Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 945
Filter
1.
J Physiol ; 602(9): 1967-1986, 2024 May.
Article in English | MEDLINE | ID: mdl-38564214

ABSTRACT

Mitochondria within skeletal muscle cells are located either between the muscle contractile apparatus (interfibrillar mitochondria, IFM) or beneath the cell membrane (subsarcolemmal mitochondria, SSM), with several structural and functional differences reported between IFM and SSM. However, recent 3D imaging studies demonstrate that mitochondria are particularly concentrated in the proximity of capillaries embedded in sarcolemmal grooves rather than in proximity to the sarcolemma itself (paravascular mitochondria, PVM). To evaluate the impact of capillary vs. sarcolemmal proximity, we compared the structure and function of skeletal muscle mitochondria located either lateral to embedded capillaries (PVM), adjacent to the sarcolemma but not in PVM pools (SSM) or interspersed between sarcomeres (IFM). Mitochondrial morphology and interactions were assessed by 3D electron microscopy coupled with machine learning segmentation, whereas mitochondrial energy conversion was assessed by two-photon microscopy of mitochondrial membrane potential, content, calcium, NADH redox and flux in live, intact cells. Structurally, although PVM and SSM were similarly larger than IFM, PVM were larger, rounder and had more physical connections to neighbouring mitochondria compared to both IFM and SSM. Functionally, PVM had similar or greater basal NADH flux compared to SSM and IFM, respectively, despite a more oxidized NADH pool and a greater membrane potential, signifying a greater activation of the electron transport chain in PVM. Together, these data indicate that proximity to capillaries has a greater impact on resting mitochondrial energy conversion and distribution in skeletal muscle than the sarcolemma alone. KEY POINTS: Capillaries have a greater impact on mitochondrial energy conversion in skeletal muscle than the sarcolemma. Paravascular mitochondria are larger, and the outer mitochondrial membrane is more connected with neighbouring mitochondria. Interfibrillar mitochondria are longer and have greater contact sites with other organelles (i.e. sarcoplasmic reticulum and lipid droplets). Paravascular mitochondria have greater activation of oxidative phosphorylation than interfibrillar mitochondria at rest, although this is not regulated by calcium.


Subject(s)
Capillaries , Mitochondria, Muscle , Muscle, Skeletal , Sarcolemma , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Sarcolemma/physiology , Animals , Capillaries/physiology , Capillaries/metabolism , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/blood supply , Mice , Energy Metabolism/physiology , Male , Mice, Inbred C57BL , Membrane Potential, Mitochondrial/physiology
2.
Biochem Biophys Res Commun ; 595: 89-95, 2022 03 05.
Article in English | MEDLINE | ID: mdl-35121232

ABSTRACT

In cardiac muscle cells, heterodimeric integrin transmembrane receptors are known to serve as mechanotransducers, translating mechanical force to biochemical signaling. However, the roles of many individual integrins have still not been delineated. In this report, we demonstrate that Itga3b is localized to the sarcolemma of cardiomyocytes from 24 to 96 hpf. We further show that heterozygous and homozygous itga3b/bdf mutant embryos display a cardiomyopathy phenotype, with decreased cardiac contractility and reduced cardiomyocyte number. Correspondingly, proliferation of ventricular and atrial cardiomyoctyes and ventricular epicardial cells is decreased in itga3b mutant hearts. The contractile dysfunction of itga3b mutants can be attributed to cardiomyocyte sarcomeric disorganization, including thin myofilaments with blurred and shortened Z-discs. Together, our results reveal that Itga3b localizes to the myocardium sarcolemma, and it is required for cardiac contractility and cardiomyocyte proliferation.


Subject(s)
Integrin alpha3/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Apoptosis/genetics , Cell Proliferation/genetics , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Situ Hybridization , Integrin alpha3/metabolism , Microscopy, Electron, Transmission , Mutation , Myocardial Contraction/genetics , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/ultrastructure , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Sarcomeres/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
3.
J Cell Biol ; 220(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34633413

ABSTRACT

The cavin proteins are essential for caveola biogenesis and function. Here, we identify a role for the muscle-specific component, Cavin4, in skeletal muscle T-tubule development by analyzing two vertebrate systems, mouse and zebrafish. In both models, Cavin4 localized to T-tubules, and loss of Cavin4 resulted in aberrant T-tubule maturation. In zebrafish, which possess duplicated cavin4 paralogs, Cavin4b was shown to directly interact with the T-tubule-associated BAR domain protein Bin1. Loss of both Cavin4a and Cavin4b caused aberrant accumulation of interconnected caveolae within the T-tubules, a fragmented T-tubule network enriched in Caveolin-3, and an impaired Ca2+ response upon mechanical stimulation. We propose a role for Cavin4 in remodeling the T-tubule membrane early in development by recycling caveolar components from the T-tubule to the sarcolemma. This generates a stable T-tubule domain lacking caveolae that is essential for T-tubule function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Sarcolemma/metabolism , Tumor Suppressor Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Caveolae/metabolism , Cell Line , Embryo, Nonmammalian/metabolism , Imaging, Three-Dimensional , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/ultrastructure , Protein Binding , Sarcolemma/ultrastructure , Zebrafish/embryology
4.
Neurogenetics ; 22(2): 117-125, 2021 05.
Article in English | MEDLINE | ID: mdl-33811585

