Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Physiol ; 600(1): 41-60, 2022 01.
Article in English | MEDLINE | ID: mdl-34761825

ABSTRACT

Injury to skeletal muscle disrupts myofibres and their microvascular supply. While the regeneration of myofibres is well described, little is known of how the microcirculation is affected by skeletal muscle injury or its recovery during regeneration. Nevertheless, the microvasculature must also recover to restore skeletal muscle function. We aimed to define the nature of microvascular damage and time course of repair during muscle injury and regeneration induced by the myotoxin BaCl2 . To test the hypothesis that microvascular disruption occurred secondary to myofibre injury, isolated microvessels were exposed to BaCl2 or the myotoxin was injected into the gluteus maximus (GM) muscle of mice. In isolated microvessels, BaCl2 depolarized smooth muscle cells (SMCs) and endothelial cells while increasing intracellular calcium in SMCs but did not elicit death of either cell type. At 1 day post-injury (dpi) of the GM, capillary fragmentation coincided with myofibre degeneration while arteriolar and venular networks remained intact; neutrophil depletion before injury did not prevent capillary damage. Perfused capillary networks reformed by 5 dpi in association with more terminal arterioles and were dilated through 10 dpi. With no change in microvascular area or branch point number in regenerating capillary networks, fewer capillaries aligned with myofibres and were no longer organized into microvascular units. By 21 dpi, capillary orientation and microvascular unit organization were no longer different from uninjured GM. We conclude that following their disruption secondary to myofibre damage, capillaries regenerate as disorganized networks that remodel into microvascular units as regenerated myofibres mature. KEY POINTS: Skeletal muscle regenerates after injury; however, the nature of microvascular damage and repair is poorly understood. Here, the myotoxin BaCl2 , a standard experimental method of acute skeletal muscle injury, was used to investigate the response of the microcirculation to local injury of intact muscle. Intramuscular injection of BaCl2 induced capillary fragmentation with myofibre degeneration; arteriolar and venular networks remained intact. Direct exposure to BaCl2 did not kill microvascular endothelial cells or smooth muscle cells. Dilated capillary networks reformed by 5 days post-injury (dpi) in association with more terminal arterioles. Capillary orientation remained disorganized through 10 dpi. Capillaries realigned with myofibres and reorganized into microvascular units by 21 dpi, which coincides with the recovery of vasomotor control and maturation of nascent myofibres. Skeletal muscle injury disrupts its capillary supply secondary to myofibre degeneration. Reorganization of regenerating microvascular networks accompanies the recovery of blood flow regulation.


Subject(s)
Capillaries , Endothelial Cells , Animals , Mice , Mice, Inbred C57BL , Microvessels , Muscle, Skeletal , Regeneration
2.
Front Mol Biosci ; 8: 809364, 2021.
Article in English | MEDLINE | ID: mdl-35096972

ABSTRACT

Although intracellular signal transduction is generally represented as a linear process that transmits stimuli from the exterior of a cell to the interior via a transmembrane receptor, interactions with additional membrane-associated proteins are often critical to its success. These molecules play a pivotal role in mediating signaling via the formation of complexes in cis (within the same membrane) with primary effectors, particularly in the context of tumorigenesis. Such secondary effectors may act to promote successful signaling by mediating receptor-ligand binding, recruitment of molecular partners for the formation of multiprotein complexes, or differential signaling outcomes. One signaling family whose contact-mediated activity is frequently modulated by lateral interactions at the cell surface is Eph/ephrin (EphA and EphB receptor tyrosine kinases and their ligands ephrin-As and ephrin-Bs). Through heterotypic interactions in cis, these molecules can promote a diverse range of cellular activities, including some that are mutually exclusive (cell proliferation and cell differentiation, or adhesion and migration). Due to their broad expression in most tissues and their promiscuous binding within and across classes, the cellular response to Eph:ephrin interaction is highly variable between cell types and is dependent on the cellular context in which binding occurs. In this review, we will discuss interactions between molecules in cis at the cell membrane, with emphasis on their role in modulating Eph/ephrin signaling.

