Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 121(22): e2402890121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38771868

ABSTRACT

Maintaining the structure of cardiac membranes and membrane organelles is essential for heart function. A critical cardiac membrane organelle is the transverse tubule system (called the t-tubule system) which is an invagination of the surface membrane. A unique structural characteristic of the cardiac muscle t-tubule system is the extension of the extracellular matrix (ECM) from the surface membrane into the t-tubule lumen. However, the importance of the ECM extending into the cardiac t-tubule lumen is not well understood. Dystroglycan (DG) is an ECM receptor in the surface membrane of many cells, and it is also expressed in t-tubules in cardiac muscle. Extensive posttranslational processing and O-glycosylation are required for DG to bind ECM proteins and the binding is mediated by a glycan structure known as matriglycan. Genetic disruption resulting in defective O-glycosylation of DG results in muscular dystrophy with cardiorespiratory pathophysiology. Here, we show that DG is essential for maintaining cardiac t-tubule structural integrity. Mice with defects in O-glycosylation of DG developed normal t-tubules but were susceptible to stress-induced t-tubule loss or severing that contributed to cardiac dysfunction and disease progression. Finally, we observed similar stress-induced cardiac t-tubule disruption in a cohort of mice that solely lacked matriglycan. Collectively, our data indicate that DG in t-tubules anchors the luminal ECM to the t-tubule membrane via the polysaccharide matriglycan, which is critical to transmitting structural strength of the ECM to the t-tubules and provides resistance to mechanical stress, ultimately preventing disruptions in cardiac t-tubule integrity.


Subject(s)
Dystroglycans , Myocardium , Animals , Mice , Myocardium/metabolism , Myocardium/pathology , Glycosylation , Dystroglycans/metabolism , Extracellular Matrix/metabolism , Mice, Knockout
2.
bioRxiv ; 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38187633

ABSTRACT

Matriglycan (-1,3-ß-glucuronic acid-1,3-α-xylose-) is a polysaccharide that is synthesized on α-dystroglycan, where it functions as a high-affinity glycan receptor for extracellular proteins, such as laminin, perlecan and agrin, thus anchoring the plasma membrane to the extracellular matrix. This biological activity is closely associated with the size of matriglycan. Using high-resolution mass spectrometry and site-specific mutant mice, we show for the first time that matriglycan on the T317/T319 and T379 sites of α-dystroglycan are not identical. T379-linked matriglycan is shorter than the previously characterized T317/T319-linked matriglycan, although it maintains its laminin binding capacity. Transgenic mice with only the shorter T379-linked matriglycan exhibited mild embryonic lethality, but those that survived were healthy. The shorter T379-linked matriglycan exists in multiple tissues and maintains neuromuscular function in adult mice. In addition, the genetic transfer of α-dystroglycan carrying just the short matriglycan restored grip strength and protected skeletal muscle from eccentric contraction-induced damage in muscle-specific dystroglycan knock-out mice. Due to the effects that matriglycan imparts on the extracellular proteome and its ability to modulate cell-matrix interactions, our work suggests that differential regulation of matriglycan length in various tissues optimizes the extracellular environment for unique cell types.

3.
Neuromuscul Disord ; 31(11): 1169-1178, 2021 11.
Article in English | MEDLINE | ID: mdl-34654610

ABSTRACT

Alpha-dystroglycan (αDG) is a highly glycosylated cell surface protein with a significant role in cell-to-extracellular matrix interactions in muscle. αDG interaction with extracellular ligands relies on the activity of the LARGE1 glycosyltransferase that synthesizes and extends the heteropolysaccharide matriglycan. Abnormalities in αDG glycosylation and formation of matriglycan are the pathogenic mechanisms for the dystroglycanopathies, a group of congenital muscular dystrophies. Muscle biopsies were evaluated from related 6-week-old Labrador retriever puppies with poor suckling, small stature compared to normal litter mates, bow-legged stance and markedly elevated creatine kinase activities. A dystrophic phenotype with marked degeneration and regeneration, multifocal mononuclear cell infiltration and endomysial fibrosis was identified on muscle cryosections. Single nucleotide polymorphism (SNP) array genotyping data on the family members identified three regions of homozygosity in 4 cases relative to 8 controls. Analysis of whole genome sequence data from one of the cases identified a stop codon mutation in the LARGE1 gene that truncates 40% of the protein. Immunofluorescent staining and western blotting demonstrated the absence of matriglycan in skeletal muscle and heart from affected dogs. Compared to control, LARGE enzyme activity was not detected. This is the first report of a dystroglycanopathy in dogs.