ABSTRACT

We report a multiplex family with extended multisystem neurological phenotype associated with a CRYAB variant. Two affected siblings were evaluated with whole exome sequencing, muscle biopsy, laser microdissection, and mass spectrometry-based proteomic analysis. Both patients and their mother manifested a combination of early-onset cataracts, cardiomyopathy, cerebellar ataxia, optic atrophy, cognitive impairment, and myopathy. Whole exome sequencing identified a heterozygous c.458C>T variant mapped to the C-terminal extension domain of the Alpha-crystallin B chain, disrupting its function as a molecular chaperone and its ability to suppress protein aggregation. In accordance with the molecular findings, muscle biopsies revealed subsarcolemmal deposits that appeared dark with H&E and trichrome staining were negative for the other routine histochemical staining and for amyloid with the Congo-red stain. Electron microscopy demonstrated that the deposits were composed of numerous parallel fibrils. Laser microdissection and mass spectrometry-based proteomic analysis revealed that the inclusions are almost exclusively composed of crystallized chaperones/heat shock proteins. Moreover,  a structural model suggests that Ser153 could be involved in monomer stabilization, dimer association, and possible binding of partner proteins. We propose that our report potentially expands the complex phenotypic spectrum of alpha B-crystallinopathies with possible effect of a CRYAB variant on the central nervous system.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cataract/genetics , Cerebellar Ataxia/genetics , Cognitive Dysfunction/genetics , Optic Atrophy/genetics , Sarcolemma/ultrastructure , alpha-Crystallin B Chain/genetics , Amino Acid Sequence , Biopsy , Death, Sudden, Cardiac/etiology , Female , Humans , Inclusion Bodies/ultrastructure , Jews/genetics , Male , Middle Aged , Models, Molecular , Muscle Weakness/genetics , Muscle, Skeletal/pathology , Phenotype , Protein Conformation , Exome Sequencing
5.
Circ Res ; 128(2): 203-215, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33228470

ABSTRACT

RATIONALE: The sarcolemma of cardiomyocytes contains many proteins that are essential for electromechanical function in general, and excitation-contraction coupling in particular. The distribution of these proteins is nonuniform between the bulk sarcolemmal surface and membrane invaginations known as transverse tubules (TT). TT form an intricate network of fluid-filled conduits that support electromechanical synchronicity within cardiomyocytes. Although continuous with the extracellular space, the narrow lumen and the tortuous structure of TT can form domains of restricted diffusion. As a result of unequal ion fluxes across cell surface and TT membranes, limited diffusion may generate ion gradients within TT, especially deep within the TT network and at high pacing rates. OBJECTIVE: We postulate that there may be an advective component to TT content exchange, wherein cyclic deformation of TT during diastolic stretch and systolic shortening serves to mix TT luminal content and assists equilibration with bulk extracellular fluid. METHODS AND RESULTS: Using electron tomography, we explore the 3-dimensional nanostructure of TT in rabbit ventricular myocytes, preserved at different stages of the dynamic cycle of cell contraction and relaxation. We show that cellular deformation affects TT shape in a sarcomere length-dependent manner and on a beat-by-beat time-scale. Using fluorescence recovery after photobleaching microscopy, we show that apparent speed of diffusion is affected by the mechanical state of cardiomyocytes, and that cyclic contractile activity of cardiomyocytes accelerates TT diffusion dynamics. CONCLUSIONS: Our data confirm the existence of an advective component to TT content exchange. This points toward a novel mechanism of cardiac autoregulation, whereby the previously implied increased propensity for TT luminal concentration imbalances at high electrical stimulation rates would be countered by elevated advection-assisted diffusion at high mechanical beating rates. The relevance of this mechanism in health and during pathological remodeling (eg, cardiac hypertrophy or failure) forms an exciting target for further research.