3.
Skelet Muscle ; 9(1): 27, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31694693

ABSTRACT

BACKGROUND: Local injection of BaCl2 is an established model of acute injury to study the regeneration of skeletal muscle. However, the mechanism by which BaCl2 causes muscle injury is unresolved. Because Ba2+ inhibits K+ channels, we hypothesized that BaCl2 induces myofiber depolarization leading to Ca2+ overload, proteolysis, and membrane disruption. While BaCl2 spares resident satellite cells, its effect on other tissue components integral to contractile function has not been defined. We therefore asked whether motor nerves and microvessels, which control and supply myofibers, are injured by BaCl2 treatment. METHODS: The intact extensor digitorum longus (EDL) muscle was isolated from male mice (aged 3-4 months) and irrigated with physiological salt solution (PSS) at 37 °C. Myofiber membrane potential (Vm) was recorded using sharp microelectrodes while intracellular calcium concentration ([Ca2+]i) was evaluated with Fura 2 dye. Isometric force production of EDL was measured in situ, proteolytic activity was quantified by calpain degradation of αII-spectrin, and membrane disruption was marked by nuclear staining with propidium iodide (PI). To test for effects on motor nerves and microvessels, tibialis anterior or gluteus maximus muscles were injected with 1.2% BaCl2 (50-75 µL) in vivo followed by immunostaining to evaluate the integrity of respective tissue elements post injury. Data were analyzed using Students t test and analysis of variance with P ≤ 0.05 considered statistically significant. RESULTS: Addition of 1.2% BaCl2 to PSS depolarized myofibers from - 79 ± 3 mV to - 17 ± 7 mV with a corresponding rise in [Ca2+]i; isometric force transiently increased from 7.4 ± 0.1 g to 11.1 ± 0.4 g. Following 1 h of BaCl2 exposure, 92 ± 3% of myonuclei stained with PI (vs. 8 ± 3% in controls) with enhanced cleavage of αII-spectrin. Eliminating Ca2+ from PSS prevented the rise in [Ca2+]i and ameliorated myonuclear staining with PI during BaCl2 exposure. Motor axons and capillary networks appeared fragmented within 24 h following injection of 1.2% BaCl2 and morphological integrity deteriorated through 72 h. CONCLUSIONS: BaCl2 injures myofibers through depolarization of the sarcolemma, causing Ca2+ overload with transient contraction, leading to proteolysis and membrane rupture. Motor innervation and capillarity appear disrupted concomitant with myofiber damage, further compromising muscle integrity.


Subject(s)
Barium Compounds/toxicity , Calcium/metabolism , Chlorides/toxicity , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/injuries , Proteolysis/drug effects , Animals , Disease Models, Animal , In Vitro Techniques , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/drug effects , Microvessels/pathology , Motor Neurons/drug effects , Motor Neurons/pathology , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/physiology , Muscle Proteins/metabolism , Muscle Strength/drug effects , Muscle, Skeletal/blood supply , Muscle, Skeletal/innervation
4.
JCI Insight ; 2(5): e89970, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28289706

ABSTRACT

Spinal muscular atrophy (SMA) is a leading genetic cause of infantile death and is caused by the loss of survival motor neuron-1 (SMN1). Importantly, a nearly identical gene is present called SMN2; however, the majority of SMN2-derived transcripts are alternatively spliced and encode a truncated, dysfunctional protein. Recently, several compounds designed to increase SMN protein have entered clinical trials, including antisense oligonucleotides (ASOs), traditional small molecules, and gene therapy. Expanding beyond SMN-centric therapeutics is important, as it is likely that the breadth of the patient spectrum and the inherent complexity of the disease will be difficult to address with a single therapeutic strategy. Several SMN-independent pathways that could impinge upon the SMA phenotype have been examined with varied success. To identify disease-modifying pathways that could serve as stand-alone therapeutic targets or could be used in combination with an SMN-inducing compound, we investigated adeno-associated virus-mediated (AAV-mediated) gene therapy using plastin-3 (PLS3). Here, we report that AAV9-PLS3 extends survival in an intermediate model of SMA mice as well as in a pharmacologically induced model of SMA using a splice-switching ASO that increases SMN production. PLS3 coadministration improves the phenotype beyond the ASO, demonstrating the potential utility of combinatorial therapeutics in SMA that target SMN-independent and SMN-dependent pathways.


