Open access peer-reviewed chapter

The Extracellular Matrix Complexome from Skeletal Muscle

Written By

Sandra Murphy and Kay Ohlendieck

Submitted: 08 October 2015 Reviewed: 15 January 2016 Published: 15 June 2016

DOI: 10.5772/62253

From the Edited Volume

Composition and Function of the Extracellular Matrix in the Human Body

Edited by Francesco Travascio

Chapter metrics overview

3,016 Chapter Downloads

View Full Metrics

Abstract

The various layers of the extracellular matrix, forming the endomysium, perimysium and epimysium of skeletal muscles, provide essential structural and mechanical support to contractile fibres. Crucial aspects of muscle elasticity and fibre contractility are dependent on proper cell–matrix interactions. A complex network of collagen fibres, non-fibrillar collagens, proteoglycans, matricellular proteins, matrix metalloproteinases, adhesion receptors and signalling molecules maintain the physical structure for force transmission within motor units, embed critical cellular structures such as capillaries and motor neurons, and enable essential sarcolemma-matrix adhesion processes and signalling cascades. The systems biological concept of protein complexomes, which assumes the existence of interconnectivities between large protein assemblies, can be readily applied to the proteins within the extracellular space of muscles. Recent proteomic studies confirm that the extracellular matrix complexome has considerable influence on the integrity and cellular functions of skeletal muscle fibres. Adaptations or changes in the organization of the extracellular matrix play a crucial role during fibre regeneration following injury, extensive neuromuscular activity or pathophysiological insults. This chapter outlines the molecular components of the matrisome from skeletal muscles and discusses the extracellular matrix in relation to myogenesis, maturation of motor units, adaptation to changed functional demands and myofibrosis in muscular disorders.

Keywords

  • collagen
  • fibrosis
  • matricellular
  • matrisome
  • proteoglycan

1. Introduction

In addition to the universal physiological, metabolic and regulatory challenges of almost all cellular entities in the body, muscle tissues have to constantly adapt to a variety of biological issues related to high energy demand, elevated levels of cellular stress and enormous physical strains during excitation–contraction–relaxation cycles [1]. The survival of skeletal muscle fibres therefore depends heavily on (i) a high degree of physiological adaptability, (ii) a unique level of tissue plasticity, (iii) efficient molecular chaperoning to prevent proteotoxic insults and (iv) a sophisticated repair machinery that can counter-act frequent cellular injuries [2]. A crucial stabilizing element that is intrinsically involved in this continuous maintenance of contractile tissues is the extracellular matrix (ECM) [3]. On the one hand, the complex layers of the muscle ECM provide the physical structure for force transmission between contracting fibres and their surrounding tissue environment [4], and on the other hand the ECM functions as an embedding medium for essential supportive components of muscles such as capillaries and motor neurons [5].

The composition and organization of the ECM adapts considerably in response to changed functional or structural demands during myogenesis, fibre maturation and exercise-induced changes [6]. During the natural aging process and in association with a variety of muscular disorders, a hyperactive connective tissue may trigger myofibrosis with a detrimental impact on muscle elasticity and fibre contractility [7]. Physiological or pathological changes in the muscle ECM frequently mirror the different phases of altered muscle structure and function [8]. The main component of the ECM is represented by collagen, which exits in a large number of isoforms that connect with proteoglycans, matricellular proteins and adhesion receptors to form an elaborate extracellular network and tight cell–matrix interactions [9].

This chapter provides an overview of the molecular components of the ECM from skeletal muscle and describes the proteomic concept of the ECM complexome. The formation, maturation and flexibility within the various layers of the ECM in developing, maturing and adapting skeletal muscles is outlined, as well as the crucial role of myofibrosis in neuromuscular pathology.

Advertisement

2. Molecular and cellular structure of the extracellular matrix

In skeletal muscles, the ECM is involved in a variety of processes during development, contractile maturation, fibre regeneration following injury, physiological adaptations to changed functional demands and the natural aging process [8]. As outlined in Figure 1, the ECM is a highly dynamic non-cellular system that undergoes frequent cycles of modifications, degradation and reassembly. The muscle ECM functions on the one hand as an embedding and stabilizing structural support and on the other hand as a cellular interaction and signalling medium. The molecular lattice of collagens and proteoglycans with its associated matricellular proteins, enzyme systems and adhesion receptors mediates various physiological and biochemical mechanisms, including (i) the overall maintenance of muscle tissue stability and elasticity, (ii) the mechanical transduction of force from the contractile fibres to their anchoring tissues, (iii) cytoskeletal coupling to enable the efficient execution of frequent excitation–contraction–relaxation cycles, (iv) the provision of signalling pathways at the fibre periphery, (v) the preservation of neuromuscular homeostasis, and (vi) the physical scaffold and embedding medium of cellular constituents, such as contractile fibres, capillaries, motor neurons and satellite cell pools. Cell–matrix interactions are of central importance during cell adhesion and cell migration and thus essential during both embryonic and adult myogenesis.

Figure 1.

Overview of the complex functions and interactions of the extracellular matrix from skeletal muscles.

The distinct layers of the ECM that surround muscle fibres, muscle fascicles and the entire skeletal muscle are formed by the endomysium, perimysium and epimysium, respectively [3]. Figure 2 shows diagrammatically the arrangements of the ECM from skeletal muscles and lists the main molecular constituents, including various isoforms of collagen, proteoglycans, matricellular proteins, crosslinking proteins and matrix metalloproteinases. Crucial adhesion systems that maintain sarcolemma–matrix interactions are marked. They include the collagen-laminin-α/β-dystroglycan-dystrophin/utrophin axis and the collagen–fibronectin–integrin axis that link the basal lamina via the plasmalemma to the underlying membrane cytoskeleton [10].

The stabilizing linkage between the outside and inside of muscle cells, provided by the ECM–sarcolemma–cytoskeleton axis, is of critical importance for maintaining normal contractile functions [8]. Primary or secondary abnormalities in individual binding partners of these surface complexes may result in severe neuromuscular disorders, as discussed in below section on the role of the ECM in skeletal muscle pathology and myofibrosis.

Collagens form tight helical structures and function as the main structural protein species in the extracellular space. Muscle-associated collagens are highly abundant in the interstitial matrix, ECM microfibrils and the basal lamina [11]. Collagen isoform COL I, the most abundant protein in the mammalian body [12], is the primary collagen in the perimysium and tendon. The interstitial matrix contains mostly collagen COL I, COL III and COL V. Minor types of collagens in this extracellular region are COL XI, XII, XIV, XV and XVIII and are mostly expressed during muscle development [3]. The main structural constituent in ECM microfibrils is collagen isoform COL VI. Microfibrils provide structural support during the physical strains of continuing contraction–relaxation cycles. Directly overlaying the sarcolemma membrane is the basal lamina consisting of the main non-fibrillar collagen isoform COL IV and a few minor constituents, including COL VI, XV and XVIII. The collagen network of the basement membrane interacts with two crucial plasmalemma adhesion complexes, the integrin complex and the dystrophin–glycoprotein complex [10]. These sarcolemma-bridging protein assemblies form the laminin–dystroglycan axis [13] and the fibronectin-integrin (α7β1) axis [14], whereby the ECM glycoprotein fibronectin mediates the connection between laminin-211 (α2β1γ1 merosin) and collagen COL IV [15].

Figure 2

Diagrammatic presentation of the various layers of the extracellular matrix (ECM) from skeletal muscle and an overview of the molecular components that form the interstitial matrix, microfibrils and the basal lamina. Abbreviations: COL, collagen; DG, dystroglycan; FN, fibronectin; LAM, laminin; INT, integrin; MMP, matrix metalloproteinase; NMJ, neuromuscular junction; PG, proteoglycan; PRELP, proline-arginine-rich end leucine-rich repeat protein; SLRP, small leucine-rich repeat proteoglycans; TIMP, tissue inhibitors of matrix metalloproteinases.

