U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Madame Curie Bioscience Database [Internet]. Austin (TX): Landes Bioscience; 2000-2013.

Cover of Madame Curie Bioscience Database

Madame Curie Bioscience Database [Internet].

Show details

Structure and Function of the Dystrophin-Glycoprotein Complex

.

Introduction

Duchenne muscular dystrophy (DMD) is the most prevalent and severe form of human muscular dystrophy. While clinical descriptions of DMD date back to the 1850's, over 100 years passed before evidence suggested that the muscle cell plasma membrane, or sarcolemma, is compromised in DMD muscle. The molecular basis for DMD and its associated sarcolemmal instability became more clear with landmark studies published in the mid-to-late 1980's which identified the gene defective in DMD.1 The DMD locus spans over 2.5 million bases distinguishing it as the largest gene in the human genome. The array of transcripts expressed from the DMD gene is complex due to the presence of multiple promoters and alternative splicing. The largest transcripts encode a four-domain protein with a predicted molecular weight of 427,000, named dystrophin. Dystrophin is the predominant DMD transcript expressed in striated muscle and DMD gene mutations, deletions or duplications most frequently result in a loss of dystrophin expression in muscle of patients afflicted with DMD. Based on its localization to the cytoplasmic face of the sarcolemma and sequence similarity with domains/motifs common to proteins of the actin-based cytoskeleton, dystrophin was hypothesized early on to play a structural role in anchoring the sarcolemma to the underlying cytoskeleton and protect the sarcolemma against stress imposed during muscle contraction or stretch. Biochemical studies aimed at confirming the hypothesized structure and function of dystrophin revealed its tight association with a multi-subunit complex, the so-named dystrophin-glycoprotein complex. Since its description, the dystrophin-glycoprotein complex has emerged as an important structural unit of muscle and also as a critical nexus for understanding muscular dystrophies arising from defects in several distinct genes.

Initial Isolation and Characterization of the Dystrophin-Glycoprotein Complex

Following close behind the identification of dystrophin as the protein missing from DMD muscle, the laboratory of Kevin Campbell demonstrated that dystrophin could be solubilized from the membrane vesicle fraction of skeletal muscle homogenates using the detergent digitonin and dramatically enriched by wheat germ agglutinin chromatography.2 Given that the primary sequence contained no hydrophobic stretches to directly anchor dystrophin within the sarcolemma, it seemed most likely that dystrophin indirectly bound to wheat germ agglutinin through association with a membrane glycoprotein embedded in the sarcolemma and this idea was confirmed by experiments showing that dystrophin binding to wheat germ agglutinin was disrupted by chaotropic agents.2 While anion-exchange chromatography further amplified and purified dystrophin over wheat germ agglutinin chromatography alone, numerous proteins remained in the peak dystrophin fractions and lectin blotting minimally revealed 12 copurifying glycoproteins as potential molecular partners for dystrophin.2 Sucrose gradient centrifugation further resolved potential candidates to 9 proteins stained by Coomassie blue that were shown to strictly cosediment with dystrophin (Fig. 1): a singlet of 88,000, a triplet of 59,000, a singlet of 50,000, a doublet of 43,000, a singlet of 35,000 (but present at a molar ratio of ˜2:1 relative to dystrophin) and a singlet of 25,000 apparent Mr. Lectin blotting identified the 50,000, 43,000 and 35,000 species as glycoproteins and further revealed a broad band with an apparent molecular weight of 156,000.3 While the 156,000 Mr protein was poorly stained by Coomassie blue, its strong staining by wheat germ agglutinin and strict cosedimentation with dystrophin nonetheless elevated its candidacy as a sarcolemmal glycoprotein receptor for dystrophin.3 Thus, the list of potential dystrophin-associated proteins was narrowed down to 10 distinct Mr proteins, 5 of which were glycosylated.

Figure 1. Protein Constituents of the Dystrophin-Glycoprotein Complex.

Figure 1

Protein Constituents of the Dystrophin-Glycoprotein Complex. Shown on the left is a Coomassie blue-stained SDS-polyacrylamide gel loaded with dystrophin-glycoprotein complex purified from rabbit skeletal muscle. Molecular weight standards are indicated (more...)

Importantly, the Campbell lab was simultaneously pursuing a long-term project aimed at generating new monoclonal antibodies to calcium channels expressed in muscle. Since several calcium channel subunits were known to be glycosylated, wheat germ agglutinin-enriched fractions from detergent solubilized muscle membranes were used to immunize mice and screen hybridomas. Screening positive clones against dystrophin-enriched preparations yielded monoclonal antibodies to dystrophin, the 156,000 and 50,000 Mr dystrophin associated glycoproteins. 3 The new antibodies were instrumental in confirming through coimmunoprecipitation experiments the tight association of proteins that cosedimented with dystrophin as well as their colocalization with dystrophin at the sarcolemma.3-5 Moreover, the monoclonal antibodies were critical in demonstrating that the abundance of the 156,000 and 50,000 Mr dystrophin-associated glycoproteins was dramatically reduced in DMD muscle.3,6,7 Since these proteins colocalized with dystrophin in situ, copurified with dystrophin in stoichiometric amounts even after several distinct protein purification methodologies, and were diminished in dystrophin-deficient muscle, it was concluded that dystrophin was part of a large, hetero-oligomeric complex that may serve to stabilize the sarcolemma. Because several constituents were glycosylated and exploitation of this characteristic was so important in their isolation, the assembly of proteins associated with dystrophin was named the dystrophin-glycoprotein complex.

Additional biochemical analyses identified the 156,000 glycoprotein and 59,000 Mr triplet as peripheral membrane proteins5 while the 50,000, 43,000, 35,000 and 25,000 Mr species behaved as a subcomplex of integral membrane proteins.5,8 Based on its extensive glycosylation5,9 and peripheral membrane association, the 156,000 Mr dystrophin-associated glycoprotein was hypothesized to reside on the extracellular face of the sarcolemma and possibly function as a receptor for a component of the extracellular matrix. These hypotheses were born out with the cloning/sequencing of the gene encoding the 156,000 dystrophin-associated glycoprotein, which is expressed from a single transcript along with one of the 43,000 Mr dystrophin-associated glycoproteins.6 The propeptide is proteolytically processed into a wholly extracellular 156,000 subunit and a 43,000 Mr single-pass transmembrane subunit. Based on its extensive glycosylation and association with dystrophin, the 156,000 and 43,000 Mr subunits were renamed α- and β-dystroglycan, respectively. A screen of known extracellular matrix molecules for skeletal muscle α-dystroglycan binding activity identified laminin as the first extracellular ligand for α-dystroglycan.6,9 Laminin-Sepharose pull-down of the entire dystrophin complex definitively demonstrated that α-dystroglycan was a stoichiometric component of the complex.9 These results also led to the hypothesis that the dystrophin-glycoprotein complex may play a role in muscle cell adhesion to the basal lamina.