Subject(s)
Membrane Glycoproteins/physiology , Microfilament Proteins/physiology , Muscular Atrophy, Spinal/pathology , Animals , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Microfilament Proteins/genetics , Motor Neurons/physiology , Muscle Fibers, Skeletal/pathology , Survival Analysis , Survival of Motor Neuron 1 Protein/genetics
5.
Genes Brain Behav ; 16(2): 260-270, 2017 02.
Article in English | MEDLINE | ID: mdl-27643807

ABSTRACT

Charcot-Marie-Tooth (CMT) is the most common inherited peripheral neuropathy, affecting approximately 2.8 million people. The CMT leads to distal neuropathy that is characterized by reduced motor nerve conduction velocity, ataxia, muscle atrophy and sensory loss. We generated a mouse model of CMT type 2E (CMT2E) expressing human neurofilament light E396K (hNF-LE396K ), which develops decreased motor nerve conduction velocity, ataxia and muscle atrophy by 4 months of age. Symptomatic hNF-LE396K mice developed phenotypes that were consistent with proprioceptive sensory defects as well as reduced sensitivity to mechanical stimulation, while thermal sensitivity and auditory brainstem responses were unaltered. Progression from presymptomatic to symptomatic included a 50% loss of large diameter sensory axons within the fifth lumbar dorsal root of hNF-LE396K mice. Owing to proprioceptive deficits and loss of large diameter sensory axons, we analyzed muscle spindle morphology in presymptomatic and symptomatic hNF-LE396K and hNF-L control mice. Muscle spindle cross-sectional area and volume were reduced in all hNF-LE396K mice analyzed, suggesting that alterations in muscle spindle morphology occurred prior to the onset of typical CMT pathology. These data suggested that CMT2E pathology initiated in the muscle spindles altering the proprioceptive sensory system. Early sensory pathology in CMT2E could provide a unifying hypothesis for the convergence of pathology observed in CMT.


Subject(s)
Charcot-Marie-Tooth Disease/physiopathology , Muscle Spindles/physiopathology , Animals , Axons/pathology , Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Muscular Atrophy/genetics , Mutation , Neural Conduction/physiology , Neurofilament Proteins/genetics , Sensorimotor Cortex/metabolism , Sensorimotor Cortex/physiopathology
6.
J Cell Biol ; 211(5): 1077-91, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26644518

ABSTRACT

Each adult mammalian skeletal muscle has a unique complement of fast and slow myofibers, reflecting patterns established during development and reinforced via their innervation by fast and slow motor neurons. Existing data support a model of postnatal "matching" whereby predetermined myofiber type identity promotes pruning of inappropriate motor axons, but no molecular mechanism has yet been identified. We present evidence that fiber type-specific repulsive interactions inhibit innervation of slow myofibers by fast motor axons during both postnatal maturation of the neuromuscular junction and myofiber reinnervation after injury. The repulsive guidance ligand ephrin-A3 is expressed only on slow myofibers, whereas its candidate receptor, EphA8, localizes exclusively to fast motor endplates. Adult mice lacking ephrin-A3 have dramatically fewer slow myofibers in fast and mixed muscles, and misexpression of ephrin-A3 on fast myofibers followed by denervation/reinnervation promotes their respecification to a slow phenotype. We therefore conclude that Eph/ephrin interactions guide the fiber type specificity of neuromuscular interactions during development and adult life.


Subject(s)
Muscle, Skeletal/growth & development , Muscle, Skeletal/innervation , Neurogenesis/physiology , Receptor, EphA3/metabolism , Animals , Axons/physiology , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Motor Neurons/physiology , Muscle, Skeletal/embryology , Myofibrils/metabolism , Neuromuscular Junction/physiology , Neuronal Plasticity , Phenotype , Receptor, EphA8/metabolism , Schwann Cells/metabolism , Sciatic Nerve/physiology
7.
Behav Neurosci ; 129(6): 812-21, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26501175