A variety of regulatory ECM proteins are involved in matrix assembly and the modulation of cell–matrix interactions, such as dermatopontin, nidogen/entactin, periostin (osteoblast-specific factor OSF-2) and osteopontin [1618]. Matricellular proteins represent non-architectural ECM components and are crucial factors during muscle development and fibre repair. Matrix metalloproteinases are an important class of ECM-associated enzymes that regulate the degradation of ECM proteins and support tissue integrity during phases of collagen deposition and muscle regeneration [19]. The main isoforms present in skeletal muscles are matrix metalloproteinases MMP-1, MMP-2, MMP-9, MMP-10 and MMP-13 [20,21]. The tissue inhibitors of matrix metalloproteinases, named TIMP, are endogenous regulatory factors involved in the formation, adaptation and controlled degradation of the ECM [22]. TIMP molecules play a crucial role in the migration and differentiation of muscle stem cells during regeneration following cellular injury [19].

A large variety of proteoglycans fill the gaps between collagen molecules and thereby form an integral part of the highly complex ECM structure. Proteoglycan molecules are highly glycosylated with glycosaminoglycans at multiple sites along the peptide backbone [23]. Skeletal muscles contain small leucine-rich repeat proteoglycans (SLRP), heparan sulfate proteoglycans and chondroitin sulphate proteoglycans. Muscle-associated proteoglycans of the type SLRP are asporin, biglycan, decorin, mimecan (osteoglycin), fibromodulin and lumican. Asporin is mostly found in the cartilage matrix. Biglycan is a small SLRP-type proteoglycan that interacts with α-sarcoglycan and γ-sarcoglycan of the dystrophin-glycoprotein complex [24]. Decorin is the primary proteoglycan molecule of the perimysium and tendon structures [25]. Fibromodulin is involved in collagen fibril formation, which is illustrated by the biomedical fact that fibromodulin-deficient tendons exhibit abnormal collagen fibrils [26]. The heparan sulfate proteoglycan syndecan is transiently up-regulated during tissue differentiation and is involved in stem cell maintenance and muscle regeneration [27]. Perlecan is located to the basement membrane and its expression is also transiently increased during muscle differentiation [28]. The chondroitin sulphate proteoglycan named aggrecan forms large aggregates in cartilage [29]. A proline-arginine-rich end leucine-rich repeat protein (PRELP) is presented by prolargin of the basal lamina. At the highly differentiated neuromuscular junction region, the large proteoglycan molecule agrin is present and forms via α-dystroglycan a tight linkage to the utrophin-glycoprotein complex. Agrin is essential for the normal development of the neuromuscular junction and agrin-induced clustering processes are crucial for the anchoring of the acetylcholine receptor complex in the junctional folds [30].

Advertisement

3. Skeletal muscle development and the extracellular matrix

Skeletal muscle fibres derive from the mesoderm [31] and represent one of the most abundant cell types in the body. Myofibres play a key physiological role in the provision and regulation of locomotion, breathing, postural control, heat homeostasis and metabolic integration [3234]. The development of contractile fibres is a highly complex process and changes in the ECM play an essential role during myogenesis. Adaptations occur at the level of the basal lamina, the interstitial ECM and the collagen-rich tendon and encompass a variety of fundamental processes of development, such as the determination of cell fate, as well as proliferation, cell division, patterning and tissue transitions [35]. The most critical developmental processes occur during the initial activation of precursor cells, various differentiation steps and the final maturation of innervated myofibres. These major developmental mechanisms are regulated by a large number of genetic and signalling factors [36]. Many specialized extracellular components are involved in muscle development, which is reflected by the transient expression patterns of certain ECM molecules, such as the minor collagen isoforms COL XI, XII, XIV, XV and XVIII [3].

The molecular and cellular events that occur during the establishment of the myogenic cell lineage and resulting formation of multi-nucleated contractile fibres entail initially a fibrillar and fibronectin-rich matrix. In developmental terms, almost all myofibres of the skeletal musculature derive from mesodermal structures named somites that develop during early embryonic segmentation on both sides of the neural tube [3739]. The developing ECM of somites is represented by a fibronectin core, a basement membrane and an outer fibronectin-containing matrix [35]. Figure 3 outlines the initiation and control of embryonic and adult myogenesis by myogenic factors and through a complex series of spatio-temporal dependent signalling cascades. Fibronectin and its interactions with the integrin complex play a central role in polarizing and guiding somitic cells [4042] and the ECM is crucial for somite formation and as a guiding cue during morphogenesis [4345]. The small SLRP-type proteoglycan decorin was shown to be majorly involved in skeletal muscle development by promoting proliferation and differentiation of muscle cells through suppressing myostatin activity [4648].

Figure 3.

Outline of the initiation and control of embryonic and adult myogenesis. Shown is the transition from mesodermal progenitor cells to myoblasts, followed by cellular fusion to produce multi-nucleated myotubes and finally innervated and functional myofibres. The critical role of stem cells during both embryonic development and regeneration of injured adult muscle fibres is highlighted, as well as changes in the ECM. Fibronectin (FN) and its interactions with the integrin complex are of central importance for polarizing and guiding somitic cells and the proteoglycan decorin is majorly involved in myogenesis by promoting proliferation and differentiation through supressing myostatin activity. Changes in the ECM and the formation of a collagen (COL) matrix during muscle development are marked.

The key regulator of early myogenesis that initiates the developmental commitment into the myogenic cell lineage is the paired-type homeobox gene PAX3 [4951]. Interestingly, another member of the PAX transcription factor family, the PAX3 orthologue PAX7, acts as a regulator of mature skeletal muscle regeneration and postnatal growth mechanisms [52]. Following the induction of mesodermal precursors, the subsequent segmentation into somites and the formation of the primary myotome involves a variety of signalling molecules and transcription factors, such as the secreted and lipid-modified family of Wnt-glycoproteins and the large group of myogenic basic helix-loop-helix muscle regulatory factors, such as MyoD, Myf5, MRF4 and myogenin [5355]. Myogenic factors act at multiple regulatory points during embryonic myogenesis, whereby the overall genetic pathway that is responsible for the transcriptional activation of skeletal muscle-specific genes is highly complex and partially redundant [56]. A key step during muscle development is the fusion of myogenic cells that result in the formation of innervated and multi-nucleated myofibres (Figure 3). Although specific regulatory processes differ between embryonic, foetal, postnatal and mature regenerative myogenesis [57], the basic biological mechanisms of skeletal muscle development and injury-related adult muscle regeneration are extraordinarily similar [58,59]. A large pool of satellite cells provides a high level of regenerative capacity of the matured post-mitotic fibres and is located between the sarcolemma and the basal lamina [60]. These mono-nucleated myogenic stem cells are activated during cycles of fibre regeneration and cellular maintenance. To provide maximum skeletal muscle performance, activated stem cells undergo a complex pattern of proliferation, differentiation and cellular fusion to form multi-nucleated and functional contractile myofibres [61]. Following maturation, the tissue mass of skeletal muscles is then regulated by catabolic and anabolic mechanisms that include signalling factors such as NF-kB and FoxO, as well as the mTOR pathway [62].

The systematic profiling of myogenesis using mass spectrometry-based proteomics has covered postnatal growth and development [63,64], but focused mostly on studying cell culture models [6567] and specifically the skeletal muscle secretome during myoblast differentiation and myotube formation [6872]. The concept that skeletal muscle cells act as secretory tissues has recently been reviewed by Pedersen [73]. Secreted myokines probably exert autocrine, paracrine or endocrine effects within the neuromuscular system and also in relation to other organ systems [74]. The skeletal muscle secretome is estimated to consist of several hundred muscle-derived peptides and proteins [75]. During muscle development, changes in ECM proteins have recently been studied in relation to human myogenesis and established that altered protein expression underlies the dramatic phenotypic conversion of primary mono-nucleated muscle cells during differentiation to form multi-nucleated myotubes [76]. The temporal profiling of the human myoblast proteome during in vitro differentiation highlighted the importance of ECM rearrangement during early myogenesis and showed a drastic increase in key ECM components, including several α-isoforms of collagen COL VI and COL XVIII, as well as the heparan sulfate proteoglycan HSPG2 of the basement membrane, the elastin–microfibril interface–located ECM glycoprotein EMILIN2 and nidogen isoform NID2 [76]. These findings confirm the developmental concept that enhanced synthesis of ECM proteins occurs during the transition from myoblasts to syncytial myotubes [72] and that complex interactions at the cell–ECM interface facilitate the fusion of myoblasts [35].