Working independently, Ozawa and colleagues corroborated10-12 many of the key findings first reported by the Campbell laboratory and they made some important original contributions in elucidating several of the protein-protein interactions within the complex (discussed below). However, Ozawa and colleagues strongly disputed two important conclusions of Campbell's group. First, they initially dismissed α-dystroglycan as an important component of the dystrophin-glycoprotein complex because it could not be stained by Coomassie blue.13 Ozawa and colleagues also strongly contested the initial identification of the 59,000 Mr α-dystrobrevin/syntrophin triplet as cytoplasmic peripheral membrane proteins because their experiments led them to conclude that one of the proteins was a transmembrane glycoprotein. 12 In subsequent work,14 it is clear that Ozawa and colleagues ultimately concurred that α-dystroglycan is an important component of the complex and that syntrophins are nonglycosylated cytoplasmic proteins. Using limited proteolysis, wheat germ agglutinin chromatography and an array of site-specific antibodies, Ozawa and colleagues first demonstrated that the cysteine-rich and first half of the C-terminal domains of dystrophin were important for its binding to the glycoprotein complex.11 By blot overlay assay, they showed that β-dystroglycan, and the 88,000 and 59,000 Mr dystrophin-associated proteins (α-dystrobrevins and syntrophins) directly bound the cysteine-rich and/or C-terminal domains of dystrophin,15 despite failing to reconcile their prior crosslinking studies where they concluded that the 50,000 and 35,000 Mr dystrophin-associated glycoproteins were directly associated with dystrophin and most important in anchoring it to the sarcolemma.10 Ozawa and colleagues also more conclusively showed16 that the dystrophin-glycoprotein complex could be dissociated into 3 sub-complexes consisting of α- and β-dystroglycan (dystroglycan complex), the 50,000, 43,000, and 35,000 Mr dystrophin-associated glycoproteins (sarcoglycan complex), dystrophin plus the 87,000 and 59,000 Mr dystrophin-associated proteins (dystrophin/dystrobrevin/syntrophin complex). However, it bears noting that several studies predating the work of Ozawa and colleagues reported data suggesting resolution of sub-complexes with similar molecular compositions. 5,8,17,18

The largely biochemical studies described above suggested that the dystrophin-glycoprotein complex may function to physically couple the sarcolemmal cytoskeleton with the extracellular matrix (Fig. 2) and that loss of this structural linkage may render the sarcolemma more susceptible to damage when exposed to mechanical stress. The purified dystrophin-glycoprotein complex also provided a substrate for peptide sequencing and antibody production which yielded new probes important in the identification of genes encoding all dystrophin associated teins and elucidation of their respective roles in Duchenne and other forms of muscular dystrophy. Notably, the genes encoding several dystrophin-associated proteins cause forms of muscular dystrophy when mutated in humans or when knocked out in mice. Since dystrophin and its associated proteins are each a story in and of themselves, I will leave their detailed discussions to be elaborated in the specific chapters that follow. Below I will summarize the large body of evidence supporting an important mechanical role for the dystrophin-glycoprotein complex and then discuss its function within the larger protein network of skeletal muscle. Finally, I will propose an engineering design analogy that I believe best fits existing data.

Figure 2. Model of the Dystrophin-Glycoprotein Complex.

Figure 2

Model of the Dystrophin-Glycoprotein Complex. Dystrophin is thought to physically couple the sarcolemma with the costameric cytoskeleton (see Fig. 3) through lateral association of its N-terminal and rod domains with cytoplasmic γ-actin filaments (more...)

In Support of a Mechanical Function for the Dystrophin-Glycoprotein Complex

Within skeletal myofibers, dystrophin is enriched in a discrete, rib-like lattice termed costameres.19,20 Costameres are protein assemblies that circumferentially align in register with the Z disk of peripheral myofibrils and physically couple force-generating sarcomeres with the sarcolemma in striated muscle cells (Fig. 3). A variety of data indicate that costameres are a striated muscle-specific elaboration of the focal adhesions expressed by nonmuscle cells.21 Classical experiments by Street22 and the Sangers23 suggest that costameres function to laterally transmit contractile forces from sarcomeres across the sarcolemma to the extracellular matrix and ultimately to neighboring muscle cells. Lateral transmission of contractile force would be useful for maintaining uniform sarcomere length between adjacent actively contracting and resting muscle cells comprising different motor units within a skeletal muscle. It is also logical that the sites of lateral force transmission across the sarcolemma would be mechanically fortified to minimize stress imposed on the relatively labile lipid bilayer. Other results suggest that costameres may also coordinate an organized folding, or “festooning” of the sarcolemma,22,24 which again may minimize stress experienced by the sarcolemmal bilayer during forceful muscle contraction or stretch. Thus, in support of its hypothesized mechanical function, dystrophin is enriched within a structure (the costamere) that likely transmits mechanical force to and through the sarcolemma.

Figure 3. The costameric cytoskeleton of striated muscle.

Figure 3

The costameric cytoskeleton of striated muscle. Dystrophin is enriched at costameres, protein assemblies that circumferentially align in register with the Z disk of peripheral myofibrils and physically couple force-generating sarcomeres with the sarcolemma. (more...)