Subject(s)
Excitation Contraction Coupling , Heart Rate , Myocardial Contraction , Myocytes, Cardiac/metabolism , Sarcolemma/metabolism , Action Potentials , Animals , Diffusion , Electron Microscope Tomography , Female , Fluorescence Recovery After Photobleaching , Myocytes, Cardiac/ultrastructure , Rabbits , Sarcolemma/ultrastructure
6.
PLoS Genet ; 16(11): e1009179, 2020 11.
Article in English | MEDLINE | ID: mdl-33175853

ABSTRACT

Gene therapy approaches for DMD using recombinant adeno-associated viral (rAAV) vectors to deliver miniaturized (or micro) dystrophin genes to striated muscles have shown significant progress. However, concerns remain about the potential for immune responses against dystrophin in some patients. Utrophin, a developmental paralogue of dystrophin, may provide a viable treatment option. Here we examine the functional capacity of an rAAV-mediated microutrophin (µUtrn) therapy in the mdx4cv mouse model of DMD. We found that rAAV-µUtrn led to improvement in dystrophic histopathology & mostly restored the architecture of the neuromuscular and myotendinous junctions. Physiological studies of tibialis anterior muscles indicated peak force maintenance, with partial improvement of specific force. A fundamental question for µUtrn therapeutics is not only can it replace critical functions of dystrophin, but whether full-length utrophin impacts the therapeutic efficacy of the smaller, highly expressed µUtrn. As such, we found that µUtrn significantly reduced the spacing of the costameric lattice relative to full-length utrophin. Further, immunostaining suggested the improvement in dystrophic pathophysiology was largely influenced by favored correction of fast 2b fibers. However, unlike µUtrn, µdystrophin (µDys) expression did not show this fiber type preference. Interestingly, µUtrn was better able to protect 2a and 2d fibers in mdx:utrn-/- mice than in mdx4cv mice where the endogenous full-length utrophin was most prevalent. Altogether, these data are consistent with the role of steric hindrance between full-length utrophin & µUtrn within the sarcolemma. Understanding the stoichiometry of this effect may be important for predicting clinical efficacy.


Subject(s)
Genetic Therapy/methods , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/therapy , Utrophin/therapeutic use , Animals , Dependovirus/genetics , Disease Models, Animal , Dystrophin/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred mdx , Microscopy, Electron , Muscle Contraction , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Neuromuscular Junction/pathology , Neuromuscular Junction/ultrastructure , Sarcolemma/pathology , Sarcolemma/ultrastructure , Utrophin/genetics
7.
Nat Commun ; 11(1): 3711, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709891

ABSTRACT

The skeletal muscle T-tubule is a specialized membrane domain essential for coordinated muscle contraction. However, in the absence of genetically tractable systems the mechanisms involved in T-tubule formation are unknown. Here, we use the optically transparent and genetically tractable zebrafish system to probe T-tubule development in vivo. By combining live imaging of transgenic markers with three-dimensional electron microscopy, we derive a four-dimensional quantitative model for T-tubule formation. To elucidate the mechanisms involved in T-tubule formation in vivo, we develop a quantitative screen for proteins that associate with and modulate early T-tubule formation, including an overexpression screen of the entire zebrafish Rab protein family. We propose an endocytic capture model involving firstly, formation of dynamic endocytic tubules at transient nucleation sites on the sarcolemma, secondly, stabilization by myofibrils/sarcoplasmic reticulum and finally, delivery of membrane from the recycling endosome and Golgi complex.


Subject(s)
Muscle Contraction/physiology , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Sarcolemma/physiology , Sarcolemma/ultrastructure , Animals , Calcium Channels/metabolism , Calcium Channels/ultrastructure , Calcium Channels, L-Type/metabolism , Carrier Proteins/metabolism , Developmental Biology , Golgi Apparatus/metabolism , Male , Microscopy, Electron , Muscle Proteins/chemistry , Muscle, Skeletal/chemistry , Myofibrils/metabolism , Sarcolemma/chemistry , Sarcoplasmic Reticulum/metabolism , Zebrafish
8.
Neuropathol Appl Neurobiol ; 46(6): 602-614, 2020 10.
Article in English | MEDLINE | ID: mdl-32573804

ABSTRACT

BACKGROUND: Dmdmdx , harbouring the c.2983C>T nonsense mutation in Dmd exon 23, is a mouse model for Duchenne muscular dystrophy (DMD), frequently used to test therapies aimed at dystrophin restoration. Current translational research is methodologically hampered by the lack of a reporter mouse model, which would allow direct visualization of dystrophin expression as well as longitudinal in vivo studies. METHODS: We generated a DmdEGFP-mdx reporter allele carrying in cis the mdx-23 mutation and a C-terminal EGFP-tag. This mouse model allows direct visualization of spontaneously and therapeutically restored dystrophin-EGFP fusion protein either after natural fibre reversion, or for example, after splice modulation using tricyclo-DNA to skip Dmd exon 23, or after gene editing using AAV-encoded CRISPR/Cas9 for Dmd exon 23 excision. RESULTS: Intravital microscopy in anaesthetized mice allowed live-imaging of sarcolemmal dystrophin-EGFP fusion protein of revertant fibres as well as following therapeutic restoration. Dystrophin-EGFP-fluorescence persisted ex vivo, allowing live-imaging of revertant and therapeutically restored dystrophin in isolated fibres ex vivo. Expression of the shorter dystrophin-EGFP isoforms Dp71 in the brain, Dp260 in the retina, and Dp116 in the peripheral nerve remained unabated by the mdx-23 mutation. CONCLUSION: Intravital imaging of DmdEGFP-mdx muscle permits novel experimental approaches such as the study of revertant and therapeutically restored dystrophin in vivo and ex vivo.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Alleles , Animals , CRISPR-Cas Systems , Exons , Gene Editing , Genetic Therapy , Humans , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscular Dystrophy, Duchenne/therapy , Retina/metabolism , Sarcolemma/metabolism , Sarcolemma/ultrastructure
9.
Int J Mol Sci ; 21(2)2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31947691