Advertisement

4. Mature fibres, skeletal muscle plasticity and the extracellular matrix

Both, proteomic cataloguing studies of various skeletal muscle specimens [7782] and the comparative expression profiling of crude skeletal muscle preparations or subcellular fractions [8386] routinely identify ECM proteins that form the core complexes of the basal lamina, microfibrils and interstitial matrix [87]. Figure 4 shows a bioinformatics STRING [88] map of core ECM components from skeletal muscles. In mature skeletal muscles, frequently identified ECM molecules include collagens of the interstitial matrix (COL I, COL III and COL V), the microfibrillar collagen isoform COL VI, the non-fibrillar collagen isoform COL IV of the basement membrane, components of cell–ECM adhesion complexes (laminin, fibronectin, integrins, dystrophin, utrophin), regulatory ECM proteins (dermatopontin, nidogen, periostin and osteopontin), SLRP-type proteoglycans (asporin, biglycan, decorin, mimecan, fibromodulin and lumican), heparan sulfate proteoglycans (syndecan and perlecan), the chondroitin sulphate proteoglycan aggrecan, the PRELP-type proteoglycan prolargin and the neuromuscular junction-specific proteoglycan agrin, as well as matrix metalloproteinases and their inhibitors (MMP-1, MMP-2, MMP-9, MMP-10, MMP-13 and TIMPs) [7786]. Figure 4 includes the protein products of the following genes: BGN, SDC1, HSPG2, AGRN, FN1, DAG1, ACAN, DPT, POSTN, MMP1, MMP2, MMP9, MMP10, MMP13, TIMP1, COL1A1, COL1A2, COL6A1, COL6A2, COL3A1, COL5A1, COL4A4, LAMA2, LAMB1, LAMB2, LAMC1, NID1, ASPN, PRELP, PGS2, LUM, OGN, FMOD, ITGA7, ITGB1. This encompasses essential members of the various collagen networks found in the basement membrane, the microfibrillar structures and the interstitial matrix. In addition, major proteoglycans, matricellular proteins and matrix metalloproteinases are shown, as well as the interaction sites between sarcolemmal adhesion receptor complexes and the ECM.

Individual skeletal muscles are characterized by their fibre type distribution pattern whereby the proportion of fast-twitching fibres, slow-twitching fibres and hybrid fibres is highly adaptable and changes according to specific physiological, biochemical and/or metabolic demands [89]. Alterations in physical activity affect the molecular and cellular composition of the neuromuscular system, including hypertrophy, i.e. the increase in fibre size and hyperplasia, i.e. the increase in fibre number [90]. The plasticity of the neuromuscular system is a well-established physiological concept. Endurance training is associated with an increased aerobic capacity and elevated utilization of fatty acid oxidation [91]. In contrast, sprint training triggers higher activities of the glycolytic and phosphocreatine pathways and enhances carbohydrate metabolism [92]. Prior to systematic proteomic studies, a large number of biochemical, cell biological and physiological studies have established ECM changes in response to exercise [9395], including collagens, adhesion receptors, growth factors, matricellular proteins and matrix metalloproteinases [96100]. Neuromuscular unloading clearly depresses collagen COL I and COL III production and reloading enhances collagen expression in fast muscles [101]. The mass spectrometric analysis of exercise indicates proteome-wide changes in the graded response of skeletal muscles to physical exercise using different training regimes [102,103]. While moderate-intensity exercise causes a shift to a more fatigue-resistant and a slower-contracting skeletal muscle phenotype, interval-exercise training is associated with changes in post-translational modifications of metabolic enzymes [104106]. The proteomic analysis of skeletal muscle plasticity in relation to acute versus chronic exercise was recently determined using human vastus lateralis muscle biopsy specimens and label-free LC-MS/MS analysis [107]. While structural and mitochondrial proteins were shown to be increased after long-term exercise, components related to energy metabolism were decreased following short-term exercise. Moderate ECM changes were described for several α-chains of collagen VI, fibronectin and decorin [107].

Figure 4.

Bioinformatics STRING map of major components that form the core of the extracellular matrix (ECM) complexome. Shown are key proteins belonging to the collagen network of the basal lamina, microfibrils and the interstitial matrix, proteoglycans, matricellular proteins and matrix metalloproteinases. The interaction sites of the ECM with sarcolemmal adhesion receptor complexes are shown.

An interesting non-physiological system is presented by external chronic low-frequency stimulation of fast muscles. This electro-stimulation method causes the complete activation of all affected motor units to a maximum extent [108]. During fast-to-slow transitions, skeletal muscles show a remarkable adaptation and transform physiologically and biochemically into motor units with an improved resistance to fatigue [109]. Chronic low-frequency stimulated fast muscles are characterized by decreased fibre calibres, an increase in the time-to-peak twitch tension, an increase in half-relaxation time and a significant elevation of aerobic-oxidative capacity [110]. The proteomic analysis of continuous electro-stimulation at 10 Hz has demonstrated complex biochemical changes with a significant shift from glycolytic to more aerobic-oxidative metabolism [111,112]. The ECM of transforming skeletal muscle undergoes distinct changes and exhibits increased collagen levels [113,114]. In the stimulated latissimus dorsi model for testing the suitability of dynamic cardiomyoplasty to treat heart failure, the collagen content was shown to be significantly elevated in the paced muscle. Although the chronically electro-stimulated muscle increased the level of fatigue resistance, distal regions of the paced latissimus dorsi muscle were characterized by muscular atrophy and myofibrosis [114].

Advertisement

5. Neuromuscular disorders and the extracellular matrix

A general myopathological parameter of a variety of acquired and inherited muscle diseases [115], as well as the gradual loss of contractile strength during the natural aging process [116], is the progressive accumulation of ECM components, especially collagens [7]. Inflammatory processes and tissue infiltration often accompany the loss of skeletal muscle fibres. Increased levels of non-contractile entities, such as fibrous connective and fatty tissue, within the neuromuscular system are a key pathological factor in the dysregulation of skeletal muscle function. Myofibrosis often correlates with poor motor outcome in neuromuscular disorders, such as the progressive loss of muscle strength and concomitant endomysial changes in the X-linked inherited disorder Duchenne muscular dystrophy [117]. In muscle pathology, changes in ECM components can be differentiated as being a consequence of a primary defect in the matrisome of muscles, such as the ECM diseases Collagen IV myopathy [118,119] and LAMA2-related congenital myopathy [120,121], or a secondary response in the form of reactive myofibrosis, as is seen in dystrophinopathies [122,123].