Consistent with its enrichment at costameres in normal muscle, the absence of dystrophin in humans and mice leads to a disorganized costameric lattice.19,25-28 Extensive data consistently report that the dystrophin-deficient sarcolemma is exceedingly fragile29-31 resulting in dramatically increased movement of membrane impermeant molecules across the sarcolemma of dystrophin-deficient muscle.30-42 Both necrosis and sarcolemmal permeability of dystrophin-deficient muscle are exacerbated by physical exercise and improved by muscle immobilization. 33,35,39,42-45 Physiological studies have demonstrated that force production by dystrophin-deficient muscle is significantly decreased when normalized against muscle cross-sectional area.40,41,46-59 Interestingly, force output by dystrophin-deficient muscle is hypersensitive to lengthening, or eccentric contraction60,61 and the force decrement exhibited by dystrophin-deficient muscle undergoing eccentric contraction positively correlates with acutely increased sarcolemmal permeability.40,41,53-55,59-63 Immunofluorescence analysis of mechanically peeled sarcolemma has demonstrated that dystrophin at costameres is tightly attached to the sarcolemma20 and its presence is necessary for strong coupling between the sarcolemma and costameric actin filaments comprised of cytoplasmic γ-actin.64 Transgenic overexpression of the dystrophin homologue utrophin, or a dystrophin construct retaining the β-dystroglycan binding site and one actin binding domain is sufficient to restore coupling between the sarcolemma and costameric actin and rescue the sarcolemmal permeability defects accompanying dystrophin deficiency.65,66 Dystrophin is also enriched in costameres of cardiac muscle.67 Like skeletal muscle, dystrophin-deficient cardiac myocytes are abnormally vulnerable to mechanical stress-induced contractile failure and injury.68,69 Finally, knockout of the dystroglycan or sarcoglycan complexes also causes muscular dystrophy that is accompanied by defects in sarcolemmal integrity.70-79 When taken together, the above studies provide compelling evidence that the dystrophin-glycoprotein complex mainly functions to anchor the sarcolemma to costameres and stabilize it against the mechanical forces transduced through costameres during muscle contraction or stretch.

Expanding beyond the Dystrophin-Glycoprotein Complex

Since its initial description in 1990, many additional proteins have been shown to interact with different dystrophin-glycoprotein complex components (Fig. 4). Once the laminin α-chain G-domain was identified as the binding site for α-dystroglycan,80 several other proteins containing homologous G-domain modules were interrogated and shown to bind α-dystroglycan with high affinity. The current list of such proteins includes agrins,81-84 neurexins85 and perlecan.86,87 Like laminins, these proteins all bind to α-dystroglycan in a manner dependent on its oligosaccharide modifications.88 In contrast, the chondroitin sulfate chains of the proteoglycan biglycan have been shown to mediate its binding to the core protein of α-dystroglycan.89 While the functional significance of α-dystroglycan binding to several different extracellular matrix molecules is not fully clear, the results suggest that the dystroglycan complex may serve multiple roles that vary with the extracellular ligand to which it is bound. That agrins, neurexins and perlecan have all been implicated in various aspects of synapse formation or function.90 further suggests participation by the dystroglycan complex in this process.

Figure 4. The Protein Interacting Network of the Dystrophin-Glycoprotein Complex.

Figure 4

The Protein Interacting Network of the Dystrophin-Glycoprotein Complex. AQP4, aquaporin 4; Cav-3, caveolin-3; nNOS, neuronal nitric oxide synthase; SAPK3, stress-activated protein kinase 3 For a full list of supporting references, refer to the supplementary (more...)

Based on its sequence similarity with the calponin homology actin binding domains of β-spectrin and α-actinin, the N-terminal domain of dystrophin was hypothesized to bind actin filaments. Indeed, recombinant proteins encoding the dystrophin N-terminal domain do bind actin filaments,91 but with relatively low affinity.92 In contrast, purified dystrophin-glycoprotein complex bound actin filaments with substantially higher affinity compared to the isolated amino-terminal domain. The stoichiometry of binding however, suggested a more extensive lateral association between dystrophin and actin filaments than could be explained by actin binding solely through the N-terminal domain alone.93 Mapping of the actin binding sites in dystrophin through F-actin cosedimentation of fragments after limited proteolysis led to the identification of a second actin binding site situated in the middle third of the dystrophin rod domain.93 The spectrin-like repeats in this novel actin binding site were found to carry an excess of basic amino acid residues and were shown to bind acidic actin filaments largely through electrostatic interaction.94 Because the middle rod actin binding site of dystrophin was separated from the amino-terminal actin binding domain by ˜1200 amino acids, the two sites were proposed to act in concert to effect an extended lateral association that could account for the measured stoichiometry of binding. In addition, the dystrophin-glycoprotein complex was shown to slow depolymerization of actin filaments in vitro.93,95 These data supported a novel actin filament side-binding model for dystrophin and are entirely consistent with a role for the dystrophin-glycoprotein complex in mechanically coupling the sarcolemma with actin filaments of the costameric cytoskeleton.64

The development of two-hybrid methodologies for the identification of protein-protein interactions has led to the discovery of several proteins that interact with the dystrophin-glycoprotein complex. Two-hybrid screens using α-dystrobrevin as bait identified several novel proteins.96-98 Two of these proteins, synemin/desmuslin98 and syncoilin,99 are structurally related to intermediate filament proteins and interact with the classical intermediate filament protein desmin. Interestingly, mice knocked out for either α-dystrobrevin100 or desmin101,102 exhibit skeletal and cardiomyopathy, which suggests that mechanical coupling of the dystrophin-glycoprotein complex to the intermediate filament cytoskeleton is necessary for normal muscle function (see Chapter 5). Two hybrid screens using the cytoplasmic domains of sarcoglycans identified a skeletal muscle-specific form of filamin (γ-filamin) as a sarcoglycan interacting protein.103 Like dystrophin, filamin contains an N-terminal calponin homology actin binding domain and a large number of repeated motifs, although the repeats in filamin differ in structure from those in dystrophin. Interestingly, filamin A is recruited to focal adhesions of nonmuscle cells in response to local mechanical stress applied via collagen-coated magnetic beads.104 Since γ-filamin is upregulated and recruited to the sarcolemma in dystrophin-deficient muscle,103 it is tempt- ing to speculate that the dystrophin-deficient costamere may “sense” increased mechanical stress and attempt to compensate by recruitment of filamin. In addition to γ-filamin, several more actin binding proteins of costameres are upregulated in dystrophin-deficient muscle including the cytolinker plectin,105 the integrin-associated proteins talin and vinculin106 and the laminin receptor α7β1 integrin.107,108 While not normally present at costameres, the dystrophin homologue utrophin is upregulated and recruited to costameres in dystrophin-deficient muscle.64,65 Based on the protein interaction network illustrated in (Fig. 4), it seems most reasonable that all of these structural proteins are upregulated by the dystrophin-deficient muscle cell in an attempt to compensate for the absence of dystrophin by fortifying the weakened costamere through the recruitment of parallel mechanical linkages. Because dystrophy persists, these parallel linkages are either not completely redundant with the dystrophin-glycoprotein complex, or the compensatory upregulation/recruitment is incomplete. In support of the latter possibility, transgenic overexpression of utrophin37,53,54 or α7 integrin109 has been shown to further compensate for dystrophin deficiency. Thus, many of the proteins found to interact with the dystrophin-glycoprotein complex, or upregulated in its absence, appear to couple the complex with other structural elements of muscle, or form parallel mechanical links between the sarcolemma and myofibrillar apparatus. As such, these findings further reinforce an important mechanical function for the dystrophin-glycoprotein complex.

