Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Skelet Muscle ; 7(1): 11, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28587652

ABSTRACT

BACKGROUND: Sarcospan (SSPN) is a transmembrane protein that interacts with the sarcoglycans (SGs) to form a tight subcomplex within the dystrophin-glycoprotein complex that spans the sarcolemma and interacts with laminin in the extracellular matrix. Overexpression of SSPN ameliorates Duchenne muscular dystrophy in murine models. METHODS: Standard cloning approaches were used to identify nanospan, and nanospan-specific polyclonal antibodies were generated and validated. Biochemical isolation of skeletal muscle membranes and two-photon laser scanning microscopy were used to analyze nanospan localization in muscle from multiple murine models. Duchenne muscular dystrophy biopsies were analyzed by immunoblot analysis of protein lysates as well as indirect immunofluorescence analysis of muscle cryosections. RESULTS: Nanospan is an alternatively spliced isoform of sarcospan. While SSPN has four transmembrane domains and is a core component of the sarcolemmal dystrophin-glycoprotein complex, nanospan is a type II transmembrane protein that does not associate with the dystrophin-glycoprotein complex. We demonstrate that nanospan is enriched in the sarcoplasmic reticulum (SR) fractions and is not present in the T-tubules. SR fractions contain membranes from three distinct structural regions: a region flanking the T-tubules (triadic SR), a SR region across the Z-line (ZSR), and a longitudinal SR region across the M-line (LSR). Analysis of isolated murine muscles reveals that nanospan is mostly associated with the ZSR and triadic SR, and only minimally with the LSR. Furthermore, nanospan is absent from the SR of δ-SG-null (Sgcd-/-) skeletal muscle, a murine model for limb girdle muscular dystrophy 2F. Analysis of skeletal muscle biopsies from Duchenne muscular dystrophy patients reveals that nanospan is preferentially expressed in type I (slow) fibers in both control and Duchenne samples. Furthermore, nanospan is significantly reduced in Duchenne biopsies. CONCLUSIONS: Alternative splicing of proteins from the SG-SSPN complex produces δ-SG3, microspan, and nanospan that localize to the ZSR and the triadic SR, where they may play a role in regulating resting calcium levels as supported by previous studies (Estrada et al., Biochem Biophys Res Commun 340:865-71, 2006). Thus, alternative splicing of SSPN mRNA generates three protein isoforms (SSPN, microspan, and nanospan) that differ in the number of transmembrane domains affecting subcellular membrane association into distinct protein complexes.


Subject(s)
Alternative Splicing , Carrier Proteins/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Sarcoglycanopathies/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Carrier Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Sarcoglycanopathies/genetics , Sarcoglycanopathies/pathology , Sarcoglycans/genetics , Sarcoplasmic Reticulum/ultrastructure
2.
J Vis Exp ; (130)2017 12 29.
Article in English | MEDLINE | ID: mdl-29364281

ABSTRACT

Measuring functional outcomes in the treatment of muscular dystrophy is an essential aspect of preclinical testing. The assessment of voluntary ambulation in mouse models is a non-invasive and reproducible activity assay that is directly analogous to measures of patient ambulation such as the 6-minute walk test and related mobility scores. Many common methods for testing mouse ambulation speed and distance are based on the open field test, where an animal's free movement within an arena is measured over time. One major downside to this approach is that commercial software and equipment for high-resolution motion tracking is expensive and may require transferring mice to specialized facilities for testing. Here, we describe a low-cost, video-based system for measuring mouse ambulation that utilizes free and open-source software. Using this protocol, we demonstrate that voluntary ambulation in the dystrophin-null mdx mouse model for Duchenne muscular dystrophy (DMD) is decreased relative to wild-type mouse activity. In mdx mice expressing the utrophin transgene, these activity deficits are not observed and the total distance traveled is indistinguishable from wild-type mice. This method is effective for measuring changes in voluntary ambulation associated with dystrophic pathology, and provides a versatile platform that can be readily adapted to diverse research settings.