The systematic profiling of changes in ECM components in Collagen IV myopathy and X-linked muscular dystrophy has resulted in interesting new findings in relation to primary ECM defects versus reactive fibrosis. Mutations in the genes encoding collagen isoform COL VI are the underlying cause of the severe UCMD type of Ullrich congenital muscular dystrophy and the milder BM type of Bethlem myopathy. Both disorders are characterized by skeletal muscle wasting, cycles of cellular degeneration and regeneration, and the substitution of contractile fibres with fat and connective tissue [119]. The cell biological and proteomic analysis of mouse models and biopsy material from patients afflicted with Collagen IV myopathy revealed metabolic dysregulation, enhanced cellular stress, autophagic impairment and alterations in mechano-transduction signalling pathways [124,125]. In the case of X-linked muscular dystrophy, a large number of proteomic studies have surveyed secondary changes downstream of the primary abnormality in the membrane cytoskeletal protein dystrophin [126]. Muscular dystrophy-related changes affect energy metabolism, cellular signalling, the excitation–contraction–relaxation cycle, the stress response, the cytoskeletal network and the ECM [127]. The recent proteomic profiling of established genetic animal models of dystrophinopathy has revealed a drastic increase in various collagens, proteoglycans and matricellular proteins [8386,128,129]. Figure 5 outlines the histo- and pathobiochemical consequences of muscular changes and a hyperactive connective tissue in dystrophin-deficient skeletal muscles.

Figure 5.

Overview of reactive myofibrosis in X-linked muscular dystrophy. The transforming growth factor TGF-β and the activity of matrix metalloproteinases (MMP) and tissue inhibitors (TIMPs) play a crucial role in ECM activation and modulation, which is characterized by increased levels of various collagens, proteoglycans, dermatopontin and periostin. Progressive ECM accumulation triggers a chronic replacement of muscle fibres by fibrotic scar tissue leading to a loss of muscle elasticity and contractile strength.

The simultaneous mass spectrometric analysis of dystrophin isoform Dp427 and collagen in moderately dystrophic mdx-4cv leg muscles revealed significant increases in collagens and associated ECM proteins, such as fibronectin, biglycan, asporin, decorin, prolargin, mimecan and lumican [85]. The pathoproteomic signature of the severely dystrophic mdx-4cv diaphragm included a significant increase in collagens and the related ECM proteins asporin, decorin, dermatopontin, prolargin and periostin [83]. Especially interesting was the proteomic identification of dermatopontin and periostin [83,128,129]. Dermatopontin, also named tyrosine-rich acidic matrix protein TRAMP [130], is involved in matrix assembly and cell–matrix interactions [17] via interactions with decorin, TGF-β and fibronectin [131]. High levels of dermatopontin in the dystrophic mdx diaphragm most likely reflect an increased demand for collagen matrix organization within Dp427-deficient fibres [128]. Periostin is a crucial matricellular protein of 93 kDa [18] that is involved in the regulation of the biomechanical properties of connective tissues and collagen fibrillogenesis [132]. Normally periostin is only temporally expressed in the muscle ECM during cellular differentiation and regeneration processes [133], making its drastic up-regulation a characteristic feature of dystrophic muscles [83,86]. Of diagnostic and therapeutic importance is the fact that muscle biopsies from Duchenne patients exhibit an elevated concentration of periostin and that the deletion of periostin clearly reduces dystrophic symptoms and myofibrosis in mice by modulating the TGF-β pathway [134]. Interestingly, laminin-deficient muscular dystrophy also shows dysregulation of matricellular proteins as an early pathophysiological feature [120,135]. Therefore altered levels of periostin and related matricellular proteins are good biomarker candidates for the characterization of myofibrosis in relation to inherited muscular dystrophies [136]. Although the natural aging process is also associated with increased collagen levels [116], which were also shown by proteomics [137], the collagen accumulation is much less pronounced in senescent muscles as compared to muscular dystrophy.

Advertisement

6. Conclusions

In conclusion, a dynamic balance exists within the ECM system from skeletal muscles. Highly regulated cycles of protein deposition, accumulation, remodelling and degradation occur in response to development, fibre transformation, neuromuscular loading, mechanical unloading, disease processes or aging. The main components of the ECM are represented by collagen fibres, non-fibrillar collagens, matricellular proteins, proteoglycans, matrix metalloproteinases, signalling molecules and adhesion complexes. The muscle ECM forms a structural scaffold that plays a central role in the maintenance of the physical structure of motor units and provides the framework for force transmission. The ECM is also involved in signalling cascades and adhesion processes at the sarcolemma–matrix interface. The systematic mass spectrometry-based proteomic analysis of the muscle ECM has established an enormous complexity and interconnectivity of matrix proteins and confirmed the dynamic nature of collagen networks and its associated proteoglycans in health and disease. Swift adaptations or alterations in the arrangement of the ECM have been established to occur during myogenesis, fibre regeneration, increased neuromuscular activity or pathological muscle wasting. Hence, concentration changes in ECM proteins are useful indicators for studying basic cell biological or pathophysiological processes in skeletal muscles. In the future, distinct ECM molecules may be useful for designing improved diagnostic, prognostic or therapy-monitoring approaches to study neuromuscular alterations.

Advertisement

Acknowledgments

Research in the author’s laboratory was supported by project grants from the Deutsche Duchenne Stiftung aktion benni & co e.V., Muscular Dystrophy Ireland and the Hume Scholarship programme of Maynooth University, as well as the Irish Higher Education Authority (HEA). The Programme for Research in Third Level Institutions PRTLI Cycle 5 is co-funded by the Irish Government and the European Union under Ireland’s EU Structural Funds Programme 2007-2013. We thank Prof. Dieter Swandulla (University of Bonn), Prof. Heinrich Brinkmeier (University of Greifswald) and Dr. Paula Meleady (Dublin City University) for their continued support of our studies into the mechanisms of muscle fibrosis.