ABSTRACT

The arrhythmogenic potential of ß1-adrenoceptor autoantibodies (ß1-AA), as well as antiarrhythmic properties of omega-3 in heart diseases, have been reported while underlying mechanisms are poorly understood. We aimed to test our hypothesis that omega-3 (eicosapentaenoic acid-EPA, docosahexaenoic acid-DHA) may inhibit matrix metalloproteinase (MMP-2) activity to prevent cleavage of ß1-AR and formation of ß1-AA resulting in attenuation of pro-arrhythmic connexin-43 (Cx43) and protein kinase C (PKC) signaling in the diseased heart. We have demonstrated that the appearance and increase of ß1-AA in blood serum of male and female 12-month-old spontaneously hypertensive rats (SHR) was associated with an increase of inducible ventricular fibrillation (VF) comparing to normotensive controls. In contrast, supplementation of hypertensive rats with omega-3 for two months suppressed ß1-AA levels and reduced incidence of VF. Suppression of ß1-AA was accompanied by a decrease of elevated myocardial MMP-2 activity, preservation of cardiac cell membrane integrity and Cx43 topology. Moreover, omega-3 abrogated decline in expression of total Cx43 as well as its phosphorylated forms at serine 368 along with PKC-ε, while decreased pro-fibrotic PKC-δ levels in hypertensive rat heart regardless the sex. The implication of MMP-2 in the action of omega-3 was also demonstrated in cultured cardiomyocytes in which desensitization of ß1-AR due to permanent activation of ß1-AR with isoproterenol was prevented by MMP-2 inhibitor or EPA. Collectively, these data support the notion that omega-3 via suppression of ß1-AA mechanistically controlled by MMP-2 may attenuate abnormal of Cx43 and PKC-ε signaling; thus, abolish arrhythmia substrate and protect rats with an advanced stage of hypertension from malignant arrhythmias.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/etiology , Autoantibodies/immunology , Autoantigens/immunology , Fatty Acids, Omega-3/pharmacology , Hypertension/complications , Receptors, Adrenergic, beta-1/immunology , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Biomarkers , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Connexin 43/metabolism , Disease Models, Animal , Disease Susceptibility , Fatty Acids, Omega-3/metabolism , Female , Male , Matrix Metalloproteinase 2/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Protein Kinase C-epsilon/metabolism , Rats , Rats, Inbred SHR , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Ventricular Fibrillation/drug therapy , Ventricular Fibrillation/etiology , Ventricular Fibrillation/physiopathology
10.
J Struct Biol ; 209(1): 107411, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31689503

ABSTRACT

Dystrophin is a large intracellular protein that prevents sarcolemmal ruptures by providing a mechanical link between the intracellular actin cytoskeleton and the transmembrane dystroglycan complex. Dystrophin deficiency leads to the severe muscle wasting disease Duchenne Muscular Dystrophy and the milder allelic variant, Becker Muscular Dystrophy (DMD and BMD). Previous work has shown that concomitant interaction of the actin binding domain 2 (ABD2) comprising spectrin like repeats 11 to 15 (R11-15) of the central domain of dystrophin, with both actin and membrane lipids, can greatly increase membrane stiffness. Based on a combination of SAXS and SANS measurements, mass spectrometry analysis of cross-linked complexes and interactive low-resolution simulations, we explored in vitro the molecular properties of dystrophin that allow the formation of ABD2-F-actin and ABD2-membrane model complexes. In dystrophin we identified two subdomains interacting with F-actin, one located in R11 and a neighbouring region in R12 and another one in R15, while a single lipid binding domain was identified at the C-terminal end of R12. Relative orientations of the dystrophin central domain with F-actin and a membrane model were obtained from docking simulation under experimental constraints. SAXS-based models were then built for an extended central subdomain from R4 to R19, including ABD2. Overall results are compatible with a potential F-actin/dystrophin/membrane lipids ternary complex. Our description of this selected part of the dystrophin associated complex bridging muscle cell membrane and cytoskeleton opens the way to a better understanding of how cell muscle scaffolding is maintained through this essential protein.