ABSTRACT

The present study explored the role of the amygdala in mediating a unique pattern of feeding behavior driven by intra-accumbens (intra-Acb) opioid activation in the rat. Temporary inactivation of the basolateral amygdala (BLA), via GABAA agonist muscimol administration prevents increased consumption following intra-Acb opioid administration of the selective µ-opioid agonist D-Ala2, NMe-Phe4, Glyol5-enkephalin (DAMGO), yet leaves food approach behaviors intact, particularly after consumption has ended. One interpretation is that inactivation of the BLA selectively blocks neural activity underlying DAMGO-driven consummatory (consumption) but not appetitive (approach) behaviors. The present experiments take advantage of this temporal dissociation of consumption and approach behaviors to investigate their associated neural activity. Following either intra-Acb saline or DAMGO administration, with or without BLA muscimol administration, rats were given 2-hr access to a limited amount of high-fat diet. Immediately following the feeding session, rats were sacrificed and brains assayed for neural activity patterns across critical brain regions known to regulate both appetitive and consummatory feeding behaviors. The results show that intra-Acb DAMGO administration increased c-Fos activation in orexin neurons within the perifornical area of the hypothalamus and that this increase in activation is blocked by BLA muscimol inactivation. Intra-Acb DAMGO administration significantly increased c-Fos activation within dopaminergic neurons of the ventral tegmental area, compared to saline controls, and BLA inactivation had no effect on this increase. Overall, these data provide underlying circuitry that may mediate the selective influence of the BLA on driving consummatory, but not appetitive, feeding behaviors in a model of hedonically driven feeding behavior.


Subject(s)
Analgesics, Opioid/pharmacology , Appetitive Behavior/physiology , Basolateral Nuclear Complex/physiology , Diet, High-Fat , Feeding Behavior/physiology , Nucleus Accumbens/drug effects , Animals , Appetitive Behavior/drug effects , Basolateral Nuclear Complex/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Feeding Behavior/drug effects , GABA-A Receptor Agonists/pharmacology , Hypothalamus/drug effects , Hypothalamus/physiology , Male , Motivation/drug effects , Motivation/physiology , Motor Activity/physiology , Muscimol/pharmacology , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/physiology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
8.
Stem Cells ; 33(10): 3138-51, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26304770

ABSTRACT

The transcription factor Pax7 regulates skeletal muscle stem cell (satellite cells) specification and maintenance through various mechanisms, including repressing the activity of the muscle regulatory factor MyoD. Hence, Pax7-to-MyoD protein ratios can determine maintenance of the committed-undifferentiated state or activation of the differentiation program. Pax7 expression decreases sharply in differentiating myoblasts but is maintained in cells (re)acquiring quiescence, yet the mechanisms regulating Pax7 levels based on differentiation status are not well understood. Here we show that Pax7 levels are directly regulated by the ubiquitin-ligase Nedd4. Our results indicate that Nedd4 is expressed in quiescent and activated satellite cells, that Nedd4 and Pax7 physically interact during early muscle differentiation-correlating with Pax7 ubiquitination and decline-and that Nedd4 loss of function prevented this effect. Furthermore, even transient nuclear accumulation of Nedd4 induced a drop in Pax7 levels and precocious muscle differentiation. Consequently, we propose that Nedd4 functions as a novel Pax7 regulator, which activity is temporally and spatially controlled to modulate the Pax7 protein levels and therefore satellite cell fate.


Subject(s)
Cell Differentiation/genetics , Endosomal Sorting Complexes Required for Transport/biosynthesis , Muscle Development , PAX7 Transcription Factor/biosynthesis , Satellite Cells, Skeletal Muscle/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Animals , Cell Proliferation/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Gene Expression Regulation, Developmental , Humans , Mice , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , MyoD Protein/biosynthesis , Nedd4 Ubiquitin Protein Ligases , PAX7 Transcription Factor/genetics , Proteasome Endopeptidase Complex/genetics , Satellite Cells, Skeletal Muscle/cytology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
Cold Spring Harb Perspect Med ; 4(11): a025650, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25368019