References

  1. 1. Pette D. The adaptive potential of skeletal muscle fibers. Canadian Journal of Applied Physiology. 2002 Aug;27(4):423-48.
  2. 2. Blaauw B, Schiaffino S, Reggiani C. Mechanisms modulating skeletal muscle phenotype. Comprehensive Physiology. 2013 Oct;3(4):1645-87.
  3. 3. Gillies AR, Lieber RL. Structure and function of the skeletal muscle extracellular matrix. Muscle & Nerve. 2011 Sep;44(3):318-31.
  4. 4. Kjaer M. Role of extracellular matrix in adaptation of tendon and skeletal muscle to mechanical loading. Physiological Reviews. 2004 Apr;84(2):649-98.
  5. 5. Purslow PP. The structure and functional significance of variations in the connective tissue within muscle. Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology. 2002 Dec;133(4):947-66.
  6. 6. Lund DK, Cornelison DD. Enter the matrix: shape, signal and superhighway. FEBS Journal. 2013 Sep;280(17):4089-99.
  7. 7. Lieber RL, Ward SR. Cellular mechanisms of tissue fibrosis. 4. Structural and functional consequences of skeletal muscle fibrosis. American Journal of Physiology: Cell Physiology. 2013 Aug;305(3):C241-52
  8. 8. Goody MF, Sher RB, Henry CA. Hanging on for the ride: adhesion to the extracellular matrix mediates cellular responses in skeletal muscle morphogenesis and disease. Developmental Biology. 2015 May;401(1):75-91.
  9. 9. Naba A, Hoersch S, Hynes RO. Towards definition of an ECM parts list: an advance on GO categories. Matrix Biology. 2012 Sep;31(7):371-2.
  10. 10. Grounds MD, Sorokin L, White J. Strength at the extracellular matrix-muscle interface. Scandinavian Journal of Medical Science in Sports. 2005 Dec;15(6):381-91.
  11. 11. Sweeney SM, Orgel JP, Fertala A, Mc Auliffe JD, TurnerKR, Di Lullo GA, Chen S, Antipova O, Perumal S, Ala-Kokko L, Forlino A, Cabral WA, Barnes AM, Marini JC, San Antonio JD. Candidate cell and matrix interaction domains on the collagen fibril, the predominant protein of vertebrates. Journal of Biological Chemistry. 2008 Jul;283(30):21187-97.
  12. 12. Stefanovic B. RNA protein interactions governing expression of the most abundant protein in human body, type I collagen. Wiley Interdisciplinary Reviews: RNA. 2013 Sep;4(5):535-45.
  13. 13. Ohlendieck K. Towards an understanding of the dystrophin–glycoprotein complex: linkage between the extracellular matrix and the membrane cytoskeleton in muscle fibers. European Journal of Cell Biology. 1996 Jan;69(1):1-10.
  14. 14. Burkin DJ, Kaufman SJ. The alpha7beta1 integrin in muscle development and disease. Cell and Tissue Research. 1999 Apr;296(1):183-90.
  15. 15. Holmberg J, Durbeej M. Laminin-211 in skeletal muscle function. Cell Adhesion & Migration. 2013 Jan;7(1):111-21.
  16. 16. Funanage VL, Smith SM, Minnich MA. Entactin promotes adhesion and long-term maintenance of cultured regenerated skeletal myotubes. Journal of Cellular Physiology. 1992 Feb;150(2):251-7.
  17. 17. Kato A, Okamoto O, Ishikawa K, Sumiyoshi H, Matsuo N, Yoshioka H, Nomizu M, Shimada T, Fujiwara S. Dermatopontin interacts with fibronectin, promotes fibronectin fibril formation, and enhances cell adhesion. Journal of Biological Chemistry. 2011 Apr;286(17):14861-9.
  18. 18. Norris RA, Damon B, Mironov V, Kasyanov V, Ramamurthi A, Moreno-Rodriguez R, Trusk T, Potts JD, Goodwin RL, Davis J, Hoffman S, Wen X, Sugi Y, Kern CB, Mjaatvedt CH, Turner DK, Oka T, Conway SJ, Molkentin JD, Forgacs G, Markwald RR. Periostin regulates collagen fibrillogenesis and the biomechanical properties of connective tissues. Journal of Cellular Biochemistry. 2007 Jun;101(3):695-711.
  19. 19. Alameddine HS. Matrix metalloproteinases in skeletal muscles: friends or foes? Neurobiology of Disease. 2012 Dec;48(3):508-18.
  20. 20. Chen X, Li Y. Role of matrix metalloproteinases in skeletal muscle: Migration, differentiation, regeneration and fibrosis. Cell Adhesion & Migration. 2009 Oct; 3(4): 337–341.
  21. 21. Lei H, Leong D, Smith LR, Barton ER. Matrix metalloproteinase 13 is a new contributor to skeletal muscle regeneration and critical for myoblast migration. American Journal of Physiology: Cell Physiology. 2013 Sep;305(5):C529-38.
  22. 22. Bellayr I, Holden K, Mu X, Pan H, Li Y. Matrix metalloproteinase inhibition negatively affects muscle stem cell behavior. International Journal of Clinical and. Experimental Pathology. 2013;6(2):124-41.
  23. 23. Heinegård D. Proteoglycans and more - from molecules to biology. International Journal of Experimental Pathology. 2009 Dec;90(6):575-86.
  24. 24. Nastase MV, Young MF, Schaefer L. Biglycan: a multivalent proteoglycan providing structure and signals. Journal of Histochemistry & Cytochemistry. 2012 Dec;60(12):963-75.
  25. 25. Goetsch KP, Kallmeyer K, Niesler CU. Decorin modulates collagen I-stimulated, but not fibronectin-stimulated, migration of C2C12 myoblasts. Matrix Biology. 2011 Mar;30(2):109-17.
  26. 26. Chakravarti S. Functions of lumican and fibromodulin: lessons from knockout mice. Glycoconjugate Journal. 2002 May;19(4-5):287-93.
  27. 27. Pisconti A, Bernet JD, Olwin BB. Syndecans in skeletal muscle development, regeneration and homeostasis. Muscles Ligaments and Tendons Journal. 2012 Jun 17;2(1):1-9.
  28. 28. Zoeller JJ, McQuillan A, Whitelock J, Ho SY, Iozzo RV. A central function for perlecan in skeletal muscle and cardiovascular development. Journal of Cell Biology. 2008 Apr;181(2):381-94.
  29. 29. Matuszewski PE, Chen YL, Szczesny SE, Lake SP, Elliott DM, Soslowsky LJ, Dodge GR. Regional variation in human supraspinatus tendon proteoglycans: decorin, biglycan, and aggrecan. Connective Tissue Research. 2012;53(5):343-8.
  30. 30. Campanelli JT, Roberds SL, Campbell KP, Scheller RH. A role for dystrophin-associated glycoproteins and utrophin in agrin-induced AChR clustering. Cell. 1994 Jun;77(5):663-74.
  31. 31. Buckingham M, Bajard L, Chang T, Daubas P, Hadchouel J, Meilhac S, Montarras D, Rocancourt D, Relaix F. The formation of skeletal muscle: from somite to limb. Journal of Anatomy. 2003 Jan;202(1):59-68.
  32. 32. Frontera WR, Ochala J. Skeletal muscle: a brief review of structure and function. Calcified Tissue International. 2015 Mar;96(3):183-95.
  33. 33. Mayeuf-Louchart A, Staels B, Duez H. Skeletal muscle functions around the clock. Diabetes, Obesity and Metabolism. 2015 Sep;17 Suppl 1:39-46.
  34. 34. Baskin K, Winders BR, Olson EN. Muscle as a “mediator” of systemic metabolism. Cell Metabolism. 2015 Feb 3;21(2):237-48.
  35. 35. Thorsteinsdóttir S, Deries M, Cachaço AS, Bajanca F. The extracellular matrix dimension of skeletal muscle development. Developmental Biology. 2011 Jun;354(2):191-207.
  36. 36. Comai G, Tajbakhsh S. Molecular and cellular regulation of skeletal myogenesis. Current Topics in Developmental Biology. 2014;110:1-73.
  37. 37. Christ B, Ordahl CP. Early stages of chick somite development. Anatomy and Embryology (Berlin). 1995 May;191(5):381-96.
  38. 38. PourquiéO. Vertebrate somitogenesis. Annual Review of Cell and Developmental Biology. 2001;17:311-50.
  39. 39. Hollway G, Currie P. Vertebrate myotome development. Birth Defects Research Part C Embryo Today. 2005 Sep;75(3):172-9.