Subject(s)
Dystrophin/ultrastructure , Muscular Dystrophy, Duchenne/genetics , Sarcolemma/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/ultrastructure , Actins/genetics , Actins/ultrastructure , Dystrophin/genetics , Humans , Lipids/chemistry , Lipids/genetics , Muscular Dystrophy, Duchenne/pathology , Protein Binding , Sarcolemma/ultrastructure , Scattering, Small Angle , Ternary Complex Factors/genetics , Ternary Complex Factors/ultrastructure , X-Ray Diffraction
11.
PLoS One ; 14(5): e0216720, 2019.
Article in English | MEDLINE | ID: mdl-31145728

ABSTRACT

Computer assisted image acquisition techniques, including confocal microscopy, require efficient tools for an automatic sorting of vast amount of generated image data. The complexity of the classification process, absence of adequate tools, and insufficient amount of reference data has made the automated processing of images challenging. Mastering of this issue would allow implementation of statistical analysis in research areas such as in research on formation of t-tubules in cardiac myocytes. We developed a system aimed at automatic assessment of cardiomyocyte development stages (SAACS). The system classifies confocal images of cardiomyocytes with fluorescent dye stained sarcolemma. We based SAACS on a densely connected convolutional network (DenseNet) topology. We created a set of labelled source images, proposed an appropriate data augmentation technique and designed a class probability graph. We showed that the DenseNet topology, in combination with the augmentation technique is suitable for the given task, and that high-resolution images are instrumental for image categorization. SAACS, in combination with the automatic high-throughput confocal imaging, will allow application of statistical analysis in the research of the tubular system development or remodelling and loss.


Subject(s)
Image Processing, Computer-Assisted/methods , Myocytes, Cardiac/cytology , Animals , Artificial Intelligence , Cell Differentiation , Fluorescent Dyes , Machine Learning , Microscopy, Confocal/methods , Models, Cardiovascular , Myocytes, Cardiac/ultrastructure , Neural Networks, Computer , Rats , Sarcolemma/ultrastructure
12.
Ann Anat ; 224: 117-123, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31117003

ABSTRACT

Specific ultrastructural anatomy of masticatory muscles is commonly referred to a general pattern assigned to striated muscles. Junctional feet consisting of calcium channels of the sarcoplasmic reticulum (i.e. the ryanodine receptors, RyRs) physically connected to the calcium channels of the t-tubules build triads within striated muscles. Functional RyRs were demonstrated in the nuclear envelopes of pancreas and of a skeletal muscle derived cell line, but not in muscle in situ. It was hypothesized that ryanodine receptors (RyRs) could also exist in the nuclear envelope in the masseter muscle, thus aiming at studying this by transmission electron microscopy. There were identified paired and consistent subsarcolemmal clusters of mitochondria, appearing as outpockets of the muscle fibers, usually flanking an endomysial microvessel. It was observed on grazing longitudinal cuts that the I-band-limited mitochondria were not strictly located in a single intermyofibrillar space but continued transversally over the I-band to the next intermyofibrillar space. It appeared that the I-band-limited transverse mitochondria participate with the column-forming mitochondria in building a rather incomplete mitochondrial reticulum of the masseter muscle. Subsarcolemmal nuclei presented nuclear envelope-associated RyRs. Moreover, t-tubules were contacting the nuclear envelope and they were seemingly filled from the perinuclear space. This could suggest that nucleoplasmic calcium could contribute to balance the cytosolic concentration via pre-built anatomical routes: (i) indirectly, via the RyRs of the nuclear envelope and (ii) directly via the communication of t-tubules and sarcoplasmic reticulum through the perinuclear space.


Subject(s)
Calcium/metabolism , Masseter Muscle/metabolism , Masseter Muscle/ultrastructure , Animals , Cell Nucleus/ultrastructure , Male , Microscopy, Electron, Transmission , Microvessels/ultrastructure , Mitochondria/ultrastructure , Models, Animal , Muscle Fibers, Skeletal/ultrastructure , Myofibrils/ultrastructure , Nuclear Envelope/ultrastructure , Rabbits , Sarcolemma/ultrastructure , Sarcomeres/ultrastructure
13.
Metabolism ; 89: 18-26, 2018 12.
Article in English | MEDLINE | ID: mdl-30253140