ABSTRACT

Rhabdomyosarcoma (RMS) represents a rare, heterogeneous group of mesodermal malignancies with skeletal muscle differentiation. One major subgroup of RMS tumors (so-called "fusion-positive" tumors) carries exclusive chromosomal translocations that join the DNA-binding domain of the PAX3 or PAX7 gene to the transactivation domain of the FOXO1 (previously known as FKHR) gene. Fusion-negative RMS represents a heterogeneous spectrum of tumors with frequent RAS pathway activation. Overtly metastatic disease at diagnosis is more frequently found in individuals with fusion-positive than in those with fusion-negative tumors. RMS is the most common pediatric soft-tissue sarcoma, and approximately 60% of all children and adolescents diagnosed with RMS are cured by currently available multimodal therapies. However, a curative outcome is achieved in <30% of high-risk individuals with RMS, including all those diagnosed as adults, those diagnosed with fusion-positive tumors during childhood (including metastatic and nonmetastatic tumors), and those diagnosed with metastatic disease during childhood (including fusion-positive and fusion-negative tumors). This white paper outlines current challenges in RMS research and their implications for developing more effective therapies. Urgent clinical problems include local control, systemic disease, need for improved risk stratification, and characterization of differences in disease course in children and adults. Biological challenges include definition of the cellular functions of PAX-FOXO1 fusion proteins, clarification of disease heterogeneity, elucidation of the cellular origins of RMS, delineation of the tumor microenvironment, and identification of means for rational selection and testing of new combination therapies. To streamline future therapeutic developments, it will be critical to improve access to fresh tumor tissue for research purposes, consider alternative trial designs to optimize early clinical testing of candidate drugs, coalesce advocacy efforts to garner public and industry support, and facilitate collaborative efforts between academia and industry.


Subject(s)
Muscle Neoplasms/therapy , Rhabdomyosarcoma/therapy , Adolescent , Adult , Age Factors , Child , Humans , Interinstitutional Relations , Muscle Neoplasms/genetics , Rhabdomyosarcoma/genetics , Young Adult
10.
Am J Physiol Cell Physiol ; 307(2): C140-9, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24898588

ABSTRACT

The twenty-five known matrix metalloproteases (MMPs) and their endogenous inhibitors, tissue inhibitors of metalloproteases (TIMPs), mediate cell invasion through the extracellular matrix (ECM). In a comparative three-dimensional assay, we analyzed human and mouse satellite cells' competence to invade an artificial ECM (collagen I). We identified a single MMP that 1) is expressed by human muscle satellite cells; 2) is induced at the mRNA/protein level by adhesion to collagen I; and 3) is necessary for invasion into a collagen I matrix. Interestingly, murine satellite cells neither express this MMP, nor invade the collagen matrix. However, exogenous human MMP-14 is not sufficient to induce invasion of a collagen matrix by murine cells, emphasizing species differences.


Subject(s)
Collagen/chemistry , Matrix Metalloproteinase 14/metabolism , Satellite Cells, Skeletal Muscle/physiology , Animals , Cell Line , Cell Movement , Gene Expression Regulation, Enzymologic/physiology , Humans , Matrix Metalloproteinase 14/genetics , Mice , Species Specificity , Tissue Scaffolds
11.
J Cell Biol ; 205(1): 97-111, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24711502

ABSTRACT

Wnt7a/Fzd7 signaling stimulates skeletal muscle growth and repair by inducing the symmetric expansion of satellite stem cells through the planar cell polarity pathway and by activating the Akt/mTOR growth pathway in muscle fibers. Here we describe a third level of activity where Wnt7a/Fzd7 increases the polarity and directional migration of mouse satellite cells and human myogenic progenitors through activation of Dvl2 and the small GTPase Rac1. Importantly, these effects can be exploited to potentiate the outcome of myogenic cell transplantation into dystrophic muscles. We observed that a short Wnt7a treatment markedly stimulated tissue dispersal and engraftment, leading to significantly improved muscle function. Moreover, myofibers at distal sites that fused with Wnt7a-treated cells were hypertrophic, suggesting that the transplanted cells deliver activated Wnt7a/Fzd7 signaling complexes to recipient myofibers. Taken together, we describe a viable and effective ex vivo cell modulation process that profoundly enhances the efficacy of stem cell therapy for skeletal muscle.


Subject(s)
Cell Movement , Muscle Strength , Muscle, Skeletal/surgery , Muscular Dystrophies/surgery , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/transplantation , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Fusion , Cell Line , Cell Polarity , Disease Models, Animal , Dishevelled Proteins , Endocytosis , Frizzled Receptors/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hypertrophy , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred mdx , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology , Myoblasts, Skeletal/pathology , Neuropeptides/metabolism , PAX7 Transcription Factor/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Wnt Proteins/genetics , rac1 GTP-Binding Protein/metabolism , Red Fluorescent Protein
12.
FEBS J ; 280(17): 4089-99, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23374506

ABSTRACT

Mammalian skeletal muscle is notable for both its highly ordered biophysical structure and its regenerative capacity following trauma. Critical to both of these features is the specialized muscle extracellular matrix, comprising both the multiple concentric sheaths of connective tissue surrounding structural units from single myofibers to whole muscles and the dense interstitial matrix that occupies the space between them. Extracellular matrix-dependent interactions affect all activities of the resident muscle stem cell population (the satellite cells), from maintenance of quiescence and stem cell potential to the regulation of proliferation and differentiation. This review focuses on the role of the extracellular matrix in muscle regeneration, with a particular emphasis on regulation of satellite-cell activity.


