
(2012). Laminin-111 protein therapy reduces muscle pathology and improves
model. Cell Tissue Res. 348, 569-578.
viability of a mouse model of merosin-deficient congenital muscular dystrophy.
Veit, G., Hansen, U., Keene, D. R., Bruckner, P., Chiquet-Ehrismann, R.,
Am. J. Pathol. 180, 1593-1602.
Chiquet, M. and Koch, M. (2006). Collagen XII interacts with avian tenascin-X
Ros, M. A., Rivero, F. B., Hinchliffe, J. R. and Hurle, J. M. (1995).
through its NC3 domain. J. Biol. Chem. 281, 27461-27470.
Immunohistological and ultrastructural study of the developing tendons of the
Volk, T. and VijayRaghavan, K. (1994). A central role for epidermal segment border
avian foot. Anat. Embryol. 192, 483-496.
cells in the induction of muscle patterning in the Drosophila embryo. Development
Rui, Y. F., Lui, P. P. Y., Wong, Y. M., Tan, Q. and Chan, K. M. (2013). Altered fate of
120, 59-70.
tendon-derived stem cells isolated from a failed tendon-healing animal model of
Wa lchli, C., Koch, M., Chiquet, M., Odermatt, B. F. and Trueb, B. (1994). Tissue-
tendinopathy. Stem Cells Dev. 22, 1076-1085.
specific expression of the fibril-associated collagens XII and XIV. J. Cell Sci. 107,
Rullman, E., Norrbom, J., Stro mberg, A., Wgsa ter, D., Rundqvist, H., Haas, T.
669-681.
and Gustafsson, T. (2009). Endurance exercise activates matrix
Wall, M. E. and Banes, A. J. (2005). Early responses to mechanical load in tendon:
metalloproteinases in human skeletal muscle. J. Appl. Physiol. 106, 804-812.
Schessl, J., Zou, Y. and Bo nnemann, C. G. (2006). Congenital muscular Role for calcium signaling, gap junctions and intercellular communication.
dystrophies and the extracellular matrix. Semin. Pediatr. Neurol. 13, 80-89. J. Musculoskelet. Neuronal Interact. 5, 70-84.
Schoenebeck, J. J., Hutchinson, S. A., Byers, A., Beale, H. C., Carrington, B., Wang, J. H.-C., Guo, Q. and Li, B. (2012). Tendon biomechanics and
Faden, D. L., Rimbault, M., Decker, B., Kidd, J. M., Sood, R. et al. (2012). mechanobiology a minireview of basic concepts and recent advancements.
Variation of BMP3 contributes to dog breed skull diversity. PLoS Genet. 8, J. Hand Ther. 25, 133-141.
e1002849. Weinreb, J. H., Sheth, C., Apostolakos, J., McCarthy, M. B., Barden, B., Cote,
Schwartz, A. G., Pasteris, J. D., Genin, G. M., Daulton, T. L. and Thomopoulos, M. P. and Mazzocca, A. D. (2014). Tendon structure, disease, and imaging.
S. (2012). Mineral distributions at the developing tendon enthesis. PLoS ONE 7, Muscles. Ligaments Tendons J. 4, 66-73.
e48630. Willett, T. L., Labow, R. S., Aldous, I. G., Avery, N. C. and Lee, J. M. (2010).
Schwartz, A. G., Lipner, J. H., Pasteris, J. D., Genin, G. M. and Thomopoulos, S. Changes in collagen with aging maintain molecular stability after overload:
(2013). Muscle loading is necessary for the formation of a functional tendon evidence from an in vitro tendon model. J. Biomech. Eng. 132, 031002.
enthesis. Bone 55, 44-51. Wipff, P.-J., Rifkin, D. B., Meister, J.-J. and Hinz, B. (2007). Myofibroblast
Schweitzer, R., Chyung, J. H., Murtaugh, L. C., Brent, A. E., Rosen, V., Olson, contraction activates latent TGF-beta1 from the extracellular matrix. J. Cell Biol.
E. N., Lassar, A. and Tabin, C. J. (2001). Analysis of the tendon cell fate using 179, 1311-1323.
Scleraxis, a specific marker for tendons and ligaments. Development 128, Yang, G., Rothrauff, B. B. and Tuan, R. S. (2013). Tendon and ligament
3855-3866. regeneration and repair: clinical relevance and developmental paradigm. Birth
Schweitzer, R., Zelzer, E. and Volk, T. (2010). Connecting muscles to tendons: Defects Res. C. Embryo Today 99, 203-222.
tendons and musculoskeletal development in flies and vertebrates. Development Yeung, C.-Y. C., Gossan, N., Lu, Y., Hughes, A., Hensman, J. J., Bayer, M. L.,
137, 2807-2817. Kjr, M., Kadler, K. E. and Meng, Q.-J. (2014). Gremlin-2 is a BMP antagonist
Shukunami, C., Takimoto, A., Oro, M. and Hiraki, Y. (2006). Scleraxis positively that is regulated by the circadian clock. Sci. Rep. 4, 5183.
DEVELOPMENT