  40. 40. Ostrovsky D, Cheney CM, Seitz AW, Lash JW. Fibronectin distribution during somitogenesis in the chick embryo. Cell Differentiation. 1983 Nov;13(3):217-23.
  41. 41. Ostrovsky D, Sanger JW, Lash JW. Somitogenesis in the mouse embryo. Cell Differentiation. 1988 Mar;23(1):17-25.
  42. 42. Lash JW, Ostrovsky D, Mittal B, Sanger JW. Alpha actinin distribution and extracellular matrix products during somitogenesis and neurulation in the chick embryo. Cell Motility. 1985;5(6):491-506.
  43. 43. Martins GG, Rifes P, Amândio R, Rodrigues G, Palmeirim I, Thorsteinsdóttir S. Dynamic 3D cell rearrangements guided by a fibronectin matrix underlie somitogenesis. PLoS One. 2009 Oct;4(10):e7429.
  44. 44. Girós A, Grgur K, Gossler A, Costell M. α5β1 integrin-mediated adhesion to fibronectin is required for axis elongation and somitogenesis in mice. PLoS One. 2011;6(7):e22002.
  45. 45. Lackner S, Schwendinger-Schreck J, Jülich D, Holley SA. Segmental assembly of fibronectin matrix requires rap1b and integrin α5. Developmental Dynamics. 2013 Feb;242(2):122-31.
  46. 46. Kishioka Y, Thomas M, Wakamatsu J, Hattori A, Sharma M, Kambadur R, Nishimura T. Decorin enhances the proliferation and differentiation of myogenic cells through suppressing myostatin activity. Journal of Cellular Physiology. 2008 Jun;215(3):856-67. doi: 10.1002/jcp.21371.
  47. 47. Velleman SG, Shin J, Li X, Song Y. Review: The skeletal muscle extracellular matrix: Possible roles in the regulation of muscle development and growth. Canadian Journal of Animal Science. 2012;92:1-10.
  48. 48. Zeng QJ, Wang LN, Shu G, Wang SB, Zhu XT, Gao P, Xi QY, Zhang YL, Zhang ZQ, Jiang QY. Decorin-induced proliferation of avian myoblasts involves the myostatin/Smad signaling pathway. Poultry Science. 2014 Jan;93(1):138-46.
  49. 49. Relaix F, Rocancourt D, Mansouri A, Buckingham M. A Pax3/Pax7-dependent population of skeletal muscle progenitor cells. Nature. 2005 Jun;435(7044):948-53.
  50. 50. Buckingham M, Relaix F. The role of Pax genes in the development of tissues and organs: Pax3 and Pax7 regulate muscle progenitor cell functions. Annual Review of Cell and Developmental Biology. 2007;23:645-73.
  51. 51. Buckingham M, Relaix F. PAX3 and PAX7 as upstream regulators of myogenesis. Seminars in Cell & Developmental Biology. 2015 Aug;44:115-25.
  52. 52. Oustanina S, Hause G, Braun T. Pax7 directs postnatal renewal and propagation of myogenic satellite cells but not their specification. EMBO Journal. 2004 Aug;23(16):3430-9.
  53. 53. Rawls A, Olson EN. Myo D meets its maker. Cell. 1997 Apr;89(1):5-8.
  54. 54. Molkentin JD, Olson EN. Defining the regulatory networks for muscle development. Current Opinion in Genetics & Development. 1996 Aug;6(4):445-53.
  55. 55. Cisternas P, Henriquez JP, Brandan E, Inestrosa NC. Wnt signaling in skeletal muscle dynamics: myogenesis, neuromuscular synapse and fibrosis. Molecular Neurobiology. 2014;49(1):574-89.
  56. 56. Tajbakhsh S, Rocancourt D, Cossu G, Buckingham M. Redefining the genetic hierarchies controlling skeletal myogenesis: Pax-3 and Myf-5 act upstream of MyoD. Cell. 1997 Apr;89(1):127-38.
  57. 57. Kang JS, Krauss RS. Muscle stem cells in developmental and regenerative myogenesis. Current Opinion in Clinical Nutrition & Metabolic Care. 2010 May;13(3):243-8.
  58. 58. Wagers AJ, Conboy IM. Cellular and molecular signatures of muscle regeneration: current concepts and controversies in adult myogenesis. Cell. 2005 Sep;122(5):659-67.
  59. 59. Karalaki M, Fili S, Philippou A, Koutsilieris M. Muscle regeneration: cellular and molecular events. In Vivo. 2009 Sep;23(5):779-96.
  60. 60. ChargéSB, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiological Reviews. 2004 Jan;84(1):209-38.
  61. 61. Yablonka-Reuveni Z, Day K, Vine A, Shefer G. Defining the transcriptional signature of skeletal muscle stem cells. Journal of Animal Science. 2008 Apr;86(14 Suppl):E207-16.
  62. 62. McCarthy JJ, Esser KA. Anabolic and catabolic pathways regulating skeletal muscle mass. Current Opinion in Clinical Nutrition & Metabolic Care. 2010 May;13(3):230-5.
  63. 63. Sun H, Zhu T, Ding F, Hu N, Gu X. Proteomic studies of rat tibialis anterior muscle during postnatal growth and development. Molecular and Cellular Biochemistry. 2009 Dec;332(1):161-71.
  64. 64. Xu Y, Qian H, Feng X, Xiong Y, Lei M, Ren Z, Zuo B, Xu D, Ma Y, Yuan H. Differential proteome and transcriptome analysis of porcine skeletal muscle during development. Journal of Proteomics. 2012 Apr;75(7):2093-108.
  65. 65. Tannu NS, Rao VK, Chaudhary RM, Giorgianni F, Saeed AE, Gao Y, Raghow R,. Comparative proteomes of the proliferating C(2)C(12) myoblasts and fully differentiated myotubes reveal the complexity of the skeletal muscle differentiation program. Molecular and Cellular Proteomics. 2004 Nov;3(11):1065-82.
  66. 66. Casadei L, Vallorani L, Gioacchini AM, Guescini M, Burattini S, D’Emilio A, Biagiotti L, Falcieri E, Stocchi V. Proteomics-based investigation in C2C12 myoblast differentiation. European Journal of Histochemistry. 2009 Dec;53(4):261-8.
  67. 67. Kislinger T, Gramolini AO, Pan Y, Rahman K, Mac Lennan DH, Emili A. Proteome dynamics during C2C12 myoblast differentiation. Molecular and Cellular Proteomics. 2005 Jul;4(7):887-901.
  68. 68. Cui Z, Chen X, Lu B, Park SK, Xu T, Xie Z, Xue P, Hou J, Hang H, Yates JR, 3rd, Yang F. Preliminary quantitative profile of differential protein expression between rat L6 myoblasts and myotubes by stable isotope labeling with amino acids in cell culture. Proteomics. 2009 Mar;9(5):1274-92.
  69. 69. Chan XC, McDermott JC, Siu KW. Identification of secreted proteins during skeletal muscle development. Journal of Proteome Research. 2007 Feb;6(2):698-710.
  70. 70. Henningsen J, Rigbolt KT, Blagoev B, Pedersen BK, Kratchmarova I. Dynamics of the skeletal muscle secretome during myoblast differentiation. Molecular and Cellular Proteomics. 2010 Nov;9(11):2482-96.
  71. 71. Chan CY, Masui O, Krakovska O, Belozerov VE, Voisin S, Ghanny S, Chen J, Moyez D, Zhu P, Evans KR, McDermott JC, Siu KW. Identification of differentially regulated secretome components during skeletal myogenesis. Molecular and Cellular Proteomics. 2011 May;10(5):M110.004804.
  72. 72. Le Bihan MC, Barrio-Hernandez I, Mortensen TP, Henningsen J, Jensen SS, Bigot A, Blagoev B, Butler-Browne G, Kratchmarova I. Cellular Proteome Dynamics during Differentiation of Human Primary Myoblasts. Journal of Proteome Research. 2015 Aug;14(8):3348-61.
  73. 73. Pedersen BK. Muscle as a secretory organ. Comprehensive Physiology. 2013 Jul;3(3):1337-62.
  74. 74. Iizuka K, Machida T, Hirafuji M. Skeletal muscle is an endocrine organ. Journal of Pharmacological Science. 2014;125(2):125-31.
  75. 75. Hartwig S, Raschke S, Knebel B, Scheler M, Irmler M, Passlack W, Muller S, Hanisch FG, Franz T, Li X, Dicken HD, Eckardt K, Beckers J, de Angelis MH, Weigert C, Häring HU, Al-Hasani H, Ouwens DM, Eckel J, Kotzka J, Lehr S. Secretome profiling of primary human skeletal muscle cells. Biochimica et Biophysica Acta. 2014 May;1844(5):1011-7.