An Engineering Design Analogy

In many respects, the bulk of experimental data indicate that the dystrophin-glycoprotein complex functions in a manner analogous to the two-by-four (˜2 inch × 4 inch) timbers used to frame the typical American stick house. The architect utilizes two-by-fours as one structural element of a sturdy support matrix (ie., the costamere) intended to securely hold in place weatherproof siding and shingles (ie., the sarcolemma) as well as doors and windows (ie., ion channels and pumps) that allow for controlled movement of occupants, air and light both into and out of the house. When built to the architect's specifications, the house (or normal muscle cell) can withstand the stress imposed on it by extremes in weather such as high winds or heavy snowfall (or muscle contraction). If, however, the house is built during a shortage of two-by-fours (ie., the dystrophin-deficient muscle cell), the carpenter may be forced to substitute two-by-two timbers instead (ie., compensatory upregulation of partially redundant structural proteins). Such an alteration from original design may indeed allow the carpenter to construct a house that stands in calm weather. Conversely, the compromised structure may distort sufficiently under the force of gravity to cause doors and windows to stick or not close tightly. Moreover, the house built with substandard structural elements is certainly less likely to remain intact when more severe weather strikes.

Up to this point, the dystrophin-glycoprotein complex as two-by-four analogy has not taken into account that several interacting proteins suggest additional roles for the dystrophin-glycoprotein complex in organizing molecules involved in cellular signaling (Fig. 4). For example, α-syntrophin anchors neuronal nitric oxide synthase to the sarcolemma,110 which is necessary to properly regulate vascular perfusion in active muscle.111,112 Other data indicate that MAP kinase signaling pathways are perturbed in dystrophin-deficient muscle.113-115 Because the putative role(s) for the dystrophin-glycoprotein complex in cell signaling will no doubt be elaborated in subsequent chapters, the reader may be left wondering whether and how a role in signaling fits with its well-supported mechanical function. However, I believe the dystrophin-glycoprotein complex as two-by-four analogy fits well with its role in anchoring signaling molecules and also can explain the signaling perturbations observed in dystrophin-deficient muscle. While the architect clearly intended the two-by-fours as structural support for the weather-proofing and controlled entry components of the house, this primary function does not prevent the electrician, plumber, telephone and cable television installers from subsequently utilizing the two-by-four framework as a support to route and organize additional regulatory and communication systems (ie., cell signaling pathways) that further enhance functionality of the house. These additional systems would very reasonably be expected to malfunction in a house constructed of substandard structural components (ie., two-by-twos instead of two-by-fours) as a secondary consequence of its distortion under gravity or when challenged by more stringent weather conditions. Alternatively, one could as reasonably argue that mechanical distortion of the structurally weak two-by-two framework house may cause a short in the electrical system (ie., altered cell signaling) that in turn destroys the house by catastrophic fire (ie., apoptosis). In either case, I believe the architect could successfully argue that compromised mechanical integrity precipitated destruction of the house (and that was not his, but the carpenter's fault!). The mere association of signaling molecules with the dystrophin-glycoprotein complex and perturbations of signal transduction pathways in dystrophin-deficient muscle are not sufficient evidence to refute the compelling data supporting a primary mechanical function for the complex. Moreover, such data fail to provide compelling support that the dystrophin-glycoprotein complex actively regulates cellular signaling, or that altered signaling initiates the pathologies observed in dystrophin-deficient muscle. While these hypotheses certainly remain attractive (especially with respect to development of treatments for muscular dystrophy), the current challenge to the field is to design and perform experiments that rigorously test their validity.

In at least one respect, the dystrophin-glycoprotein complex as two-by-four analogy fails. When a house becomes too small for its occupants, the two-by-fours supporting static walls are demolished in order to expand rooms. In the case of muscle however, cells simultaneously grow under the influence of muscle contraction so the “two-by-four” framework of muscle cells must be sufficiently dynamic to expand with growth while simultaneously protecting against stress-induced membrane damage. Several studies have demonstrated that the dystrophin-glycoprotein complex and costameres are indeed dynamic structures capable of remodeling in vivo.21 However, a remaining challenge is to understand how such fascinating and opposing functions are effected at the molecular level, or perhaps at the level of interacting protein networks.

Acknowledgements

I thank Ariana Combs and Inna Rybakova for the blot images used in (Fig. 1) and Kevin Sonnemann for helpful discussions. The author is supported by grants from the Muscular Dystrophy Association and the National Institutes of Health (AR42423).