Subject(s)
Muscular Dystrophy, Animal/physiopathology , Utrophin/biosynthesis , Animals , Disease Models, Animal , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Transgenes , Utrophin/genetics , Video Recording
3.
Hum Mol Genet ; 25(24): 5395-5406, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27798107

ABSTRACT

Duchenne muscular dystrophy (DMD) is a genetic disorder that causes progressive muscle weakness, ultimately leading to early mortality in affected teenagers and young adults. Previous work from our lab has shown that a small transmembrane protein called sarcospan (SSPN) can enhance the recruitment of adhesion complex proteins to the cell surface. When human SSPN is expressed at three-fold levels in mdx mice, this increase in adhesion complex abundance improves muscle membrane stability, preventing many of the histopathological changes associated with DMD. However, expressing higher levels of human SSPN (ten-fold transgenic expression) causes a severe degenerative muscle phenotype in wild-type mice. Since SSPN-mediated stabilization of the sarcolemma represents a promising therapeutic strategy in DMD, it is important to determine whether SSPN can be introduced at high levels without toxicity. Here, we show that mouse SSPN (mSSPN) can be overexpressed at 30-fold levels in wild-type mice with no deleterious effects. In mdx mice, mSSPN overexpression improves dystrophic pathology and sarcolemmal stability. We show that these mice exhibit increased resistance to eccentric contraction-induced damage and reduced fatigue following exercise. mSSPN overexpression improved pulmonary function and reduced dystrophic histopathology in the diaphragm. Together, these results demonstrate that SSPN overexpression is well tolerated in mdx mice and improves sarcolemma defects that underlie skeletal muscle and pulmonary dysfunction in DMD.


Subject(s)
Carrier Proteins/genetics , Membrane Proteins/genetics , Muscular Dystrophy, Duchenne/genetics , Neoplasm Proteins/genetics , Sarcolemma/genetics , Animals , Carrier Proteins/biosynthesis , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Lung Diseases/genetics , Lung Diseases/pathology , Membrane Proteins/biosynthesis , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle Contraction/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Neoplasm Proteins/biosynthesis , Sarcolemma/pathology
4.
Glycobiology ; 26(10): 1120-1132, 2016 10.
Article in English | MEDLINE | ID: mdl-27236198

ABSTRACT

The neuromuscular junction (NMJ) is enriched with glycoproteins modified with N-acetylgalactosamine (GalNAc) residues, and four nominally GalNAc-specific plant lectins have historically been used to identify the NMJ and the utrophin-glycoprotein complex. However, little is known about the specific glycan epitopes on skeletal muscle that are bound by these lectins, the glycoproteins that bear these epitopes or how creation of these glycan epitopes is regulated. Here, we profile changes in cell surface glycosylation during muscle cell differentiation and identify distinct differences in the binding preferences of GalNAc-specific lectins, Wisteria floribunda agglutinin (WFA), Vicia villosa agglutinin (VVA), soybean agglutinin (SBA) and Dolichos biflorus agglutinin (DBA). While we find that all four GalNAc binding lectins specifically label the NMJ, each of the four lectins binds distinct sets of muscle glycoproteins; furthermore, none of the major adhesion complexes are required for binding of any of the four GalNAc-specific lectins. Analysis of glycosylation-related transcripts identified target glycosyltransferases and glycosidases that could potentially create GalNAc-containing epitopes; reducing expression of these transcripts by siRNA highlighted differences in lectin binding specificities. In addition, we found that complex N-glycans are required for binding of WFA and SBA to murine C2C12 myotubes and for WFA binding to wild-type skeletal muscle, but not for binding of VVA or DBA. These results demonstrate that muscle cell surface glycosylation is finely regulated during muscle differentiation in a domain- and acceptor-substrate-specific manner, suggesting that temporal- and site-specific glycosylation are important for skeletal muscle cell function.


Subject(s)
Epitopes/immunology , Glycocalyx/metabolism , Muscle, Skeletal/metabolism , Polysaccharides/immunology , Animals , Cell Differentiation , Cell Line , Chickens , Glycocalyx/chemistry , Glycocalyx/immunology , Mice , Mice, Knockout , Muscle, Skeletal/chemistry , Muscle, Skeletal/cytology , Muscle, Skeletal/immunology
5.
J Am Heart Assoc ; 4(12)2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26702077