ABSTRACT

BACKGROUND: Skeletal muscle mitochondrial content and function appear to be altered in obesity. Mitochondria in muscle are found in well-defined regions within cells, and they are arranged in a way that form distinct subpopulations of subsarcolemmal (SS) and intermyofibrillar (IMF) mitochondria. We sought to investigate differences in the proteomes of SS and IMF mitochondria between lean subjects and subjects with obesity. METHODS: We performed comparative proteomic analyses on SS and IMF mitochondria isolated from muscle samples obtained from lean subjects and subjects with obesity. Mitochondria were isolated using differential centrifugation, and proteins were subjected to label-free quantitative tandem mass spectrometry analyses. Collected data were evaluated for abundance of mitochondrial proteins using spectral counting. The Reactome pathway database was used to determine metabolic pathways that are altered in obesity. RESULTS: Among proteins, 73 and 41 proteins showed different (mostly lower) expression in subjects with obesity in the SS and IMF mitochondria, respectively (false discovery rate-adjusted P ≤ 0.05). We specifically found an increase in proteins forming the tricarboxylic acid cycle and electron transport chain (ETC) complex II, but a decrease in proteins forming protein complexes I and III of the ETC and adenosine triphosphate (ATP) synthase in subjects with obesity in the IMF, but not SS, mitochondria. Obesity was associated with differential effects on metabolic pathways linked to protein translation in the SS mitochondria and ATP formation in the IMF mitochondria. CONCLUSIONS: Obesity alters the expression of mitochondrial proteins regulating key metabolic processes in skeletal muscle, and these effects are distinct to mitochondrial subpopulations located in different regions of the muscle fibers. TRIAL REGISTRATION: ClinicalTrials.gov (NCT01824173).


Subject(s)
Mitochondria, Muscle/ultrastructure , Mitochondrial Proteins/metabolism , Obesity/metabolism , ATP Synthetase Complexes/metabolism , Adult , Female , Healthy Volunteers , Humans , Male , Metabolic Networks and Pathways , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Obesity/pathology , Proteomics , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , Tandem Mass Spectrometry
14.
Dis Model Mech ; 11(4)2018 04 16.
Article in English | MEDLINE | ID: mdl-29666144

ABSTRACT

Skeletal muscle atrophy is the most prominent feature of amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease of motor neurons. However, the contribution of skeletal muscle to disease progression remains elusive. Our previous studies have shown that intrathecal injection of cerebrospinal fluid from sporadic ALS patients (ALS-CSF) induces several degenerative changes in motor neurons and glia of neonatal rats. Here, we describe various pathologic events in the rat extensor digitorum longus muscle following intrathecal injection of ALS-CSF. Adenosine triphosphatase staining and electron microscopic (EM) analysis revealed significant atrophy and grouping of type 2 fibres in ALS-CSF-injected rats. Profound neuromuscular junction (NMJ) damage, such as fragmentation accompanied by denervation, were revealed by α-bungarotoxin immunostaining. Altered expression of key NMJ proteins, rapsyn and calpain, was also observed by immunoblotting. In addition, EM analysis showed sarcolemmal folding, Z-line streaming, structural alterations of mitochondria and dilated sarcoplasmic reticulum. The expression of trophic factors was affected, with significant downregulation of vascular endothelial growth factor (VEGF), marginal reduction in insulin-like growth factor-1 (IGF-1), and upregulation of brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF). However, motor neurons might be unable to harness the enhanced levels of BDNF and GDNF, owing to impaired NMJs. We propose that ALS-CSF triggers motor neuronal degeneration, resulting in pathological changes in the skeletal muscle. Muscle damage further aggravates the motor neuronal pathology, because of the interdependency between them. This sets in a vicious cycle, leading to rapid and progressive loss of motor neurons, which could explain the relentless course of ALS.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Muscle, Skeletal/pathology , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Newborn , Atrophy , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Down-Regulation , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Neuromuscular Junction/pathology , Oxidative Stress , Rats, Wistar , Sarcolemma/pathology , Sarcolemma/ultrastructure , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
15.
Muscle Nerve ; 58(2): 286-292, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29603301

ABSTRACT

INTRODUCTION: GNE myopathy is an adult-onset muscle disorder characterized by impaired sialylation of (muscle) glycans, detectable by lectin histochemistry. We describe a standardized method to quantify (lectin-) fluorescence in muscle sections, applicable for diagnosis and response to therapy for GNE myopathy. METHODS: Muscle sections were fluorescently labeled with the sialic acid-binding Sambucus nigra agglutinin (SNA) lectin and antibodies to sarcolemma residence protein caveolin-3 (CAV-3). Entire tissue sections were imaged in tiles and fluorescence was quantified. RESULTS: SNA fluorescence co-localizing with CAV-3 was ∼50% decreased in GNE myopathy biopsies compared with muscle-matched controls, confirming previous qualitative results. DISCUSSION: This quantitative fluorescence method can accurately determine sialylation status of GNE myopathy muscle biopsies. This method is adaptable for expression of other membrane-associated muscle proteins, and may be of benefit for disorders in which therapeutic changes in expression are subtle and difficult to assess by other methods. Muscle Nerve 58: 286-292, 2018.