Subject(s)
Dog Diseases/genetics , Muscular Dystrophy, Animal/genetics , Animals , Dogs , Dystroglycans/metabolism , Glycosylation , Muscle, Skeletal/pathology , Mutation , Phenotype
4.
Elife ; 92020 09 25.
Article in English | MEDLINE | ID: mdl-32975514

ABSTRACT

Matriglycan [-GlcA-ß1,3-Xyl-α1,3-]n serves as a scaffold in many tissues for extracellular matrix proteins containing laminin-G domains including laminin, agrin, and perlecan. Like-acetyl-glucosaminyltransferase 1 (LARGE1) synthesizes and extends matriglycan on α-dystroglycan (α-DG) during skeletal muscle differentiation and regeneration; however, the mechanisms which regulate matriglycan elongation are unknown. Here, we show that Protein O-Mannose Kinase (POMK), which phosphorylates mannose of core M3 (GalNAc-ß1,3-GlcNAc-ß1,4-Man) preceding matriglycan synthesis, is required for LARGE1-mediated generation of full-length matriglycan on α-DG (~150 kDa). In the absence of Pomk gene expression in mouse skeletal muscle, LARGE1 synthesizes a very short matriglycan resulting in a ~ 90 kDa α-DG which binds laminin but cannot prevent eccentric contraction-induced force loss or muscle pathology. Solution NMR spectroscopy studies demonstrate that LARGE1 directly interacts with core M3 and binds preferentially to the phosphorylated form. Collectively, our study demonstrates that phosphorylation of core M3 by POMK enables LARGE1 to elongate matriglycan on α-DG, thereby preventing muscular dystrophy.


Subject(s)
Dystroglycans/metabolism , Gene Expression , Muscle, Skeletal/physiology , N-Acetylglucosaminyltransferases/genetics , Protein Kinases/genetics , Animals , Male , Mannose/chemistry , Mice , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation , Protein Kinases/metabolism
5.
Proc Natl Acad Sci U S A ; 113(39): 10992-7, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27625424

ABSTRACT

Dystroglycan (DG) is a highly expressed extracellular matrix receptor that is linked to the cytoskeleton in skeletal muscle. DG is critical for the function of skeletal muscle, and muscle with primary defects in the expression and/or function of DG throughout development has many pathological features and a severe muscular dystrophy phenotype. In addition, reduction in DG at the sarcolemma is a common feature in muscle biopsies from patients with various types of muscular dystrophy. However, the consequence of disrupting DG in mature muscle is not known. Here, we investigated muscles of transgenic mice several months after genetic knockdown of DG at maturity. In our study, an increase in susceptibility to contraction-induced injury was the first pathological feature observed after the levels of DG at the sarcolemma were reduced. The contraction-induced injury was not accompanied by increased necrosis, excitation-contraction uncoupling, or fragility of the sarcolemma. Rather, disruption of the sarcomeric cytoskeleton was evident as reduced passive tension and decreased titin immunostaining. These results reveal a role for DG in maintaining the stability of the sarcomeric cytoskeleton during contraction and provide mechanistic insight into the cause of the reduction in strength that occurs in muscular dystrophy after lengthening contractions.


Subject(s)
Cytoskeleton/metabolism , Dystroglycans/metabolism , Muscle Contraction , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Sarcomeres/metabolism , Animals , Connectin/metabolism , Cytoskeleton/drug effects , Excitation Contraction Coupling/drug effects , Female , Isometric Contraction/drug effects , Male , Mice, Knockout , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Necrosis , Organ Size , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sarcolemma/metabolism , Sarcomeres/drug effects , Tamoxifen/pharmacology
6.
Science ; 302(5649): 1416-8, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-14631046

ABSTRACT

Calcium ion (Ca2+) influx through voltage-gated Ca2+ channels is important for the regulation of vascular tone. Activation of L-type Ca2+ channels initiates muscle contraction; however, the role of T-type Ca2+ channels (T-channels) is not clear. We show that mice deficient in the alpha1H T-type Ca2+ channel (alpha(1)3.2-null) have constitutively constricted coronary arterioles and focal myocardial fibrosis. Coronary arteries isolated from alpha(1)3.2-null arteries showed normal contractile responses, but reduced relaxation in response to acetylcholine and nitroprusside. Furthermore, acute blockade of T-channels with Ni2+ prevented relaxation of wild-type coronary arteries. Thus, Ca2+ influx through alpha1H T-type Ca2+ channels is essential for normal relaxation of coronary arteries.


Subject(s)
Arteries/physiology , Calcium Channels, T-Type/physiology , Calcium/metabolism , Coronary Vessels/physiology , Vasodilation , Acetylcholine/pharmacology , Animals , Arteries/drug effects , Calcium Channels, T-Type/genetics , Coronary Vessels/drug effects , Coronary Vessels/pathology , Echocardiography , Electrocardiography , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Fibrosis , Ganglia, Spinal/cytology , Gene Targeting , Heart/physiology , Heart Rate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/physiology , Myocardium/pathology , Neurons/metabolism , Nickel/pharmacology , Nitric Oxide/physiology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Patch-Clamp Techniques , Vasoconstriction/drug effects , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...