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy is a fatal cardiac and skeletal muscle disease resulting from mutations in the dystrophin gene. We have previously demonstrated that a dystrophin-associated protein, sarcospan (SSPN), ameliorated Duchenne muscular dystrophy skeletal muscle degeneration by activating compensatory pathways that regulate muscle cell adhesion (laminin-binding) to the extracellular matrix. Conversely, loss of SSPN destabilized skeletal muscle adhesion, hampered muscle regeneration, and reduced force properties. Given the importance of SSPN to skeletal muscle, we investigated the consequences of SSPN ablation in cardiac muscle and determined whether overexpression of SSPN into mdx mice ameliorates cardiac disease symptoms associated with Duchenne muscular dystrophy cardiomyopathy. METHODS AND RESULTS: SSPN-null mice exhibited cardiac enlargement, exacerbated cardiomyocyte hypertrophy, and increased fibrosis in response to ß-adrenergic challenge (isoproterenol; 0.8 mg/day per 2 weeks). Biochemical analysis of SSPN-null cardiac muscle revealed reduced sarcolemma localization of many proteins with a known role in cardiomyopathy pathogenesis: dystrophin, the sarcoglycans (α-, δ-, and γ-subunits), and ß1D integrin. Transgenic overexpression of SSPN in Duchenne muscular dystrophy mice (mdx(TG)) improved cardiomyofiber cell adhesion, sarcolemma integrity, cardiac functional parameters, as well as increased expression of compensatory transmembrane proteins that mediate attachment to the extracellular matrix. CONCLUSIONS: SSPN regulates sarcolemmal expression of laminin-binding complexes that are critical to cardiac muscle function and protects against transient and chronic injury, including inherited cardiomyopathy.


Subject(s)
Cardiomyopathies/etiology , Carrier Proteins/physiology , Heart/drug effects , Isoproterenol/pharmacology , Membrane Proteins/physiology , Muscular Dystrophy, Duchenne/complications , Neoplasm Proteins/physiology , Animals , Cardiomyopathies/pathology , Creatine Kinase, MB Form/blood , Echocardiography , Fluorescent Antibody Technique , Heart/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscular Dystrophy, Duchenne/pathology , Myocardium/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sarcolemma/physiology
6.
Hum Mol Genet ; 24(7): 2011-22, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25504048

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene that result in loss of the dystrophin-glycoprotein complex, a laminin receptor that connects the myofiber to its surrounding extracellular matrix. Utrophin, a dystrophin ortholog that is normally localized to the neuromuscular junction, is naturally upregulated in DMD muscle, which partially compensates for the loss of dystrophin. Transgenic overexpression of utrophin causes broad sarcolemma localization of utrophin, restoration of laminin binding and amelioration of disease in the mdx mouse model of DMD. We previously demonstrated that overexpression of sarcospan, a dystrophin- and utrophin-binding protein, ameliorates mdx muscular dystrophy. Sarcospan boosts levels of utrophin to therapeutic levels at the sarcolemma, where attachment to laminin is restored. However, understanding the compensatory mechanism is complicated by concomitant upregulation of α7ß1 integrin, which also binds laminin. Similar to the effects of utrophin, transgenic overexpression of α7 integrin prevents DMD disease in mice and is accompanied by increased abundance of utrophin around the extra-synaptic sarcolemma. In order to investigate the mechanisms underlying sarcospan 'rescue' of muscular dystrophy, we created double-knockout mice to test the contributions of utrophin or α7 integrin. We show that sarcospan-mediated amelioration of muscular dystrophy in DMD mice is dependent on the presence of both utrophin and α7ß1 integrin, even when they are individually expressed at therapeutic levels. Furthermore, we found that association of sarcospan into laminin-binding complexes is dependent on utrophin and α7ß1 integrin.


Subject(s)
Antigens, CD/metabolism , Carrier Proteins/metabolism , Integrin alpha Chains/metabolism , Laminin/metabolism , Membrane Proteins/metabolism , Muscular Dystrophy, Duchenne/metabolism , Neoplasm Proteins/metabolism , Utrophin/metabolism , Animals , Antigens, CD/genetics , Carrier Proteins/genetics , Female , Humans , Integrin alpha Chains/genetics , Integrins/genetics , Integrins/metabolism , Laminin/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Neoplasm Proteins/genetics , Protein Binding , Utrophin/genetics
7.
FEBS J ; 280(17): 4210-29, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23601082

ABSTRACT

Three adhesion complexes span the sarcolemma and facilitate critical connections between the extracellular matrix and the actin cytoskeleton: the dystrophin- and utrophin-glycoprotein complexes and α7ß1 integrin. Loss of individual protein components results in a loss of the entire protein complex and muscular dystrophy. Muscular dystrophy is a progressive, lethal wasting disease characterized by repetitive cycles of myofiber degeneration and regeneration. Protein-replacement therapy offers a promising approach for the treatment of muscular dystrophy. Recently, we demonstrated that sarcospan facilitates protein-protein interactions amongst the adhesion complexes and is an important potential therapeutic target. Here, we review current protein-replacement strategies, discuss the potential benefits of sarcospan expression, and identify important experiments that must be addressed for sarcospan to move to the clinic.