References

1.
O'Brien KF, Kunkel LM. Dystrophin and muscular dystrophy: Past, present, and future. Mol Genet Metab. 2001;74:75–88. [PubMed: 11592805]
2.
Campbell KP, Kahl SD. Association of dystrophin and an integral membrane glycoprotein. Nature. 1989;338:259–262. [PubMed: 2493582]
3.
Ervasti JM, Ohlendieck K, Kahl SD. et al. Deficiency of a glycoprotein component of the dystrophin complex in dystrophic muscle. Nature. 1990;345:315–319. [PubMed: 2188135]
4.
Ohlendieck K, Ervasti JM, Snook JB. et al. Dystrophin-glycoprotein complex is highly enriched in isolated skeletal muscle sarcolemma. J Cell Biol. 1991;112:135–148. [PMC free article: PMC2288808] [PubMed: 1986002]
5.
Ervasti JM, Campbell KP. Membrane organization of the dystrophin-glycoprotein complex. Cell. 1991;66:1121–1131. [PubMed: 1913804]
6.
Ibraghimov-Beskrovnaya O, Ervasti JM, Leveille CJ. et al. Primary structure of dystrophin-associated glycoproteins linking dystrophin to the extracellular matrix. Nature. 1992;355:696–702. [PubMed: 1741056]
7.
Ohlendieck K, Matsumura K, Ionasescu VV. et al. Duchenne muscular dystrophy: Deficiency of dystrophin-associated proteins in the sarcolemma. Neurology. 1993;43:795–800. [PubMed: 8469343]
8.
Ervasti JM, Kahl SD, Campbell KP. Purification of dystrophin from skeletal muscle. J Biol Chem. 1991;266:9161–9165. [PubMed: 2026615]
9.
Ervasti JM, Campbell KP. A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. J Cell Biol. 1993;122:809–823. [PMC free article: PMC2119587] [PubMed: 8349731]
10.
Yoshida M, Ozawa E. Glycoprotein complex anchoring dystrophin to sarcolemma. J Biochem. 1990;108:748–752. [PubMed: 2081733]
11.
Suzuki A, Yoshida M, Yamamoto H. et al. Glycoprotein-binding site of dystrophin is confined to the cysteine-rich domain and the first half of the carboxy-terminal domain. FEBS Lett. 1992;308:154–160. [PubMed: 1499724]
12.
Yamamoto H, Hagiwara Y, Mizuno Y. et al. Heterogeneity of dystrophin-associated proteins. J Biochem (Tokyo). 1993;114:132–139. [PubMed: 8407865]
13.
Ozawa E, Yoshida M, Hagiwara Y. et al. Formation of dystrophin lining on muscle cell membrane in myogenesis in vivo. In: Ozawa E, Masaki T, Nabeshima Y, eds Frontiers in Muscle ResearchAmsterdam: Elsevier,1991305–319.
14.
Suzuki A, Yoshida M, Ozawa E. Mammalian α1- and β1-syntrophin bind to the alternative splice-prone region of the dystrophin COOH terminus. J Cell Biol. 1995;128:373–381. [PMC free article: PMC2120347] [PubMed: 7844151]
15.
Suzuki A, Yoshida M, Hayashi K. et al. Molecular organization at the glycoprotein-complex-binding site of dystrophin-Three dystrophin-associated proteins bind directly to the carboxy-terminal portion of dystrophin. Eur J Biochem. 1994;220:283–292. [PubMed: 8125086]
16.
Yoshida M, Suzuki A, Yamamoto H. et al. Dissociation of the complex of dystrophin and its associated proteins into several unique groups by n-octyl β-D-glucoside. Eur J Biochem. 1994;222:1055–1061. [PubMed: 8026484]
17.
Butler MH, Douville K, Murname AA. et al. Association of the Mr 58,000 postsynaptic protein of electric tissue with Torpedo dystrophin and the Mr 87,000 postsynaptic protein. J Biol Chem. 1992;267:6213–6218. [PubMed: 1556129]
18.
Kramarcy NR, Vidal A, Froehner SC. et al. Association of utrophin and multiple dystrophin short forms with the mammalian Mr 58,000 dystrophin-associated protein (syntrophin). J Biol Chem. 1994;269:2870–2876. [PubMed: 8300622]
19.
Porter GA, Dmytrenko GM, Winkelmann JC. et al. Dystrophin colocalizes with β-spectrin in distinct subsarcolemmal domains in mammalian skeletal muscle. J Cell Biol. 1992;117:997–1005. [PMC free article: PMC2289490] [PubMed: 1577872]
20.
Straub V, Bittner RE, Léger JJ. et al. Direct visualization of the dystrophin network on skeletal muscle fiber membrane. J Cell Biol. 1992;119:1183–1191. [PMC free article: PMC2289725] [PubMed: 1447296]
21.
Ervasti JM. Costameres: The Achilles' heel of Herculean muscle. J Biol Chem. 2003;278:13591–13594. [PubMed: 12556452]
22.
Street SF. Lateral transmission of tension in frog myofibers: A myofibrillar network and transverse cytoskeletal connections are possible transmitters. J Cell Physiol. 1983;114:346–364. [PubMed: 6601109]
23.
Danowski BA, Imanaka-Yoshida K, Sanger JM. et al. Costameres are sites of force transmission to the substratum in adult rat cardiomyocytes. J Cell Biol. 1992;118:1411–1420. [PMC free article: PMC2289604] [PubMed: 1522115]
24.
Shear CR, Bloch RJ. Vinculin in subsarcolemmal densities in chicken skeletal muscle: Localization and relationship to intracellular and extracellular structures. J Cell Biol. 1985;101:240–256. [PMC free article: PMC2113626] [PubMed: 3924918]
25.
Minetti C, Tanji K, Bonilla E. Immunologic study of vinculin in Duchenne muscular dystrophy. Neurology. 1992;42:1751–1754. [PubMed: 1513465]
26.
Minetti C, Tanji K, Rippa PG. et al. Abnormalities in the expression of β-spectrin in Duchenne muscular dystrophy. Neurology. 1994;44:1149–1153. [PubMed: 8208414]
27.
Ehmer S, Herrmann R, Bittner R. et al. Spatial distribution of β-spectrin in normal and dystrophic human skeletal muscle. Acta Neuropathol (Berl ). 1997;94:240–246. [PubMed: 9292693]
28.
Williams MW, Bloch RJ. Extensive but coordinated reorganization of the membrane skeleton in myofibers of dystrophic (mdx) mice. J Cell Biol. 1999;144:1259–1270. [PMC free article: PMC2150591] [PubMed: 10087268]
29.
Mokri B, Engel AG. Duchenne dystrophy: Electron microscopic findings pointing to a basic or early abnormality in the plasma membrane of the muscle fiber. Neurology. 1975;25:1111–1120. [PubMed: 1105232]
30.
Menke A, Jockusch H. Decreased osmotic stability of dystrophin-less muscle cells from the mdx mouse. Nature. 1991;349:69–71. [PubMed: 1985268]
31.