Subject(s)
Extracellular Matrix/metabolism , Muscle Development/physiology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Humans , Satellite Cells, Skeletal Muscle/physiology , Signal Transduction , Stem Cells/physiology
13.
Development ; 138(24): 5279-89, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22071104

ABSTRACT

During development and regeneration, directed migration of cells, including neural crest cells, endothelial cells, axonal growth cones and many types of adult stem cells, to specific areas distant from their origin is necessary for their function. We have recently shown that adult skeletal muscle stem cells (satellite cells), once activated by isolation or injury, are a highly motile population with the potential to respond to multiple guidance cues, based on their expression of classical guidance receptors. We show here that, in vivo, differentiated and regenerating myofibers dynamically express a subset of ephrin guidance ligands, as well as Eph receptors. This expression has previously only been examined in the context of muscle-nerve interactions; however, we propose that it might also play a role in satellite cell-mediated muscle repair. Therefore, we investigated whether Eph-ephrin signaling would produce changes in satellite cell directional motility. Using a classical ephrin 'stripe' assay, we found that satellite cells respond to a subset of ephrins with repulsive behavior in vitro; patterning of differentiating myotubes is also parallel to ephrin stripes. This behavior can be replicated in a heterologous in vivo system, the hindbrain of the developing quail, in which neural crest cells are directed in streams to the branchial arches and to the forelimb of the developing quail, where presumptive limb myoblasts emigrate from the somite. We hypothesize that guidance signaling might impact multiple steps in muscle regeneration, including escape from the niche, directed migration to sites of injury, cell-cell interactions among satellite cell progeny, and differentiation and patterning of regenerated muscle.


Subject(s)
Body Patterning/physiology , Cell Movement/physiology , Ephrins/physiology , Receptors, Eph Family/physiology , Satellite Cells, Skeletal Muscle/physiology , Animals , Branchial Region/growth & development , Cells, Cultured , Ephrins/metabolism , Female , Mice , Mice, Inbred CBA , Muscle Development , Neural Crest/growth & development , Quail/growth & development , Quail/metabolism , Receptors, Eph Family/metabolism , Rhombencephalon/growth & development
14.
Stem Cells ; 29(12): 2030-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21997891

ABSTRACT

Expression of the cell surface sialomucin CD34 is common to many adult stem cell types, including muscle satellite cells. However, no clear stem cell or regeneration-related phenotype has ever been reported in mice lacking CD34, and its function on these cells remains poorly understood. Here, we assess the functional role of CD34 on satellite cell-mediated muscle regeneration. We show that Cd34(-/-) mice, which have no obvious developmental phenotype, display a defect in muscle regeneration when challenged with either acute or chronic muscle injury. This regenerative defect is caused by impaired entry into proliferation and delayed myogenic progression. Consistent with the reported antiadhesive function of CD34, knockout satellite cells also show decreased motility along their host myofiber. Altogether, our results identify a role for CD34 in the poorly understood early steps of satellite cell activation and provide the first evidence that beyond being a stem cell marker, CD34 may play an important function in modulating stem cell activity.


Subject(s)
Antigens, CD34/metabolism , Cell Movement , Cell Proliferation , Muscle, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Animals , Antigens, CD34/genetics , Elapid Venoms/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Point Mutation , Satellite Cells, Skeletal Muscle/physiology , Time-Lapse Imaging
15.
Skelet Muscle ; 1(1): 7, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21798086