Subject(s)
Carrier Proteins/metabolism , Dystrophin/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Muscular Dystrophies/therapy , Neoplasm Proteins/metabolism , Sarcolemma/metabolism , Utrophin/metabolism , Animals , Humans , Muscular Dystrophies/metabolism , Signal Transduction
8.
Skelet Muscle ; 3(1): 1, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23282144

ABSTRACT

Purification of the proteins associated with dystrophin, the gene product responsible for Duchenne muscular dystrophy, led to the discovery of the dystrophin-glycoprotein complex. Sarcospan, a 25-kDa transmembrane protein, was the last component to be identified and its function in skeletal muscle has been elusive. This review will focus on progress over the last decade revealing that sarcospan is an important regulator of muscle cell adhesion, strength, and regeneration. Investigations using several transgenic mouse models demonstrate that overexpression of sarcospan in the mouse model for Duchenne muscular dystrophy ameliorates pathology and restores muscle cell binding to laminin. Sarcospan improves cell surface expression of the dystrophin- and utrophin-glycoprotein complexes as well as α7ß1 integrin, which are the three major laminin-binding complexes in muscle. Utrophin and α7ß1 integrin compensate for the loss of dystrophin and the finding that sarcospan increases their abundance at the extra-synaptic sarcolemma supports the use of sarcospan as a therapeutic target. Newly discovered phenotypes in sarcospan-deficient mice, including a reduction in specific force output and increased drop in force in the diaphragm muscle, result from decreased utrophin and dystrophin expression and further reveal sarcospan's role in determining abundance of these complexes. Dystrophin protein levels and the specific force output of the diaphragm muscle are further reduced upon genetic removal of α7 integrin (Itga7) in SSPN-deficient mice, demonstrating that interactions between integrin and sarcospan are critical for maintenance of the dystrophin-glycoprotein complex and force production of the diaphragm muscle. Sarcospan is a major regulator of Akt signaling pathways and sarcospan-deficiency significantly impairs muscle regeneration, a process that is dependent on Akt activation. Intriguingly, sarcospan regulates glycosylation of a specific subpopulation of α-dystroglycan, the laminin-binding receptor associated with dystrophin and utrophin, localized to the neuromuscular junction. Understanding the basic mechanisms responsible for assembly and trafficking of the dystrophin- and utrophin-glycoprotein complexes to the cell surface is lacking and recent studies suggest that sarcospan plays a role in these essential processes.

9.
Neuromuscul Disord ; 23(2): 188-91, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22800409

ABSTRACT

The Myomatrix 2012 conference held April 22-24th, 2012 at the University of Nevada, Reno convened 73 international participants to discuss the dynamic relationship between muscle and its matrix in muscular dystrophy with a specific focus on congenital muscular dystrophy. Seven sessions over 2½ days defined three central themes: (1) the role of extracellular matrix proteins and compartments in development and specifically in congenital muscular dystrophy (CMD) (2) the role of extracellular matrix signaling and adhesion to membrane receptors and (3) the balance and interplay between inflammation and fibrosis as drivers of altered matrix stiffness, impaired regeneration and progressive dystrophy. This report highlights major conference findings and the translational roadmap as defined by conference attendees.


Subject(s)
Extracellular Matrix/physiology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/physiopathology , Fibrosis , Humans , Muscle, Skeletal/pathology , Sarcolemma/physiology , Signal Transduction/physiology
10.
Hum Mol Genet ; 21(20): 4378-93, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22798625

ABSTRACT

Sarcospan (SSPN) is a core component of the major adhesion complexes in skeletal muscle, the dystrophin- and utrophin (Utr)-glycoprotein complexes (DGC and UGC). We performed a rigorous analysis of SSPN-null mice and discovered that loss of SSPN decreased DGC and UGC abundance, leading to impaired laminin-binding activity and susceptibility to eccentric contraction-induced injury in skeletal muscle. We show that loss of SSPN increased levels of α7ß1 integrin. To genetically test whether integrin compensates for the loss of DGC and UGC function in SSPN-nulls, we generated mice lacking both SSPN and α7 integrin (DKO, double knockout). Muscle regeneration, sarcolemma integrity and fibrosis were exacerbated in DKO mice and were remarkably similar to muscle from Duchenne muscular dystrophy (DMD) patients, suggesting that secondary loss of integrin contributes significantly to pathogenesis. Expression of the DGC and UGC, laminin binding and Akt signaling were negatively impacted in DKO muscle, resulting in severely diminished specific force properties. We demonstrate that SSPN is a necessary component of dystrophin and Utr function and that SSPN modulation of integrin signaling is required for extracellular matrix attachment and muscle force development.