Menke A, Jockusch H. Extent of shock-induced membrane leakage in human and mouse myotubes depends on dystrophin. J Cell Sci. 1995;108:727–733. [PubMed: 7769014]
32.
Engel AG. Duchenne Dystrophy. In: Engel AG, Banker BQ, eds Myology: Basic and ClinicalNew York: McGraw-Hill,19861185–1240.
33.
Weller B, Karpati G, Carpenter S. Dystrophin-deficient mdx muscle fibers are preferentially vulnerable to necrosis induced by experimental lengthening contractions. J Neurol Sci. 1990;100:9–13. [PubMed: 2089145]
34.
Cox GA, Cole NM, Matsumura K. et al. Overexpression of dystrophin in transgenic mdx mice eliminates dystrophic symptoms without toxicity. Nature. 1993;364:725–729. [PubMed: 8355788]
35.
Clarke MSF, Khakee R, McNeil PL. Loss of cytoplasmic basic fibroblast growth factor from physiologically wounded myofibers of normal and dystrophic muscle. J Cell Sci. 1993;106:121–133. [PubMed: 8270618]
36.
Matsuda R, Nishikawa A, Tanaka H. Visualization of dystrophic muscle fibers in mdx mouse by vital staining with evans blue: Evidence of apoptosis in dystrophin-deficient muscle. J Biochem (Tokyo). 1995;118:959–964. [PubMed: 8749313]
37.
Tinsley JM, Potter AC, Phelps SR. et al. Amelioration of the dystrophic phenotype of mdx mice using a truncated utrophin transgene. Nature. 1996;384:349–353. [PubMed: 8934518]
38.
Straub V, Rafael JA, Chamberlain JS. et al. Animal models for muscular dystrophy show different patterns of sarcolemmal disruption. J Cell Biol. 1997;139:375–385. [PMC free article: PMC2139791] [PubMed: 9334342]
39.
Vilquin JT, Brussee V, Asselin I. et al. Evidence of mdx mouse skeletal muscle fragility in vivo by eccentric running exercise. Muscle Nerve. 1998;21:567–576. [PubMed: 9572235]
40.
Harper SQ, Hauser MA, DelloRusso C. et al. Modular flexibility of dystrophin: Implications for gene therapy of Duchenne muscular dystrophy. Nat Med. 2002;8:253–261. [PubMed: 11875496]
41.
Barton ER, Morris L, Musaro A. et al. Muscle-specific expression of insulin-like growth factor I counters muscle decline in mdx mice. J Cell Biol. 2002;157:137–148. [PMC free article: PMC2173262] [PubMed: 11927606]
42.
Bansal D, Miyake K, Vogel SS. et al. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature. 2003;423:168–172. [PubMed: 12736685]
43.
Karpati G, Carpenter S. Small-caliber skeletal muscle fibers do not suffer deleterious consequences of dystrophic gene expression. Am J Med Genet. 1986;25:653–658. [PubMed: 3789023]
44.
Mizuno Y. Prevention of myonecrosis in mdx mice: Effect of immobilization by the local tetanus method. Brain Dev. 1992;14:319–322. [PubMed: 1456387]
45.
Mokhtarian A, Lefaucheur JP, Even PC. et al. Hindlimb immobilization applied to 21-day-old mdx mice prevents the occurrence of muscle degeneration. J Appl Physiol. 1999;86:924–931. [PubMed: 10066706]
46.
Coulton GR, Curtin NA, Morgan JE. et al. The mdx mouse skeletal muscle myopathy: II. Contractile properties. Neuropathol Appl Neurobiol. 1988;14:299–314. [PubMed: 3221977]
47.
Kometani K, Tsugeno H, Yamada K. Mechanical and energetic properties of dystrophic (mdx) mouse muscle. Jpn J Physiol. 1990;40:541–549. [PubMed: 2150208]
48.
Stedman HH, Sweeney HL, Shrager JB. et al. The mdx mouse diaphragm reproduces the degenerative changes of Duchenne muscular dystrophy. Nature. 1991;352:536–539. [PubMed: 1865908]
49.
Sacco P, Jones DA, Dick JR. et al. Contractile properties and susceptibility to exercise-induced damage of normal and mdx mouse tibialis anterior muscle. Clin Sci (Lond). 1992;82(2):227–236. [PubMed: 1311662]
50.
Quinlan JG, Johnson SR, McKee MK. et al. Twitch and tetanus in mdx mouse muscle. Muscle Nerve. 1992;15:837–842. [PubMed: 1501627]
51.
Cox GA, Cole NM, Matsumura K. et al. Overexpression of dystrophin in transgenic mdx mice eliminates dystrophic symptoms without toxicity. Nature. 1993;364:725–729. [PubMed: 8355788]
52.
Pastoret C, Sebille A. Time course study of the isometric contractile properties of mdx mouse striated muscles. J Muscle Res Cell Motil. 1993;14:423–431. [PubMed: 8227301]
53.
Deconinck N, Tinsley J, De Backer F. et al. Expression of truncated utrophin leads to major functional improvements in dystrophin-deficient muscles of mice. Nature Med. 1997;3:1216–1221. [PubMed: 9359695]
54.
Tinsley J, Deconinck N, Fisher R. et al. Expression of full-length utrophin prevents muscular dystrophy in mdx mice. Nature Med. 1998;4:1441–1444. [PubMed: 9846586]
55.
Deconinck N, Rafael JA, Beckers-Bleukx G. et al. Consequences of the combined deficiency in dystrophin and utrophin on the mechanical properties and myosin composition of some limb and respiratory muscles of the mouse. Neuromuscular Disorders. 1998;8:362–370. [PubMed: 9713852]
56.
Bobet J, Mooney RF, Gordon T. Force and stiffness of old dystrophic (mdx) mouse skeletal muscles. Muscle Nerve. 1998;21:536–539. [PubMed: 9533791]
57.
Stevens ED, Faulkner JA. The capacity of mdx mouse diaphragm muscle to do oscillatory work. J Physiol. 2000;522:457–466. [PMC free article: PMC2269768] [PubMed: 10713969]
58.
Lynch GS, Hinkle RT, Chamberlain JS. et al. Force and power output of fast and slow skeletal muscles from mdx mice 6-28 months old. J Physiol. 2001;535:591–600. [PMC free article: PMC2278782] [PubMed: 11533147]
59.
DelloRusso C, Crawford RW, Chamberlain JS. et al. Tibialis anterior muscles in mdx mice are highly susceptible to contraction-induced injury. J Muscle Res Cell Motil. 2001;22:467–475. [PubMed: 11964072]
60.
Petrof BJ, Shrager JB, Stedman HH. et al. Dystrophin protects the sarcolemma from stresses developed during muscle contraction. Proc Natl Acad Sci USA. 1993;90:3710–3714. [PMC free article: PMC46371] [PubMed: 8475120]
61.
Moens P, Baatsen PH, Marechal G. Increased susceptibility of EDL muscles from mdx mice to damage induced by contractions with stretch. J Muscle Res Cell Motil. 1993;14:446–451. [PubMed: 7693747]