ABSTRACT

BACKGROUND: As the resident stem cells of skeletal muscle, satellite cells are activated by extracellular cues associated with local damage. Once activated, satellite cells will re-enter the cell cycle to proliferate and supply a population of myoblasts, which will repair or replace damaged myofibers by differentiating and fusing either with an existing myofiber or with each other. There is also evidence that the orientation of cell division with respect to the myofiber may indicate or convey asymmetry in the two daughter cells. Our recent studies with time-lapse imaging of myofiber-associated satellite cells in vitro have yielded new data on the timing and orientation of satellite cell divisions, and revealed persistent differences in the behavior of daughter cells from planar versus vertical divisions. RESULTS: We analyzed 244 individual fiber-associated satellite cells in time-lapse video from 24 to 48 hours after myofiber harvest. We found that initial cell division in fiber culture is not synchronous, although presumably all cells were activated by the initial trauma of harvest; that cell cycling time is significantly shorter than previously thought (as short as 4.8 hours, averaging 10 hours between the first and second divisions and eight hours between the second and third); and that timing of subsequent divisions is not strongly correlated with timing of the initial division. Approximately 65% of first and 80% of second cell divisions occur parallel to the axis of the myofiber, whereas the remainder occur outside the plane of the fiber surface (vertical division). We previously demonstrated that daughter cells frequently remain associated with each other after division or reassociate after a brief separation, and that unrelated cells may also associate for significant periods of time. We show in this paper that daughter cells resulting from a vertical division remain associated with one another several times longer than do daughters from a horizontal division. However, the total average time of association between sister cells is not significantly different from the total average time of association between unrelated cells. CONCLUSIONS: These longitudinal characterizations of satellite cell behavior shortly after activation provide new insights into cell proliferation and association as a function of relatedness, and indicate significant and consistent heterogeneity within the population based on these metrics.

16.
Neuromuscul Disord ; 21(4): 282-90, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21277207

ABSTRACT

Duchenne muscular dystrophy is a neuromuscular degenerative disorder caused by the absence of dystrophin protein. It is characterized by progressive muscle weakness and cycles of degeneration/regeneration accompanying chronic muscle damage and repair. Canine models of muscular dystrophy, including the dystrophin-deficient golden retriever muscular dystrophy (GRMD), are the most promising animal models for evaluation of potential therapies, however canine-specific molecular tools are limited. In particular, few immune reagents for extracellular epitopes marking canine satellite cells (muscle stem cells) are available. We generated an antibody to the satellite cell marker syndecan-4 that identifies canine satellite cells. We then characterized isolated satellite cells from GRMD muscle and wildtype muscle by several in vitro metrics, and surprisingly found no significant differences between the two populations. We discuss whether accumulated adverse changes in the muscle environment rather than cell-intrinsic defects may be implicated in the eventual failure of satellite cell efficacy in vivo.


Subject(s)
Muscular Dystrophy, Animal/pathology , Satellite Cells, Skeletal Muscle/pathology , Amino Acid Sequence , Animals , Blotting, Western , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation , Cloning, Molecular , Dogs , Flow Cytometry , Immunohistochemistry , Molecular Sequence Data , Syndecan-4/genetics , Syndecan-4/immunology
17.
J Cell Biol ; 190(3): 427-41, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20696709

ABSTRACT

Skeletal muscle postnatal growth and repair depend on satellite cells and are regulated by molecular signals within the satellite cell niche. We investigated the molecular and cellular events that lead to altered myogenesis upon genetic ablation of Syndecan-3, a component of the satellite cell niche. In the absence of Syndecan-3, satellite cells stall in S phase, leading to reduced proliferation, increased cell death, delayed onset of differentiation, and markedly reduced numbers of Pax7(+) satellite cells accompanied by myofiber hypertrophy and an increased number of centrally nucleated myofibers. We show that the aberrant cell cycle and impaired self-renewal of explanted Syndecan-3-null satellite cells are rescued by ectopic expression of the constitutively active Notch intracellular domain. Furthermore, we show that Syndecan-3 interacts with Notch and is required for Notch processing by ADAM17/tumor necrosis factor-alpha-converting enzyme (TACE) and signal transduction. Together, our data support the conclusion that Syndecan-3 and Notch cooperate in regulating homeostasis of the satellite cell population and myofiber size.