Subject(s)
Antigens, CD/genetics , Carrier Proteins/genetics , Dystrophin/genetics , Integrin alpha Chains/genetics , Membrane Proteins/genetics , Muscle, Skeletal/metabolism , Neoplasm Proteins/genetics , Phenotype , Utrophin/genetics , Animals , Antigens, CD/metabolism , Carrier Proteins/metabolism , Disease Models, Animal , Female , Integrin alpha Chains/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Neoplasm Proteins/metabolism , Sarcolemma/metabolism
11.
J Cell Biol ; 197(7): 1009-27, 2012 Jun 25.
Article in English | MEDLINE | ID: mdl-22734004

ABSTRACT

Utrophin is normally confined to the neuromuscular junction (NMJ) in adult muscle and partially compensates for the loss of dystrophin in mdx mice. We show that Akt signaling and utrophin levels were diminished in sarcospan (SSPN)-deficient muscle. By creating several transgenic and knockout mice, we demonstrate that SSPN regulates Akt signaling to control utrophin expression. SSPN determined α-dystroglycan (α-DG) glycosylation by affecting levels of the NMJ-specific glycosyltransferase Galgt2. After cardiotoxin (CTX) injury, regenerating myofibers express utrophin and Galgt2-modified α-DG around the sarcolemma. SSPN-null mice displayed delayed differentiation after CTX injury caused by loss of utrophin and Akt signaling. Treatment of SSPN-null mice with viral Akt increased utrophin and restored muscle repair after injury, revealing an important role for the SSPN-Akt-utrophin signaling axis in regeneration. SSPN improved cell surface expression of utrophin by increasing transportation of utrophin and DG from endoplasmic reticulum/Golgi membranes. Our experiments reveal functions of utrophin in regeneration and new pathways that regulate utrophin expression at the cell surface.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/physiology , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Regeneration , Utrophin/metabolism , Animals , Carrier Proteins/genetics , Cell Adhesion , Disease Models, Animal , Dystroglycans/metabolism , Enzyme Activation , Glycosylation , Humans , Membrane Proteins/genetics , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Neoplasm Proteins/genetics , Signal Transduction
12.
J Biol Chem ; 287(27): 22759-70, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22570487

ABSTRACT

Duchenne muscular dystrophy is an X-linked disorder characterized by loss of dystrophin, a cytoskeletal protein that connects the actin cytoskeleton in skeletal muscle cells to extracellular matrix. Dystrophin binds to the cytoplasmic domain of the transmembrane glycoprotein ß-dystroglycan (ß-DG), which associates with cell surface α-dystroglycan (α-DG) that binds laminin in the extracellular matrix. ß-DG can also associate with utrophin, and this differential association correlates with specific glycosylation changes on α-DG. Genetic modification of α-DG glycosylation can promote utrophin binding and rescue dystrophic phenotypes in mouse dystrophy models. We used high throughput screening with the plant lectin Wisteria floribunda agglutinin (WFA) to identify compounds that altered muscle cell surface glycosylation, with the goal of finding compounds that increase abundance of α-DG and associated sarcolemmal glycoproteins, increase utrophin usage, and increase laminin binding. We identified one compound, lobeline, from the Prestwick library of Food and Drug Administration-approved compounds that fulfilled these criteria, increasing WFA binding to C2C12 cells and to primary muscle cells from wild type and mdx mice. WFA binding and enhancement by lobeline required complex N-glycans but not O-mannose glycans that bind laminin. However, inhibiting complex N-glycan processing reduced laminin binding to muscle cell glycoproteins, although O-mannosylation was intact. Glycan analysis demonstrated a general increase in N-glycans on lobeline-treated cells rather than specific alterations in cell surface glycosylation, consistent with increased abundance of multiple sarcolemmal glycoproteins. This demonstrates the feasibility of high throughput screening with plant lectins to identify compounds that alter muscle cell glycosylation and identifies a novel role for N-glycans in regulating muscle cell function.


Subject(s)
Glycomics/methods , Laminin/metabolism , Myoblasts, Skeletal/metabolism , Polysaccharides/metabolism , Quadriceps Muscle/metabolism , Sarcolemma/metabolism , Animals , Cells, Cultured , Glycomics/instrumentation , Glycosylation , Lobeline/pharmacology , Male , Mannosyltransferases/genetics , Mannosyltransferases/metabolism , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Nicotinic Agonists/pharmacology , Plant Lectins/pharmacology , Quadriceps Muscle/cytology , RNA, Small Interfering/genetics , Receptors, N-Acetylglucosamine , Sarcolemma/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...