regulates the expression of tenomodulin, a differentiation marker of tenocytes. Yoon, J. H. and Halper, J. (2005). Tendon proteoglycans: biochemistry and
Dev. Biol. 298, 234-247. function. J. Musculoskelet. Neuronal Interact. 5, 22-34.
Shwartz, Y., Blitz, E. and Zelzer, E. (2013). One load to rule them all: mechanical Yu, Q. and Stamenkovic, I. (2000). Cell surface-localized matrix mealloproteinase-
control of the musculoskeletal system in development and aging. Differentiation 9 proteolytically activates TGF-beta and promotes tumor invasion and
86, 104-111. angiogenesis. Genes Dev. 14, 163-176.
Snow, C. J. and Henry, C. A. (2009). Dynamic formation of microenvironments at Yurchenco, P. D., Amenta, P. S. and Patton, B. L. (2004). Basement membrane
the myotendinous junction correlates with muscle fiber morphogenesis in assembly, stability and activities observed through a developmental lens. Matrix
zebrafish. Gene Expr. Patterns 9, 37-42. Biol. 22, 521-538.
Snow, C. J., Peterson, M. T., Khalil, A. and Henry, C. A. (2008). Muscle Zelzer, E., Blitz, E., Killian, M. L. and Thomopoulos, S. (2014). Tendon-to-bone
development is disrupted in zebrafish embryos deficient for fibronectin. Dev. Dyn. attachment: from development to maturity. Birth Defects Res. C Embryo Today
237, 2542-2553. 102, 101-112.

4203
REVIEW Development (2015) 142, 4191-4204 doi:10.1242/dev.114777

Zhang, J. and Wang, J. H. C. (2010). Mechanobiological response of tendon stem assembly of collagen fibrils and acquisition of biomechanical properties during
cells: implications of tendon homeostasis and pathogenesis of tendinopathy. tendon development. J. Cell Biochem. 98, 1436-1449.
J. Orthop. Res. 28, 639-643. Zhou, B., Zhou, Y. and Tang, K. (2014). An overview of structure, mechanical
Zhang, J. and Wang, J. H.-C. (2013). The effects of mechanical loading on tendons properties, and treatment for age-related tendinopathy. J. Nutr. Health Aging 18,
an in vivo and in vitro model study. PLoS ONE 8, e71740. 441-448.
Zhang, J. and Wang, J. H.-C. (2015). Moderate exercise mitigates the detrimental Zou, Y., Zwolanek, D., Izu, Y., Gandhy, S., Schreiber, G., Brockmann, K.,
effects of aging on tendon stem cells. PLoS ONE 10, e0130454. Devoto, M., Tian, Z., Hu, Y., Veit, G. et al. (2014). Recessive and dominant
Zhang, G., Ezura, Y., Chervoneva, I., Robinson, P. S., Beason, D. P., Carine, mutations in COL12A1 cause a novel EDS/myopathy overlap syndrome in
E. T., Soslowsky, L. J., Iozzo, R. V. and Birk, D. E. (2006). Decorin regulates humans and mice. Hum. Mol. Genet. 23, 2339-2352.

DEVELOPMENT

4204

Potrebbero piacerti anche