  76. 76. Le Bihan MC, Bigot A, Jensen SS, Dennis JL, Rogowska-Wrzesinska A, Lainé J, Gache V, Furling D, Jensen ON, Voit T, Mouly V, Coulton GR, Butler-Browne G. In-depth analysis of the secretome identifies three major independent secretory pathways in differentiating human myoblasts. Journal of Proteomics. 2012 Dec;77:344-56.
  77. 77. Højlund K, Yi Z, Hwang H, Bowen B, Lefort N, Flynn CR, Langlais P, Weintraub ST, Mandarino LJ. Characterization of the human skeletal muscle proteome by one-dimensional gel electrophoresis and HPLC-ESI-MS/MS. Molecular and Cellular Proteomics. 2008;7:257-67.
  78. 78. Parker KC, Walsh RJ, Salajegheh M, Amato AA, Krastins B, Sarracino DA, Greenberg SA. Characterization of human skeletal muscle biopsy samples using shotgun proteomics. Journal of Proteome Research. 2009;8:3265-77.
  79. 79. Drexler HC, Ruhs A, Konzer A, Mendler L, Bruckskotten M, Looso M, Günther S, Boettger T, Krüger M, Braun T. On marathons and Sprints: an integrated quantitative proteomics and transcriptomics analysis of differences between slow and fast muscle fibers. Molecular and Cellular Proteomics. 2012;11:M111.010801.
  80. 80. Burniston JG, Connolly J, Kainulainen H, Britton SL, Koch LG. Label-free profiling of skeletal muscle using high-definition mass spectrometry. Proteomics. 2014;14:2339-44.
  81. 81. Murgia M, Nagaraj N, Deshmukh AS, Zeiler M, Cancellara P, Moretti I, Reggiani C, Schiaffino S, Mann M. Single muscle fiber proteomics reveals unexpected mitochondrial specialization. EMBO Reports. 2015;16:387-95.
  82. 82. Deshmukh AS, Murgia M, Nagaraja N, Treebak JT, Cox J, Mann M. Deep proteomics of mouse skeletal muscle enables quantitation of protein isoforms, metabolic pathways and transcription factors. Molecular and Cellular Proteomics. 2015:14:841-53.
  83. 83. Holland A, Dowling P, Meleady P, Henry M, Zweyer M, Mundegar RR, Swandulla D, Ohlendieck K. Label-free mass spectrometric analysis of the mdx-4cv diaphragm identifies the matricellular protein periostin as a potential factor involved in dystrophinopathy-related fibrosis. Proteomics. 2015;15:2318-31.
  84. 84. Murphy S, Henry M, Meleady P, Zweyer M, Mundegar RR, Swandulla D, Ohlendieck K. Simultaneous Pathoproteomic Evaluation of the Dystrophin-Glycoprotein Complex and Secondary Changes in the mdx-4cv Mouse Model of Duchenne Muscular Dystrophy. Biology (Basel) 2015;4:397-423.
  85. 85. Murphy S, Zweyer M, Mundegar RR, Henry M, Meleady P, Swandulla D, Ohlendieck K. Concurrent Label-Free Mass Spectrometric Analysis of Dystrophin Isoform Dp427 and the Myofibrosis Marker Collagen in Crude Extracts from mdx-4cv Skeletal Muscles. Proteomes 2015;3:298-327.
  86. 86. Holland A, Murphy S, Dowling P, Ohlendieck K. Pathoproteomic profiling of the skeletal muscle matrisome in dystrophinopathy associated myofibrosis. Proteomics. 2016 Jan;16(2):345-66.
  87. 87. Smith LR, Meyer G, Lieber RL. Systems analysis of biological networks in skeletal muscle function. Wiley Interdisciplinary Reviews: Systems Biology and Medicine. 2013 Jan-Feb;5(1):55-71.
  88. 88. Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M. STRING v9.1: Protein–protein interaction networks, with increased coverage and integration. Nucleic Acids Research. 2013;41:D808-15.
  89. 89. Booth FW, Thomason DB. Molecular and cellular adaptation of muscle in response to exercise: perspectives of various models. Physiological Reviews. 1991;71(2):541-85.
  90. 90. Wisdom KM, Delp SL, Kuhl E. Use it or lose it: multiscale skeletal muscle adaptation to mechanical stimuli. Biomechanics and Modeling in Mechanobiology. 2015 Apr;14(2):195-215.
  91. 91. Hawley JA. Adaptations of skeletal muscle to prolonged, intense endurance training. Clinical and Experimental Pharmacology and Physiology. 2002 Mar;29(3):218-22.
  92. 92. Ross A, Leveritt M. Long-term metabolic and skeletal muscle adaptations to short-sprint training: implications for sprint training and tapering. Sports Medicine. 2001;31(15):1063-82.
  93. 93. Kjaer M, Magnusson P, Krogsgaard M, Boysen Møller J, Olesen J, Heinemeier K, Hansen M, Haraldsson B, Koskinen S, Esmarck B, Langberg H. Extracellular matrix adaptation of tendon and skeletal muscle to exercise. Journal of Anatomy. 2006 Apr;208(4):445-50.
  94. 94. Coutinho EL, DeLuca C, Salvini TF, Vidal BC. Bouts of passive stretching after immobilization of the rat soleus muscle increase collagen macromolecular organization and muscle fiber area. Connect Tissue Research. 2006;47(5):278-86.
  95. 95. Hyldahl RD, Nelson B, Xin L, Welling T, Groscost L, Hubal MJ, Chipkin S, Clarkson PM, Parcell AC. Extracellular matrix remodeling and its contribution to protective adaptation following lengthening contractions in human muscle. FASEB Journal. 2015 Jul;29(7):2894-904.
  96. 96. Heinemeier KM, Olesen JL, Haddad F, Langberg H, Kjaer M, Baldwin KM, Schjerling P. Expression of collagen and related growth factors in rat tendon and skeletal muscle in response to specific contraction types. Journal of Physiology. 2007 Aug;582(3):1303-16.
  97. 97. Rullman E, Rundqvist H, Wågsäter D, Fischer H, Eriksson P, Sundberg CJ, Jansson E, Gustafsson T. A single bout of exercise activates matrix metalloproteinase in human skeletal muscle. Journal of Applied Physiology. 2007 Jun;102(6):2346-51.
  98. 98. Heinemeier KM, Bjerrum SS, Schjerling P, Kjaer M. Expression of extracellular matrix components and related growth factors in human tendon and muscle after acute exercise. Scandinavian Journal of Medicine & Science in Sports. 2013 Jun;23(3):e150-61.
  99. 99. Ogasawara R, Nakazato K, Sato K, Boppart MD, Fujita S. Resistance exercise increases active MMP and β1-integrin protein expression in skeletal muscle. Physiological Reports. 2014 Nov;2(11):e12212.
  100. 100. Pincu Y, Linden MA, Zou K, Baynard T, Boppart MD. The effects of high fat diet and moderate exercise on TGFβ1 and collagen deposition in mouse skeletal muscle. Cytokine. 2015 May;73(1):23-9.
  101. 101. Kaasik P, Riso EM, Seene T. Extracellular matrix and myofibrils during unloading and reloading of skeletal muscle. International Journal of Sports Medicine. 2011 Apr;32(4):247-53.
  102. 102. Burniston JG, Hoffman EP. Proteomic responses of skeletal and cardiac muscle to exercise. Expert Review of Proteomics. 2011 Jun;8(3):361-77.
  103. 103. Ohlendieck K. Proteomics of exercise-induced skeletal muscle adaptations. OA Sports Medicine. 2013 Mar;1(1):3.
  104. 104. Burniston JG. Changes in the rat skeletal muscle proteome induced by moderate-intensity endurance exercise. Biochimica et Biophysica Acta. 2008 Jul;1784(7):1077-86.
  105. 105. Egan B, Dowling P, O’Connor PL, Henry M, Meleady P, Zierath JR, O’Gorman DJ. 2-D DIGE analysis of the mitochondrial proteome from human skeletal muscle reveals time course-dependent remodelling in response to 14 consecutive days of endurance exercise training. Proteomics. 2011 Apr;11(8):1413-28.
  106. 106. Holloway KV, O’Gorman M, Woods P, Morton JP, Evans L, Cable NT, Goldspink DF, Burniston JG. Proteomic investigation of changes in human vastus lateralis muscle in response to interval-exercise training. Proteomics. 2009 Nov;9(22):5155-74.