Subject(s)
Distal Myopathies/pathology , Lectins , Muscle, Skeletal/pathology , Adult , Caveolin 3/genetics , Distal Myopathies/genetics , Female , Fluorescent Dyes , Humans , Image Processing, Computer-Assisted , Male , Microscopy, Confocal , Middle Aged , Plant Lectins , Ribosome Inactivating Proteins , Sarcolemma/pathology , Sarcolemma/ultrastructure
16.
Bull Exp Biol Med ; 164(4): 508-513, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29504114

ABSTRACT

The study examined the myocardial ultrastructural alterations in rats maintained on various atherogenic diets. It revealed the complex ultrastructural alterations of cardiomyocytes and endotheliocytes (including the lytic and destructive changes of the intracellular organelles, upregulation of the autophagocytosis in the cardiomyocytes, and necrobiosis with apoptosis of endotheliocytes) reflecting the cytopathic features of circulating cholesterol and lipoproteins, whose elevation determined the intensity of destructive processes. The revealed peculiarities in the changes of lipid inclusions (their osmiophilic transformation) in cardiomyocytes can be provoked by entry of cholesterol into the cells and its further metabolic modifications. During moderate dyslipidemia, the cardiomyocytes demonstrated the ultrastructural signs of induction of intracellular regeneration (marked with the clusters of polysomes in the intermyofibrillar and subsarcolemmal spaces with appearance of neogenic myofilaments) and upregulated pinocytotic activity. In all cases, up-regulated autophagocytosis in cardiomyocytes was accompanied by accumulation of myelin- and vacuole-like structures in the intercellular spaces and capillary lumens paralleled with appearance of activated forms of macrophages and fibroblasts in the myocardium.


Subject(s)
Cardiomyopathies/pathology , Dyslipidemias/pathology , Endothelial Cells/ultrastructure , Myocytes, Cardiac/ultrastructure , Myofibrils/ultrastructure , Animals , Antithyroid Agents/administration & dosage , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cholesterol/administration & dosage , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Diet, Atherogenic/adverse effects , Diet, High-Fat/adverse effects , Dyslipidemias/etiology , Dyslipidemias/metabolism , Endothelial Cells/metabolism , Methimazole/administration & dosage , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Polyribosomes/metabolism , Polyribosomes/ultrastructure , Rats , Rats, Wistar , Sarcolemma/metabolism , Sarcolemma/ultrastructure
17.
Mitochondrion ; 38: 1-5, 2018 01.
Article in English | MEDLINE | ID: mdl-28716666

ABSTRACT

Cardiomyocytes consist of longitudinally oriented myofibril bundles with a misaligned composition caused by the uneven contours of the intercalated discs. The cytoplasmic space harbors the organelles, including mitochondria. This study investigated whether cardiomyocytes contain spatially and ultrastructurally discrete pools of mitochondria that can be separated for structurally and functionally appraisal in (patho)physiology. Transmission electron microscopy disclosed continuous transitions of mitochondria without attributable characteristics from beneath the sarcolemma directly into the barrier-free cytoplasmic space between myofibrils. The various shapes and sizes of mitochondria are formed by myofibril positioning and the space available independent of their localization within the cardiomyocytes. Furthermore, the established enzymatic isolation procedure including proteinase treatment resulted in loss of mitochondrial proteins, as evidenced by immunogold labeling of Connexin43 in situ, a postulated marker for distinguishing mitochondrial subpopulations. Moreover, mitochondrial ATP produced in those mitochondria was not different. These findings preclude a spatial and ultrastructural grading of cardiac mitochondria and their distinct separation and classification in subsarcolemmal and interfibrillar subpopulations.


Subject(s)
Mitochondria/ultrastructure , Myocytes, Cardiac/ultrastructure , Animals , Male , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Myofibrils/ultrastructure , Sarcolemma/ultrastructure
18.
J Mol Cell Cardiol ; 112: 123-130, 2017 11.
Article in English | MEDLINE | ID: mdl-28822805

ABSTRACT

The cardiac transverse (T)-tubule membrane system is the safeguard for cardiac function and undergoes dramatic remodeling in response to cardiac stress. However, the mechanism by which cardiomyocytes repair damaged T-tubule network remains unclear. In the present study, we tested the hypothesis that MG53, a muscle-specific membrane repair protein, antagonizes T-tubule damage to protect against maladaptive remodeling and thereby loss of excitation-contraction coupling and cardiac function. Using MG53-knockout (MG53-KO) mice, we first established that deficiency of MG53 had no impact on maturation of the T-tubule network in developing hearts. Additionally, MG53 ablation did not influence T-tubule integrity in unstressed adult hearts as late as 10months of age. Following left ventricular pressure overload-induced cardiac stress, MG53 protein levels were increased by approximately three-fold in wild-type mice, indicating that pathological stress induces a significant upregulation of MG53. MG53-deficient mice had worsened T-tubule disruption and pronounced dysregulation of Ca2+ handling properties, including decreased Ca2+ transient amplitude and prolonged time to peak and decay. Moreover, MG53 deficiency exacerbated cardiac hypertrophy and dysfunction and decreased survival following cardiac stress. Our data suggest MG53 is not required for T-tubule development and maintenance in normal physiology. However, MG53 is essential to preserve T-tubule integrity and thereby Ca2+ handling properties and cardiac function under pathological cardiac stress.