Subject(s)
Muscle Development , Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Syndecan-3/metabolism , Animals , Cell Cycle , Cell Differentiation , Cell Membrane/enzymology , Cell Membrane/metabolism , Cell Proliferation , Cells, Cultured , Mice , Mice, Inbred Strains , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction , Syndecan-3/deficiency , Syndecan-3/genetics
18.
Stem Cells ; 27(10): 2527-38, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19609936

ABSTRACT

Skeletal muscle repair and regeneration requires the activity of satellite cells, a population of myogenic stem cells scattered throughout the tissue and activated to proliferate and differentiate in response to myotrauma or disease. While it seems likely that satellite cells would need to navigate local muscle tissue to reach damaged areas, relatively little data on such motility exist, and most studies have been with immortalized cell lines. We find that primary satellite cells are significantly more motile than myoblast cell lines, and that adhesion to laminin promotes primary cell motility more than fourfold over other substrates. Using timelapse videomicroscopy to assess satellite cell motility on single living myofibers, we have identified a requirement for the laminin-binding integrin alpha 7 beta 1 in satellite cell motility, as well as a role for hepatocyte growth factor in promoting directional persistence. The extensive migratory behavior of satellite cells resident on muscle fibers suggests caution when determining, based on fixed specimens, whether adjacent cells are daughters from the same mother cell. We also observed more persistent long-term contact between individual satellite cells than has been previously supposed, potential cell-cell attractive and repulsive interactions, and migration between host myofibers. Based on such activity, we assayed for expression of "pathfinding" cues, and found that satellite cells express multiple guidance ligands and receptors. Together, these data suggest that satellite cell migration in vivo may be more extensive than currently thought, and could be regulated by combinations of signals, including adhesive haptotaxis, soluble factors, and guidance cues.


Subject(s)
Cell Movement/physiology , Imaging, Three-Dimensional/methods , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Animals , Antigens, CD/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/physiology , Cell Lineage/physiology , Cells, Cultured , Chemotaxis/physiology , Cues , Female , Hepatocyte Growth Factor/metabolism , Integrin alpha Chains/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Laminin/metabolism , Laminin/pharmacology , Mice , Microscopy, Video/methods , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Receptors, Growth Factor/drug effects , Receptors, Growth Factor/metabolism , Satellite Cells, Skeletal Muscle/metabolism
19.
Cell Stem Cell ; 4(3): 217-25, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19265661

ABSTRACT

Skeletal muscle satellite cells, located between the basal lamina and plasma membrane of myofibers, are required for skeletal muscle regeneration. The capacity of satellite cells as well as other cell lineages including mesoangioblasts, mesenchymal stem cells, and side population (SP) cells to contribute to muscle regeneration has complicated the identification of a satellite stem cell. We have characterized a rare subset of the muscle SP that efficiently engrafts into the host satellite cell niche when transplanted into regenerating muscle, providing 75% of the satellite cell population and 30% of the myonuclear population, respectively. These cells are found in the satellite cell position, adhere to isolated myofibers, and spontaneously undergo myogenesis in culture. We propose that this subset of SP cells (satellite-SP cells), characterized by ABCG2, Syndecan-4, and Pax7 expression, constitutes a self-renewing muscle stem cell capable of generating both satellite cells and their myonuclear progeny in vivo.


Subject(s)
Muscle, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/physiology , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Niche/physiology , Syndecan-4/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Animals , Female , Mice , Mice, Inbred C57BL , Muscle Development , PAX7 Transcription Factor/biosynthesis , Satellite Cells, Skeletal Muscle/metabolism , Syndecan-3/biosynthesis
20.
J Cell Biochem ; 105(3): 663-9, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18759329

ABSTRACT

Skeletal muscle is formed during development by the progressive specification, proliferation, migration, and fusion of myoblasts to form terminally differentiated, contractile, highly patterned myofibers. Skeletal muscle is repaired or replaced postnatally by a similar process, involving a resident myogenic stem cell population referred to as satellite cells. In both cases, the activity of the myogenic precursor cells in question is regulated by local signals from the environment, frequently involving other, non-muscle cell types. However, while the majority of studies on muscle development were done in the context of the whole embryo, much of the current work on muscle satellite cells has been done in vitro, or on satellite cell-derived cell lines. While significant practical reasons for these approaches exist, it is almost certain that important influences from the context of the injured and regenerating muscle are lost, while potential tissue culture artifacts are introduced. This review will briefly address extracellular influences on satellite cells in vivo and in vitro that would be expected to impinge on their activity.


Subject(s)
Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...