62.
Deconinck N, Ragot T, Maréchal G. et al. Functional protection of dystrophic mouse (mdx ) muscles after adenovirus-mediated transfer of a dystrophin minigene. Proc Natl Acad Sci USA. 1996;93:3570–3574. [PMC free article: PMC39651] [PubMed: 8622977]
63.
Brooks SV. Rapid recovery following contraction-induced injury to in situ skeletal muscles in mdx mice. J Muscle Res Cell Motil. 1998;19:179–187. [PubMed: 9536444]
64.
Rybakova IN, Patel JR, Ervasti JM. The dystrophin complex forms a mechanically strong link between the sarcolemma and costameric actin. J Cell Biol. 2000;150:1209–1214. [PMC free article: PMC2175263] [PubMed: 10974007]
65.
Rybakova IN, Patel JR, Davies KE. et al. Utrophin binds laterally along actin filaments and can couple costameric actin with the sarcolemma when overexpressed in dystrophin-deficient muscle. Mol Biol Cell. 2002;13:1512–1521. [PMC free article: PMC111123] [PubMed: 12006649]
66.
Warner LE, DelloRusso C, Crawford RW. et al. Expression of Dp260 in muscle tethers the actin cytoskeleton to the dystrophin-glycoprotein complex. Hum Mol Genet. 2002;11:1095–1105. [PubMed: 11978768]
67.
Kaprielian RR, Stevenson S, Rothery SM. et al. Distinct patterns of dystrophin organization in myocyte sarcolemma and transverse tubules of normal and diseased human myocardium. Circulation. 2000;101:2586–2594. [PubMed: 10840009]
68.
Danialou G, Comtois AS, Dudley R. et al. Dystrophin-deficient cardiomyocytes are abnormally vulnerable to mechanical stress-induced contractile failure and injury. FASEB J. 2001;15:1655–1658. [PubMed: 11427517]
69.
Kamogawa Y, Biro S, Maeda M. et al. Dystrophin-deficient myocardium is vulnerable to pressure overload in vivo. Cardiovasc Res. 2001;50:509–515. [PubMed: 11376626]
70.
Hack AA, Ly CT, Jiang F. et al. γ-Sarcoglycan deficiency leads to muscle membrane defects and apoptosis independent of dystrophin. J Cell Biol. 1998;142:1279–1287. [PMC free article: PMC2149352] [PubMed: 9732288]
71.
Duclos F, Straub V, Moore SA. et al. Progressive muscular dystrophy in α-sarcoglycan-deficient mice. J Cell Biol. 1998;142:1461–1471. [PMC free article: PMC2141773] [PubMed: 9744877]
72.
Araishi K, Sasaoka T, Imamura M. et al. Loss of the sarcoglycan complex and sarcospan leads to muscular dystrophy in β-sarcoglycan-deficient mice. Hum Mol Genet. 1999;8:1589–1598. [PubMed: 10441321]
73.
Coral-Vazquez R, Cohn RD, Moore SA. et al. Disruption of the sarcoglycan-sarcospan complex in vascular smooth muscle: A novel mechanism for cardiomyopathy and muscular dystrophy. Cell. 1999;98:465–474. [PubMed: 10481911]
74.
Cote PD, Moukhles H, Lindenbaum M. et al. Chimaeric mice deficient in dystroglycans develop muscular dystrophy and have disrupted myoneural synapses. Nat Genet. 1999;23:338–342. [PubMed: 10610181]
75.
Hack AA, Lam MY, Cordier L. et al. Differential requirement for individual sarcoglycans and dystrophin in the assembly and function of the dystrophin-glycoprotein complex. J Cell Sci. 2000;113:2535–2544. [PubMed: 10862711]
76.
Durbeej M, Cohn RD, Hrstka RF. et al. Disruption of the β-sarcoglycan gene reveals pathogenetic complexity of limb-girdle muscular dystrophy type 2E. Mol Cell. 2000;5:141–151. [PubMed: 10678176]
77.
Straub V, Donahue KM, Allamand V. et al. Contrast agent-enhanced magnetic resonance imaging of skeletal muscle damage in animal models of muscular dystrophy. Magn Reson Med. 2000;44:655–659. [PubMed: 11025524]
78.
Cohn R, Henry M, Michele D. et al. Disruption of Dag1 in differentiated skeletal muscle reveals a role for dystroglycan in muscle regeneration. Cell. 2002;110:639–648. [PubMed: 12230980]
79.
Sasaoka T, Imamura M, Araishi K. et al. Pathological analysis of muscle hypertrophy and degeneration in muscular dystrophy in γ-sarcoglycan-deficient mice. Neuromuscul Disord. 2003;13:193–206. [PubMed: 12609501]
80.
Gee SH, Blacher RW, Douville PJ. et al. Laminin-binding protein 120 from brain is closely related to the dystrophin-associated glycoprotein, dystroglycan, and binds with high affinity to the major heparin binding domain of laminin. J Biol Chem. 1993;268:14972–14980. [PubMed: 8325873]
81.
Bowe MA, Deyst KA, Leszyk JD. et al. Identification and purification of an agrin receptor from Torpedo postsynaptic membranes: A heteromeric complex related to the dystroglycans. Neuron. 1994;12:1173–1180. [PubMed: 8185951]
82.
Campanelli JT, Roberds SL, Campbell KP. et al. A role for dystrophin-associated glycoproteins and utrophin in agrin-induced AChR clustering. Cell. 1994;77:663–674. [PubMed: 8205616]
83.
Gee SH, Montanaro F, Lindenbaum MH. et al. Dystroglycan-α, a dystrophin-associated glycoprotein, is a functional agrin receptor. Cell. 1994;77:675–686. [PubMed: 8205617]
84.
Sugiyama J, Bowen DC, Hall ZW. Dystroglycan binds nerve and muscle agrin. Neuron. 1994;13:103–115. [PubMed: 8043271]
85.
Sugita S, Saito F, Tang J. et al. A stoichiometric complex of neurexins and dystroglycan in brain. J Cell Biol. 2001;154:435–445. [PMC free article: PMC2150755] [PubMed: 11470830]
86.
Peng HB, Ali AA, Daggett DF. et al. The relationship between perlecan and dystroglycan and its implication in the formation of the neuromuscular junction. Cell Adhes Commun. 1998;5:475–489. [PubMed: 9791728]
87.
Peng HB, Xie HB, Rossi SG. et al. Acetylcholinesterase clustering at the neuromuscular junction involves perlecan and dystroglycan. J Cell Biol. 1999;145:911–921. [PMC free article: PMC2133180] [PubMed: 10330416]
88.
Michele DE, Campbell KP. Dystrophin-Glycoprotein Complex: Post-translational Processing and Dystroglycan Function. J Biol Chem. 2003;278:15457–15460. [PubMed: 12556455]
89.