Subject(s)
Carrier Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Sarcolemma/metabolism , Animals , Calcium Signaling , Down-Regulation , Excitation Contraction Coupling , Heart/embryology , Male , Membrane Proteins , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Sarcolemma/ultrastructure , Sodium-Calcium Exchanger/metabolism
19.
Int J Mol Med ; 40(4): 1078-1088, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28765879

ABSTRACT

Supramolecular membrane complexes of low abundance are difficult to study by routine bioanalytical techniques. The plasmalemmal complex consisting of sarcoglycans, dystroglycans, dystrobrevins and syntrophins, which is closely associated with the membrane cytoskeletal protein dystrophin, represents such a high­molecular­mass protein assembly in skeletal muscles. The almost complete loss of the dystrophin isoform Dp427­M and concomitant reduction in the dystrophin­associated glycoprotein complex is the underlying cause of the highly progressive neuromuscular disorder named Duchenne muscular dystrophy. This gives the detailed characterization of the dystrophin complex considerable pathophysiological importance. In order to carry out a comprehensive mass spectrometric identification of the dystrophin­glycoprotein complex, in this study, we used extensive subcellular fractionation and enrichment procedures prior to subproteomic analysis. Mass spectrometry identified high levels of full­length dystrophin isoform Dp427­M, α/ß­dystroglycans, α/ß/γ/δ­sarcoglycans, α1/ß1/ß2­syntrophins and α/ß­dystrobrevins in highly purified sarcolemma vesicles. By contrast, lower levels were detected in transverse tubules and no components of the dystrophin complex were identified in triads. For comparative purposes, the presence of organellar marker proteins was studied in crude surface membrane preparations vs. enriched fractions from the sarcolemma, transverse tubules and triad junctions using gradient gel electrophoresis and on­membrane digestion. This involved the subproteomic assessment of various ion­regulatory proteins and excitation­contraction coupling components. The comparative profiling of skeletal muscle fractions established a relatively restricted subcellular localization of the dystrophin­glycoprotein complex in the muscle fibre periphery by proteomic means and clearly demonstrated the absence of dystrophin from triad junctions by sensitive mass spectrometric analysis.


Subject(s)
Dystrophin-Associated Proteins/isolation & purification , Dystrophin/isolation & purification , Sarcoglycans/isolation & purification , Sarcolemma/chemistry , Sarcoplasmic Reticulum/chemistry , Animals , Excitation Contraction Coupling/physiology , Mass Spectrometry/methods , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Protein Isoforms/isolation & purification , Rabbits , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure
20.
J Am Heart Assoc ; 6(5)2017 May 04.
Article in English | MEDLINE | ID: mdl-28473402

ABSTRACT

BACKGROUND: Heart failure is a common secondary complication following a myocardial infarction (MI), characterized by impaired cardiac contraction and t-tubule (t-t) loss. However, post-MI nano-scale morphological changes to the remaining t-ts are poorly understood. METHOD AND RESULTS: We utilized a porcine model of MI, using a nonlethal microembolization method to generate controlled microinfarcts. Using serial block face scanning electron microscopy, we report that post-MI, after mild left-ventricular dysfunction has developed, t-ts are not only lost in the peri-infarct region, but also the remnant t-ts form enlarged, highly branched disordered structures, containing a dense intricate inner membrane. Biochemical and proteomics analyses showed that the calcium release channel, ryanodine receptor 2 (RyR2), abundance is unchanged, but junctophilin-2 (JP2), important for maintaining t-t trajectory, is depressed (-0.5×) in keeping with the t-ts being disorganized. However, immunolabeling shows that populations of RyR2 and JP2 remain associated with the remodeled t-ts. The bridging integrator 1 protein (BIN-1), a regulator of tubulogensis, is upregulated (+5.4×), consistent with an overdeveloped internal membrane system, a feature not present in control t-ts. Importantly, we have determined that t-ts, in the remote region, are narrowed and also contain dense membrane folds (BIN-1 is up-regulated +3.4×), whereas the t-ts have a radial organization comparable to control JP2 is upregulated +1.7×. CONCLUSIONS: This study reveals previously unidentified remodeling of the t-t nano-architecture in the post-MI heart that extends to the remote region. Our findings highlight that targeting JP2 may be beneficial for preserving the orientation of the t-ts, attenuating the development of hypocontractility post-MI.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Sarcolemma/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Disease Models, Animal , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Myocardial Contraction , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/ultrastructure , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcolemma/ultrastructure , Sus scrofa , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL