Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.687
Filter
1.
Clin Anat ; 36(8): 1138-1146, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37092576

ABSTRACT

Textbooks and atlases of human macroscopic and microscopic anatomy of the larynx generally provide, if at all, only sparse information on the laryngeal Musculus ventricularis. However, several studies indicate that this muscle takes over the function of vestibular (ventricular) fold phonation after denervation of the Musculus vocalis. In the present study, 29 laryngeal specimens were coronally dissected at different levels, i.e. the anterior (L1), middle (L2), and posterior third of the vestibular fold (L3), and they underwent histological analysis. In all specimens the vestibular folds of both hemi-larynxes contained striated muscle bundles in variable amounts, representing a ventricularis muscle. These muscle bundles obviously originated from the lateral (external) and thyroepiglottic part of the thyroarytenoid muscle and the aryepiglottic part of the oblique arytenoid muscle, as has been described by other authors. The areas of vestibular folds and their amounts of ventricularis muscle bundles were measured using image analysis software (imageJ) by manual tracing. The mean area of the vestibular folds of both hemi-larynxes was 27.9 mm2 (SD [standard deviation] ± 9.17), and the area occupied by fibers of the ventricularis muscle was 1.5 mm2 (SD ± 1.78). Statistical analysis comparing the areas of both hemi-larynxes and levels resulted in no significant differences, except for the levels 2 and 3. In level 2, significantly more muscle fibers (2.0 mm2 ; SD ± 2.21) were detectable within the vestibular fold than in level 3 (0.9 mm2 ; SD ± 1.43). Level 1 also contained more muscle fibers (1.1 mm2 ; SD ± 1.06) than level 3, however, without significance. In conclusion, the laryngeal ventricularis muscle is present in the majority of reported cases. Since the muscle is of clinical relevance, it should be included in anatomical textbooks by default.


Subject(s)
Larynx , Humans , Larynx/anatomy & histology , Laryngeal Muscles/anatomy & histology , Laryngeal Muscles/physiology , Vocal Cords/anatomy & histology , Vocal Cords/physiology , Muscle Fibers, Skeletal/ultrastructure , Clinical Relevance , Image Processing, Computer-Assisted
2.
Science ; 374(6565): 355-359, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34648328

ABSTRACT

Regeneration of skeletal muscle is a highly synchronized process that requires muscle stem cells (satellite cells). We found that localized injuries, as experienced through exercise, activate a myofiber self-repair mechanism that is independent of satellite cells in mice and humans. Mouse muscle injury triggers a signaling cascade involving calcium, Cdc42, and phosphokinase C that attracts myonuclei to the damaged site via microtubules and dynein. These nuclear movements accelerate sarcomere repair and locally deliver messenger RNA (mRNA) for cellular reconstruction. Myofiber self-repair is a cell-autonomous protective mechanism and represents an alternative model for understanding the restoration of muscle architecture in health and disease.


Subject(s)
Cell Nucleus/physiology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Regeneration , Sarcomeres/physiology , Animals , Calcium/metabolism , Dyneins/metabolism , Mice , Microtubules/metabolism , Muscle Contraction , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/ultrastructure , RNA, Messenger/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/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.
Sci Rep ; 11(1): 18161, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34518586

ABSTRACT

Megaconial Congenital Muscular Dystrophy (CMD) is a rare autosomal recessive disorder characterized by enlarged mitochondria located mainly at the periphery of muscle fibers and caused by mutations in the Choline Kinase Beta (CHKB) gene. Although the pathogenesis of this disease is not well understood, there is accumulating evidence for the presence of mitochondrial dysfunction. In this study, we aimed to investigate whether imbalanced mitochondrial dynamics affects mitochondrial function and bioenergetic efficiency in skeletal muscle cells of Megaconial CMD. Immunofluorescence, confocal and transmission electron microscopy studies revealed impaired mitochondrial network, morphology, and localization in primary skeletal muscle cells of Megaconial CMD. The organelle disruption was specific only to skeletal muscle cells grown in culture. The expression levels of mitochondrial fission proteins (DRP1, MFF, FIS1) were found to be decreased significantly in both primary skeletal muscle cells and tissue sections of Megaconial CMD by Western blotting and/or immunofluorescence analysis. The metabolomic and fluxomic analysis, which were performed in Megaconial CMD for the first time, revealed decreased levels of phosphonucleotides, Krebs cycle intermediates, ATP, and altered energy metabolism pathways. Our results indicate that reduced mitochondrial fission and altered mitochondrial energy metabolism contribute to mitochondrial dysmorphology and dysfunction in the pathogenesis of Megaconial CMD.


Subject(s)
Energy Metabolism , Mitochondrial Dynamics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Cells, Cultured , Fluorescence , Humans , Metabolic Flux Analysis , Metabolomics , Mitochondrial Proteins/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/ultrastructure
5.
Elife ; 102021 08 27.
Article in English | MEDLINE | ID: mdl-34448452

ABSTRACT

Skeletal muscles are composed of hundreds of multinucleated muscle fibers (myofibers) whose myonuclei are regularly positioned all along the myofiber's periphery except the few ones clustered underneath the neuromuscular junction (NMJ) at the synaptic zone. This precise myonuclei organization is altered in different types of muscle disease, including centronuclear myopathies (CNMs). However, the molecular machinery regulating myonuclei position and organization in mature myofibers remains largely unknown. Conversely, it is also unclear how peripheral myonuclei positioning is lost in the related muscle diseases. Here, we describe the microtubule-associated protein, MACF1, as an essential and evolutionary conserved regulator of myonuclei positioning and maintenance, in cultured mammalian myotubes, in Drosophila muscle, and in adult mammalian muscle using a conditional muscle-specific knockout mouse model. In vitro, we show that MACF1 controls microtubules dynamics and contributes to microtubule stabilization during myofiber's maturation. In addition, we demonstrate that MACF1 regulates the microtubules density specifically around myonuclei, and, as a consequence, governs myonuclei motion. Our in vivo studies show that MACF1 deficiency is associated with alteration of extra-synaptic myonuclei positioning and microtubules network organization, both preceding NMJ fragmentation. Accordingly, MACF1 deficiency results in reduced muscle excitability and disorganized triads, leaving voltage-activated sarcoplasmic reticulum Ca2+ release and maximal muscle force unchanged. Finally, adult MACF1-KO mice present an improved resistance to fatigue correlated with a strong increase in mitochondria biogenesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microfilament Proteins/metabolism , Microtubules/metabolism , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Neuromuscular Junction/metabolism , Organelle Biogenesis , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Excitation Contraction Coupling , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microtubules/genetics , Microtubules/ultrastructure , Mitochondria, Muscle/genetics , Mitochondria, Muscle/ultrastructure , Muscle Fatigue , Muscle Fibers, Skeletal/ultrastructure , Muscle Strength , Myoblasts, Skeletal/ultrastructure , Neuromuscular Junction/genetics , Neuromuscular Junction/ultrastructure , Time Factors
6.
Am J Physiol Cell Physiol ; 321(4): C749-C759, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34406904

ABSTRACT

Recently, methods for creating three-dimensional (3-D) human skeletal muscle tissues from myogenic cell lines have been reported. Bioengineered muscle tissues are contractile and respond to electrical and chemical stimulation. In this study, we provide an electrophysiological analysis of healthy and dystrophic 3-D bioengineered skeletal muscle tissues, focusing on Duchenne muscular dystrophy (DMD). We enlist the 3-D in vitro model of DMD muscle tissue to evaluate muscle cell electrical properties uncoupled from presynaptic neural inputs, an understudied aspect of DMD. Our data show that previously reported electrophysiological aspects of DMD, including effects on membrane potential and membrane resistance, are replicated in the 3-D muscle tissue model. Furthermore, we test a potential therapeutic compound, poloxamer 188, and demonstrate capacity for improving the membrane potential in DMD muscle. Therefore, this study serves as a baseline for a new in vitro method to examine potential therapies for muscular disorders.


Subject(s)
Dystrophin/metabolism , Membrane Potentials , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myoblasts, Skeletal/metabolism , Tissue Engineering , Adolescent , Case-Control Studies , Cell Culture Techniques , Cell Line , Child , Dystrophin/genetics , Electric Impedance , Humans , Male , Membrane Potentials/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/ultrastructure , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Mutation , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/ultrastructure , Poloxamer/pharmacology , Sodium/metabolism
7.
Physiol Rep ; 9(13): e14927, 2021 07.
Article in English | MEDLINE | ID: mdl-34197700

ABSTRACT

Cachexia, a condition prevalent in many chronically ill patients, is characterized by weight loss, fatigue, and decreases in muscle mass and function. Cachexia is associated with tumor burden and disease-related malnutrition, but other studies implicate chemotherapy as being causative. We investigated the effects of a chemotherapy drug cocktail on myofibrillar protein abundance and synthesis, anabolic signaling mechanisms, and substrate availability. On day 4 of differentiation, L6 myotubes were treated with vehicle (1.4 µl/ml DMSO) or a chemotherapy drug cocktail (a mixture of cisplatin [20 µg/ml], leucovorin [10 µg/ml], and 5-fluorouracil [5-FLU; 50 µg/ml]) for 24-72 h. Compared to myotubes treated with vehicle, those treated with the drug cocktail showed 50%-80% reductions in the abundance of myofibrillar proteins, including myosin heavy chain-1, troponin, and tropomyosin (p < 0.05). Cells treated with only a mixture of cisplatin and 5-FLU had identical reductions in myofibrillar protein abundance. Myotubes treated with the drug cocktail also showed >50% reductions in the phosphorylation of AKTSer473 and of mTORC1 substrates ribosomal protein S6Ser235/236 , its kinase S6K1Thr389 and eukaryotic translation initiation factor 4E-binding protein 1 (all p < 0.05). Drug treatment impaired peptide chain initiation in myofibrillar protein fractions and insulin-stimulated glucose uptake (p = 0.06) but increased the expression of autophagy markers beclin-1 and microtubule-associated proteins 1A/1B light chain 3B (p < 0.05), and of apoptotic marker, cleaved caspase 3 (p < 0.05). Drug treatment reduced the expression of mitochondrial markers cytochrome oxidase and succinate dehydrogenase (p < 0.05). The observed profound negative effects of this chemotherapy drug cocktail on myotubes underlie a need for approaches that can reduce the negative effects of these drugs on muscle metabolism.


Subject(s)
Muscle Fibers, Skeletal/drug effects , Muscle Proteins/drug effects , Animals , Blotting, Western , Cachexia/chemically induced , Cells, Cultured , Cisplatin/administration & dosage , Cisplatin/pharmacology , Drug Therapy, Combination , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Leucovorin/administration & dosage , Leucovorin/pharmacology , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/analysis , Muscle Proteins/physiology , Myosin Heavy Chains/analysis , Rats , Tropomyosin/analysis , Troponin/analysis
8.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209663

ABSTRACT

The myotendinous junction (MTJ) is the muscle-tendon interface and constitutes an integrated mechanical unit to force transmission. Joint immobilization promotes muscle atrophy via disuse, while physical exercise can be used as an adaptative stimulus. In this study, we aimed to investigate the components of the MTJ and their adaptations and the associated elements triggered with aquatic training after joint immobilization. Forty-four male Wistar rats were divided into sedentary (SD), aquatic training (AT), immobilization (IM), and immobilization/aquatic training (IMAT) groups. The samples were processed to measure fiber area, nuclear fractal dimension, MTJ nuclear density, identification of telocytes, sarcomeres, and MTJ perimeter length. In the AT group, the maintenance of ultrastructure and elements in the MTJ region were observed; the IM group presented muscle atrophy effects with reduced MTJ perimeter; the IMAT group demonstrated that aquatic training after joint immobilization promotes benefits in the muscle fiber area and fractal dimension, in the MTJ region shows longer sarcomeres and MTJ perimeter. We identified the presence of telocytes in the MTJ region in all experimental groups. We concluded that aquatic training is an effective rehabilitation method after joint immobilization due to reduced muscle atrophy and regeneration effects on MTJ in rats.


Subject(s)
Adaptation, Physiological , Immobilization , Joints , Physical Conditioning, Animal , Physical Exertion , Tendons/physiology , Animals , Male , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Rats , Sarcomeres/ultrastructure , Tendons/cytology , Tendons/ultrastructure
9.
Meat Sci ; 179: 108527, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33962166

ABSTRACT

This study investigated the effect of low voltage electrostatic field (LVEF) on the microstructure damage and protein structure changes of prepared beef steak during freezing. The scanning electron microscopy results showed that LVEF-assisted freezing (LVEFF) minimized the gaps in the cross section between muscle fibers induced by freezing and thus improved fiber compactness. Furthermore, LVEFF reduced the length of the enlarged sarcomere, repaired the Z-line fractures, and intensified the dismission of the A band in the air-blast freezing (AF) process. The decreased carbonyl content and increased total sulfhydryl content indicated that LVEFF reduced protein oxidation in the freezing process. In addition, the results of Raman spectroscopy and fluorescence spectroscopy revealed that LVEFF minimized the changes in protein secondary and tertiary structures during freezing. In conclusion, utilization of LVEF in the freezing of prepared beef steak could reduce both the microstructure damage and protein structure changes in the freezing process.


Subject(s)
Frozen Foods , Meat Products/analysis , Static Electricity , Animals , Cattle , Food Handling/methods , Freezing , Male , Microscopy, Electron, Scanning/veterinary , Muscle Fibers, Skeletal/ultrastructure , Sarcomeres
10.
Meat Sci ; 179: 108530, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33946021

ABSTRACT

This study investigated the effects of l-arginine and l-lysine on the water holding capacity, shear force, color, and protein denaturation of frozen porcine Longissimus lumborum. Four batches were prepared, each corresponding to samples of an experimental treatment: without a cryoprotective solution, injecting a 0.3% sodium tripolyphosphate and 0.5% NaCl solution, a 0.5% l-arginine solution, or a 0.5% l-lysine solution. The results showed that both l-arginine and l-lysine decreased thawing loss, cooking loss, shear force, L⁎ values, b⁎ values, and surface hydrophobicity, but they increased pH values, a⁎ values, percentages of peak areas for T21 relaxation times, and Ca2+-ATPase activity. Additionally, both histological and transmission electron microscopy images showed that l-lysine, and especially l-arginine could inhibit the formation of gaps between fiber bundles, alleviate the disruption of intracellular spaces, and maintain the structural integrity of sarcomeres. Overall, the results showed that both l-arginine and l-lysine hindered the structural damage of muscle fibers during freezing and protected myofibrillar proteins from denaturation, ultimately contributing to superior quality attributes.


Subject(s)
Arginine/chemistry , Freezing , Lysine/chemistry , Pork Meat/analysis , Animals , Color , Cooking , Cryoprotective Agents , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/metabolism , Shear Strength , Sus scrofa , Water
11.
PLoS Genet ; 17(3): e1009488, 2021 03.
Article in English | MEDLINE | ID: mdl-33780446

ABSTRACT

Mitochondria are essential for maintaining skeletal muscle metabolic homeostasis during adaptive response to a myriad of physiologic or pathophysiological stresses. The mechanisms by which mitochondrial function and contractile fiber type are concordantly regulated to ensure muscle function remain poorly understood. Evidence is emerging that the Folliculin interacting protein 1 (Fnip1) is involved in skeletal muscle fiber type specification, function, and disease. In this study, Fnip1 was specifically expressed in skeletal muscle in Fnip1-transgenic (Fnip1Tg) mice. Fnip1Tg mice were crossed with Fnip1-knockout (Fnip1KO) mice to generate Fnip1TgKO mice expressing Fnip1 only in skeletal muscle but not in other tissues. Our results indicate that, in addition to the known role in type I fiber program, FNIP1 exerts control upon muscle mitochondrial oxidative program through AMPK signaling. Indeed, basal levels of FNIP1 are sufficient to inhibit AMPK but not mTORC1 activity in skeletal muscle cells. Gain-of-function and loss-of-function strategies in mice, together with assessment of primary muscle cells, demonstrated that skeletal muscle mitochondrial program is suppressed via the inhibitory actions of FNIP1 on AMPK. Surprisingly, the FNIP1 actions on type I fiber program is independent of AMPK and its downstream PGC-1α. These studies provide a vital framework for understanding the intrinsic role of FNIP1 as a crucial factor in the concerted regulation of mitochondrial function and muscle fiber type that determine muscle fitness.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Female , Gene Expression Profiling , Male , Mice , Mice, Transgenic , Mitochondria, Muscle/ultrastructure , Muscle Fibers, Skeletal/ultrastructure , Organ Specificity , Oxidation-Reduction , Oxidative Stress
12.
Meat Sci ; 175: 108442, 2021 May.
Article in English | MEDLINE | ID: mdl-33540360

ABSTRACT

Thai beef (Bos indicus) samples were sous-vide-cooked at temperatures of 60°C, 70°C or 80°C for 2 to 36 hrs and prepared for microstructure characterization by light and electron microscopy. Muscle fibers showed a first phase of lateral shrinkage during the first 6 hrs of cooking at 60-70°C and the first 2 hrs at 80°C followed by a second phase of significant alternations of shrinkage and swelling independently of water transfers. Swelling peaked at 12 hrs. Microstructural changes were more variable for samples cooked at 60-70°C than for samples cooked at 80°C that showed a larger cross-sectional myofibrillar mass area (CSA). Hypercontracted fibers were evidenced at all temperature-time combinations and were associated with adjacent wavy fibers and a characteristic structural evolution in the mitochondria. The role of thermal denaturation of proteins and the ultrastructural analogy of hypercontracted fibers with cold-shortened fibers are discussed.


Subject(s)
Cooking/methods , Muscle Fibers, Skeletal/ultrastructure , Red Meat/analysis , Animals , Cattle , Microscopy, Electron, Transmission , Temperature , Time Factors
13.
Int J Mol Sci ; 22(3)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540821

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer-related death, and the prevalence of CRC in young adults is on the rise, making this a largescale clinical concern. Advanced CRC patients often present with liver metastases (LM) and an increased incidence of cachexia, i.e., musculoskeletal wasting. Despite its high incidence in CRC patients, cachexia remains an unresolved issue, and animal models for the study of CRC cachexia, in particular, metastatic CRC cachexia, remain limited; therefore, we aimed to establish a new model of metastatic CRC cachexia. C57BL/6 male mice (8 weeks old) were subcutaneously (MC38) or intrasplenically injected (mMC38) with MC38 murine CRC cells to disseminate LM, while experimental controls received saline (n = 5-8/group). The growth of subcutaneous MC38 tumors was accompanied by a reduction in skeletal muscle mass (-16%; quadriceps muscle), plantarflexion force (-22%) and extensor digitorum longus (EDL) contractility (-20%) compared to experimental controls. Meanwhile, the formation of MC38 LM (mMC38) led to heighted reductions in skeletal muscle mass (-30%; quadriceps), plantarflexion force (-28%) and EDL contractility (-35%) compared to sham-operated controls, suggesting exacerbated cachexia associated with LM. Moreover, both MC38 and mMC38 tumor hosts demonstrated a marked loss of bone indicated by reductions in trabecular (Tb.BV/TV: -49% in MC38, and -46% in mMC38) and cortical (C.BV/TV: -12% in MC38, and -8% in mMC38) bone. Cell culture experiments revealed that MC38 tumor-derived factors directly promote myotube wasting (-18%) and STAT3 phosphorylation (+5-fold), while the pharmacologic blockade of STAT3 signaling was sufficient to preserve myotube atrophy in the presence of MC38 cells (+21%). Overall, these results reinforce the notion that the formation of LM heightens cachexia in an experimental model of CRC.


Subject(s)
Adenocarcinoma/secondary , Cachexia/etiology , Colorectal Neoplasms/complications , Liver Neoplasms/secondary , Adenocarcinoma/complications , Adenocarcinoma/pathology , Animals , Cachexia/pathology , Cachexia/physiopathology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Progression , Femur/pathology , Liver Neoplasms/complications , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Muscle Contraction , Muscle Fibers, Skeletal/ultrastructure , Muscle Weakness/etiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/etiology , STAT3 Transcription Factor , Subcutaneous Tissue , X-Ray Microtomography
14.
Theranostics ; 11(7): 3331-3347, 2021.
Article in English | MEDLINE | ID: mdl-33537090

ABSTRACT

A spheroid is an aggregation of single cells with structural and functional characteristics similar to those of 3D native tissues, and it has been utilized as one of the typical in vitro three-dimensional (3D) cell models. Scaffold-free spheroids provide outstanding reflection of tissue complexity in a 3D in vivo-like environment, but they can neither fabricate realistic macroscale 3D complex structures without avoiding necrosis nor receive direct external stimuli (i.e., stimuli from mechanical or topographical cues). Here, we propose a spheroid-laden electrospinning process to obtain in vitro model achieved using the synergistic effect of the unique bioactive components provided by the spheroids and stimulating effects provided by the aligned nanofibers. Methods: To show the functional activity of the spheroid-laden structures, we used myoblast-spheroids to obtain skeletal muscle, comprising highly aligned myotubes, utilizing an uniaxially arranged topographical cue. The spheroid-electrospinning was used to align spheroids directly by embedding them in aligned alginate nanofibers, which were controlled with various materials and processing parameters. Results: The spheroids laden in the alginate nanofibers showed high cell viability (>90%) and was compared with that of a cell-laden alginate nanofiber that was electrospun with single cells. Consequently, the spheroids laden in the aligned nanofibers showed a significantly higher degree of myotube formation and maturation. Conclusion: Results suggested that the in vitro model using electrospun spheroids could potentially be employed to understand myogenic responses for various in vitro drug tests.


Subject(s)
Muscle Development/drug effects , Muscle Fibers, Skeletal/drug effects , Myoblasts/drug effects , Spheroids, Cellular/drug effects , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Alginates/chemistry , Alginates/pharmacology , Animals , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Electrochemical Techniques , Mice , Models, Biological , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/ultrastructure , Myoblasts/physiology , Myoblasts/ultrastructure , Spheroids, Cellular/physiology , Spheroids, Cellular/ultrastructure
15.
Cells ; 11(1)2021 12 27.
Article in English | MEDLINE | ID: mdl-35011629

ABSTRACT

High-resolution 3D images of organelles are of paramount importance in cellular biology. Although light microscopy and transmission electron microscopy (TEM) have provided the standard for imaging cellular structures, they cannot provide 3D images. However, recent technological advances such as serial block-face scanning electron microscopy (SBF-SEM) and focused ion beam scanning electron microscopy (FIB-SEM) provide the tools to create 3D images for the ultrastructural analysis of organelles. Here, we describe a standardized protocol using the visualization software, Amira, to quantify organelle morphologies in 3D, thereby providing accurate and reproducible measurements of these cellular substructures. We demonstrate applications of SBF-SEM and Amira to quantify mitochondria and endoplasmic reticulum (ER) structures.


Subject(s)
Algorithms , Imaging, Three-Dimensional , Microscopy, Electron, Scanning , Organelles/ultrastructure , Animals , Drosophila , Endoplasmic Reticulum , GTP Phosphohydrolases/deficiency , GTP Phosphohydrolases/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/ultrastructure
16.
J Gerontol A Biol Sci Med Sci ; 76(2): 244-252, 2021 01 18.
Article in English | MEDLINE | ID: mdl-32738046

ABSTRACT

The purpose of this investigation was to determine the effects of vocal training on neuromuscular junction (NMJ) morphology and muscle fiber size and composition in the thyroarytenoid muscle, the primary muscle in the vocal fold, in younger (9-month) and older (24-month) Fischer 344 × Brown Norway male rats. Over 4 or 8 weeks of vocal training, rats of both ages progressively increased their daily number of ultrasonic vocalizations (USVs) through operant conditioning and were then compared to an untrained control group. Neuromuscular junction morphology and myofiber size and composition were measured from the thyroarytenoid muscle. Acoustic analysis of USVs before and after training quantified the functional effect of training. Both 4- and 8-week training resulted in less NMJ motor endplate dispersion in the lateral portion of the thyroarytenoid muscle in rats of both ages. Vocal training and age had no significant effects on laryngeal myofiber size or type. Vocal training resulted in a greater number of USVs with longer duration and increased intensity. This study demonstrated that vocal training induces laryngeal NMJ morphology and acoustic changes. The lack of significant effects of vocal training on muscle fiber type and size suggests vocal training significantly improves neuromuscular efficiency but does not significantly influence muscle strength changes.


Subject(s)
Aging/physiology , Aging/psychology , Laryngeal Muscles/innervation , Laryngeal Muscles/physiology , Vocalization, Animal/physiology , Acoustics , Aging/pathology , Animals , Female , Laryngeal Muscles/anatomy & histology , Male , Motor Endplate/anatomy & histology , Motor Endplate/physiology , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/ultrastructure , Neuromuscular Junction/anatomy & histology , Neuromuscular Junction/physiology , Rats , Rats, Inbred BN , Rats, Inbred F344 , Ultrasonics
17.
Scand J Med Sci Sports ; 31(2): 303-312, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33038024

ABSTRACT

The repair, remodeling, and regeneration of myofibers are dependent on satellite cells (SCs), although, the distribution of SCs in different fiber types of human muscle remains inconclusive. There is also a paucity of research comparing muscle fiber characteristics in a sex-specific manner. Therefore, the aim of this study was to investigate fiber type-specific SC content in men and women. Muscle biopsies from vastus lateralis were collected from 64 young (mean age 27 ± 5), moderately trained men (n = 34) and women (n = 30). SCs were identified by Pax7-staining together with immunofluorescent analyses of fiber type composition, fiber size, and myonuclei content. In a mixed population, comparable number of SCs was associated to type I and type II fibers (0.07 ± 0.02 vs 0.07 ± 0.02 SCs per fiber, respectively). However, unlike men, women displayed a fiber type-specific distribution, with SC content being lower in type II than type I fibers (P = .041). Sex-based differences were found specifically for type II fibers, where women displayed lower SC content compared to men (P < .001). In addition, positive correlations (r-values between 0.36-0.56) were found between SC content and type I and type II fiber size in men (P = .03 and P < .01, respectively), whereas similar relationships could not be detected in women. Sex-based differences were also noted for fiber type composition and fiber size, but not for myonuclei content. We hereby provide evidence for sex-based differences present at the myocellular level, which may have important implications when studying exercise- and training-induced myogenic responses in skeletal muscle.


Subject(s)
Muscle Fibers, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Sex Factors , Adult , Cell Nucleus , Exercise/physiology , Female , Humans , Immunohistochemistry , Male , Muscle Fibers, Skeletal/classification , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/chemistry , Muscle, Skeletal/cytology , PAX7 Transcription Factor/analysis , Quadriceps Muscle/anatomy & histology , Quadriceps Muscle/chemistry , Quadriceps Muscle/cytology , Satellite Cells, Skeletal Muscle/ultrastructure , Time Factors , Young Adult
18.
PLoS One ; 15(12): e0243163, 2020.
Article in English | MEDLINE | ID: mdl-33362264

ABSTRACT

Currently available software tools for automated segmentation and analysis of muscle cross-section images often perform poorly in cases of weak or non-uniform staining conditions. To address these issues, our group has developed the MyoSAT (Myofiber Segmentation and Analysis Tool) image-processing pipeline. MyoSAT combines several unconventional approaches including advanced background leveling, Perona-Malik anisotropic diffusion filtering, and Steger's line detection algorithm to aid in pre-processing and enhancement of the muscle image. Final segmentation is based upon marker-based watershed segmentation. Validation tests using collagen V labeled murine and canine muscle tissue demonstrate that MyoSAT can determine mean muscle fiber diameter with an average accuracy of ~92.4%. The software has been tested to work on full muscle cross-sections and works well even under non-optimal staining conditions. The MyoSAT software tool has been implemented as a macro for the freely available ImageJ software platform. This new segmentation tool allows scientists to efficiently analyze large muscle cross-sections for use in research studies and diagnostics.


Subject(s)
Muscle Fibers, Skeletal/ultrastructure , Animals , Automation/methods , Dogs , Female , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy
19.
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
20.
Biotechniques ; 69(5): 388-391, 2020 11.
Article in English | MEDLINE | ID: mdl-33000629

ABSTRACT

Isolated myofibers are commonly used to understand the function of skeletal muscle in vivo. This can involve single isolated myofibers obtained from dissection or from enzymatic dissociation. Isolation via dissection allows control of sarcomere length and preserves tendon attachment but is labor-intensive, time-consuming and yields few viable myofibers. In contrast, enzymatic dissociation is fast and facile, produces hundreds of myofibers, and more importantly reduces the number of muscles/animals needed for studies. Biomechanical properties of the sarcolemma have been studied using myofibers from the extensor digitorum longus, but this has been limited to dissected myofibers, making data collection slow and difficult. We have modified this tool to perform biomechanical measurements of the sarcolemma in dissociated myofibers from the flexor digitorum brevis.


Subject(s)
Cell Culture Techniques/methods , Muscle Fibers, Skeletal/cytology , Sarcolemma/physiology , Animals , Biomechanical Phenomena , Elasticity , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...