Bowe MA, Mendis DB, Fallon JR. The small leucine-rich repeat proteoglycan biglycan binds to α-dystroglycan and is upregulated in dystrophic muscle. J Cell Biol. 2000;148:801–810. [PMC free article: PMC2169361] [PubMed: 10684260]
90.
Montanaro F, Carbonetto S. Targeting dystroglycan in the brain. Neuron. 2003;37:193–196. [PubMed: 12546815]
91.
Hemmings L, Kuhlman PA, Critchley DR. Analysis of the actin-binding domain of α-actinin by mutagenesis and demonstration that dystrophin contains a functionally homologous domain. J Cell Biol. 1992;116:1369–1380. [PMC free article: PMC2289384] [PubMed: 1541634]
92.
Way M, Pope B, Cross RA. et al. Expression of the N-terminal domain of dystrophin in E. coli and demonstration of binding to F-actin. FEBS Lett. 1992;301:243–245. [PubMed: 1577159]
93.
Rybakova IN, Amann KJ, Ervasti JM. A new model for the interaction of dystrophin with F-actin. J Cell Biol. 1996;135:661–672. [PMC free article: PMC2121071] [PubMed: 8909541]
94.
Amann KJ, Renley BA, Ervasti JM. A cluster of basic repeats in the dystrophin rod domain binds F- actin through an electrostatic interaction. J Biol Chem. 1998;273:28419–28423. [PubMed: 9774469]
95.
Rybakova IN, Ervasti JM. Dystrophin-glycoprotein complex is monomeric and stabilizes actin filaments in vitro through a lateral association. J Biol Chem. 1997;272:28771–28778. [PubMed: 9353348]
96.
Newey SE, Howman EV, Ponting CP. et al. Syncoilin, a novel member of the intermediate filament superfamily that interacts with α-dystrobrevin in skeletal muscle. J Biol Chem. 2001;276:6645–6655. [PubMed: 11053421]
97.
Benson MA, Newey SE, Martin-Rendon E. et al. Dysbindin, a novel coiled-coil-containing protein that interacts with the dystrobrevins in muscle and brain. J Biol Chem. 2001;276:24232–24241. [PubMed: 11316798]
98.
Mizuno Y, Thompson TG, Guyon JR. et al. Desmuslin, an intermediate filament protein that interacts with α-dystrobrevin and desmin. Proc Natl Acad Sci USA. 2001;98:6156–6161. [PMC free article: PMC33438] [PubMed: 11353857]
99.
Poon E, Howman EV, Newey SE. et al. Association of syncoilin and desmin: linking intermediate filament proteins to the dystrophin-associated protein complex. J Biol Chem. 2002;277:3433–3439. [PubMed: 11694502]
100.
Grady RM, Grange RW, Lau KS. et al. Role for α-dystrobrevin in the pathogenesis of dystrophin-dependent muscular dystrophies. Nature Cell Biol. 1999;1:215–220. [PubMed: 10559919]
101.
Milner DJ, Weitzer G, Tran D. et al. Disruption of muscle arhitecture and myocardial degeneration in mice lacking desmin. J Cell Biol. 1996;134:1255–1270. [PMC free article: PMC2120972] [PubMed: 8794866]
102.
Li Z, Mericskay M, Agbulut O. et al. Desmin is essential for the tensile strength and integrity of myofibrils but not for myogenic commitment, differentiation, and fusion of skeletal muscle. J Cell Biol. 1997;139:129–144. [PMC free article: PMC2139820] [PubMed: 9314534]
103.
Thompson TG, Chan YM, Hack AA. et al. Filamin 2 (FLN2): A muscle-specific sarcoglycan interacting protein. J Cell Biol. 2000;148:115–126. [PMC free article: PMC3207142] [PubMed: 10629222]
104.
Glogauer M, Arora P, Chou D. et al. The role of actin-binding protein 280 in integrin-dependent mechanoprotection. J Biol Chem. 1998;273:1689–1698. [PubMed: 9430714]
105.
Schroder R, Mundegar RR, Treusch M. et al. Altered distribution of plectin/HD1 in dystrophinopathies. Eur J Cell Biol. 1997;74:165–171. [PubMed: 9352221]
106.
Law DJ, Allen DL, Tidball JG. Talin, vinculin and DRP (utrophin) concentrations are increased at mdx myotendinous junctions following onset of necrosis. J Cell Sci. 1994;107:1477–1483. [PubMed: 7962191]
107.
Vachon PH, Xu H, Liu L. et al. Integrins (α7β1) in muscle function and survival - Disrupted expression in merosin-deficient congenital muscular dystrophy. J Clin Invest. 1997;100:1870–1881. [PMC free article: PMC508374] [PubMed: 9312189]
108.
Hodges BL, Hayashi YK, Nonaka I. et al. Altered expression of the α7β1 integrin in human and murine muscular dystrophies. J Cell Sci. 1997;110:2873–2881. [PubMed: 9427295]
109.
Burkin DJ, Wallace GQ, Nicol KJ. et al. Enhanced expression of the α7β1 integrin reduces muscular dystrophy and restores viability in dystrophic mice. J Cell Biol. 2001;152:1207–1218. [PMC free article: PMC2199213] [PubMed: 11257121]
110.
Adams ME, Mueller HA, Froehner SC. In vivo requirement of the α-syntrophin PDZ domain for the sarcolemmal localization of nNOS and aquaporin-4. J Cell Biol. 2001;155:2–10. [PMC free article: PMC2150783] [PubMed: 11571312]
111.
Thomas GD, Sander M, Lau KS. et al. Impaired metabolic modulation of α-adrenergic vasoconstriction in dystrophin-deficient skeletal muscle. Proc Natl Acad Sci USA. 1998;95:15090–15095. [PMC free article: PMC24580] [PubMed: 9844020]
112.
Sander M, Chavoshan B, Harris SA. et al. Functional muscle ischemia in neuronal nitric oxide synthase-deficient skeletal muscle of children with Duchenne muscular dystrophy. Proc Natl Acad Sci USA. 2000;97:13818–13823. [PMC free article: PMC17659] [PubMed: 11087833]
113.
Kolodziejczyk SM, Walsh GS, Balazsi K. et al. Activation of JNK1 contributes to dystrophic muscle pathogenesis. Curr Biol. 2001;11:1278–1282. [PubMed: 11525743]
114.
Nakamura A, Harrod GV, Davies KE. Activation of calcineurin and stress activated protein kinase/p38-mitogen activated protein kinase in hearts of utrophin-dystrophin knockout mice. Neuromusc Disord. 2001;11:251–259. [PubMed: 11297940]
115.
Nakamura A, Yoshida K, Takeda S. et al. Progression of dystrophic features and activation of mitogen-activated protein kinases and calcineurin by physical exercise, in hearts of mdx mice. FEBS Lett. 2002;520:18–24. [PubMed: 12044863]
Copyright © 2000-2013, Landes Bioscience.
Bookshelf ID: NBK6193

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...