  107. 107. Schild M, Ruhs A, Beiter T, Zügel M, Hudemann J, Reimer A, Krumholz-Wagner I, Wagner C, Keller J, Eder K, Krüger K, Krüger M, Braun T, NießA, Steinacker J, Mooren FC. Basal and exercise induced label-free quantitative protein profiling of m. vastus lateralis in trained and untrained individuals. Journal of Proteomics. 2015 Jun;122:119-32.
  108. 108. Ljubicic V, Adhihetty PJ, Hood DA. Application of animal models: chronic electrical stimulation-induced contractile activity. Canadian Journal of Applied Physiology. 2005 Oct;30(5):625-43.
  109. 109. Pette D, Staron RS. Transitions of muscle fiber phenotypic profiles. Histochemistry & Cell Biology. 2001 May;115(5):359-72
  110. 110. Hicks A, Ohlendieck K, Göpel SO, Pette D. Early functional and biochemical adaptations to low-frequency stimulation of rabbit fast-twitch muscle. American Journal of Physiology. 1997 Jul;273(1):C297-305.
  111. 111. Donoghue P, Doran P, Dowling P, Ohlendieck K. Differential expression of the fast skeletal muscle proteome following chronic low-frequency stimulation. Biochimica et Biophysica Acta. 2005 Sep;1752(2):166-76.
  112. 112. Donoghue P, Doran P, Wynne K, Pedersen K, Dunn MJ, Ohlendieck K. Proteomic profiling of chronic low-frequency stimulated fast muscle. Proteomics. 2007 Sep;7(18):3417-30.
  113. 113. Henriksson J, Chi MM, Hintz CS, Young DA, Kaiser KK, Salmons S, Lowry OH. Chronic stimulation of mammalian muscle: changes in enzymes of six metabolic pathways. American Journal of Physiology. 1986 Oct;251(4):C614-32.
  114. 114. Kratz JM, Johnson WS, Mukherjee R, Hu J, Crawford FA, Spinale FG. The relation between latissimus dorsi skeletal muscle structure and contractile function after cardiomyoplasty. Journal of Thoracic and Cardiovascular Surgery. 1994 Mar;107(3):868-78.
  115. 115. Carmignac V, Durbeej M. Cell–matrix interactions in muscle disease. Journal of Pathology. 2012 Jan;226(2):200-18.
  116. 116. Kragstrup TW, Kjaer M, Mackey AL. Structural, biochemical, cellular, and functional changes in skeletal muscle extracellular matrix with aging. Scandinavian Journal of Medicine & Science in Sports. 2011 Dec;21(6):749-57.
  117. 117. Desguerre I, Mayer M, Leturcq F, Barbet JP, Gherardi RK, Christov C. Endomysial fibrosis in Duchenne muscular dystrophy: a marker of poor outcome associated with macrophage alternative activation. Journal of Neuropathology & Experimental Neurology. 2009 Jul;68(7):762-73.
  118. 118. Merlini L, Bernardi P. Therapy of collagen VI-related myopathies (Bethlem and Ullrich). Neurotherapeutics. 2008 Oct;5(4):613-8.
  119. 119. Bönnemann CG. The collagen VI-related myopathies: muscle meets its matrix. Nat Rev Neurol. 2011 Jun 21;7(7):379-90.
  120. 120. Mehuron T, Kumar A, Duarte L, Yamauchi J, Accorsi A, Girgenrath M. Dysregulation of matricellular proteins is an early signature of pathology in laminin-deficient muscular dystrophy. Skeletal Muscle. 2014 Jul;4:14.
  121. 121. Løkken N, Born AP, Duno M, Vissing J. LAMA2-related myopathy: Frequency among congenital and limb-girdle muscular dystrophies. Muscle & Nerve. 2015 Oct;52(4):547-53.
  122. 122. Klingler W, Jurkat-Rott K, Lehmann-Horn F, Schleip R. The role of fibrosis in Duchenne muscular dystrophy. Acta Myologica. 2012 Dec;31(3):184-95.
  123. 123. Kharraz Y, Guerra J, Pessina P, Serrano AL, Muñoz-Cánoves P. Understanding the process of fibrosis in Duchenne muscular dystrophy. Biomedical Research International. 2014;2014:965631.
  124. 124. De Palma S, Leone R, Grumati P, Vasso M, Polishchuk R, Capitanio D, Braghetta P, Bernardi P, Bonaldo P, Gelfi C. Changes in muscle cell metabolism and mechanotransduction are associated with myopathic phenotype in a mouse model of collagen VI deficiency. PLoS One. 2013;8(2):e56716.
  125. 125. De Palma S, Capitanio D, Vasso M, Braghetta P, Scotton C, Bonaldo P, Lochmüller H, Muntoni F, Ferlini A, Gelfi C. Muscle proteomics reveals novel insights into the pathophysiological mechanisms of collagen VI myopathies. Journal of Proteome Research. 2014 Nov;13(11):5022-30.
  126. 126. Holland A, Carberry S, Ohlendieck K. Proteomics of the dystrophin-glycoprotein complex and dystrophinopathy. Current Protein and Peptide Science. 2013 Dec;14(8):680-97.
  127. 127. Dowling P., Holland A., Ohlendieck K., Mass spectrometry-based identification of muscle-associated and muscle-derived proteomic biomarkers of dystrophinopathies. Journal of Neuromuscular Disorders. 2014;1:15-40.
  128. 128. Carberry S, Zweyer M, Swandulla D, Ohlendieck K. Proteomics reveals drastic increase of extracellular matrix proteins collagen and dermatopontin in the aged mdx diaphragm model of Duchenne muscular dystrophy. International Journal of Molecular Medicine. 2012 Aug;30(2):229-34.
  129. 129. Carberry S, Zweyer M, Swandulla D, Ohlendieck K. Application of fluorescence two-dimensional difference in-gel electrophoresis as a proteomic biomarker discovery tool in muscular dystrophy research. Biology (Basel). 2013 Dec;2(4):1438-64.
  130. 130. Forbes EG, Cronshaw AD, MacBeath JR and Hulmes DJ: Tyrosine-rich acidic matrix protein (TRAMP) is a tyrosine-sulphated and widely distributed protein of the extracellular matrix. FEBS Letters. 1994 Sep;351(3):433-6.
  131. 131. Okamoto O and Fujiwara S: Dermatopontin, a novel player in the biology of the extracellular matrix. Connective Tissue Research. 2006;47(4):177-89.
  132. 132. Conway S.J., Izuhara K, Kudo Y, Litvin J, Markwald R, Ouyang G, Arron JR, Holweg CT, Kudo A. The role of periostin in tissue remodeling across health and disease. Cellular and Molecular Life Science. 2014 Apr;71(7):1279-88.
  133. 133. Ozdemir C., Akpulat U., Sharafi P., Yıldız Y. Onbaşılar I, Kocaefe C. Periostin is temporally expressed as an extracellular matrix component in skeletal muscle regeneration and differentiation. Gene. 2014 Dec;553(2):130-9.
  134. 134. Lorts A, Schwanekamp JA, Baudino TA, McNally EM, Molkentin JD. Deletion of periostin reduces muscular dystrophy and fibrosis in mice by modulating the transforming growth factor-β pathway. Proceedings of the National Acadamy of Science USA. 2012 Jul;109(27):10978-83.
  135. 135. Menezes de Oliveira, B., Matsumura C.Y., Fontes-Oliveira C.C., Gawlik K.I. Acosta H, Wernhoff P, Durbeej M. Quantitative proteomic analysis reveals metabolic alterations, calcium dysregulation and increased expression of extracellular matrix proteins in laminin α2 chain-deficient muscle. Molecular and Cellular Proteomics. 2014 Nov;13(11):3001-13.
  136. 136. Pessina P., Cabrera D., Morales M.G., Riquelme C.A., Gutiérrez J, Serrano AL, Brandan E, Muñoz-Cánoves P. Novel and optimized strategies for inducing fibrosis in vivo: focus on Duchenne Muscular Dystrophy. Skeletal Muscle. 2014 Aug;4:7.
  137. 137. Carberry S, Ohlendieck K. Gel electrophoresis-based proteomics of senescent tissues. Methods in Molecular Biology. 2013;1048:229-46.

Written By

Sandra Murphy and Kay Ohlendieck

Submitted: 08 October 2015 Reviewed: 15 January 2016 Published: 15 June 2016