Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Eur J Cell Biol ; 103(2): 151409, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38579603

ABSTRACT

Neuromuscular junctions transmit signals from the nervous system to skeletal muscles, triggering their contraction, and their proper organization is essential for breathing and voluntary movements. αDystrobrevin-1 is a cytoplasmic component of the dystrophin-glycoprotein complex and has pivotal functions in regulating the integrity of muscle fibers and neuromuscular junctions. Previous studies identified that αDystrobrevin-1 functions in the organization of the neuromuscular junction and that its phosphorylation in the C-terminus is required in this process. Our proteomic screen identified several putative αDystrobrevin-1 interactors recruited to the Y730 site in phosphorylated and unphosphorylated states. Amongst various actin-modulating proteins, we identified the Arp2/3 complex regulator cortactin. We showed that similarly to αDystrobrevin-1, cortactin is strongly enriched at the neuromuscular postsynaptic machinery and obtained results suggesting that these two proteins interact in cell homogenates and at the neuromuscular junctions. Analysis of synaptic morphology in cortactin knockout mice showed abnormalities in the slow-twitching soleus muscle and not in the fast-twitching tibialis anterior. However, muscle strength examination did not reveal apparent deficits in knockout animals.


Subject(s)
Cortactin , Dystrophin-Associated Proteins , Mice, Knockout , Neuromuscular Junction , Animals , Neuromuscular Junction/metabolism , Cortactin/metabolism , Cortactin/genetics , Mice , Dystrophin-Associated Proteins/metabolism , Dystrophin-Associated Proteins/genetics , Muscle, Skeletal/metabolism , Humans , Phosphorylation
2.
Nucleic Acids Res ; 52(2): 600-624, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38048326

ABSTRACT

We examined YAP1/TAZ-TEAD signaling pathway activity at neuromuscular junctions (NMJs) of skeletal muscle fibers in adult mice. Our investigations revealed that muscle-specific knockouts of Yap1 or Taz, or both, demonstrate that these transcriptional coactivators regulate synaptic gene expression, the number and morphology of NMJs, and synaptic nuclei. Yap1 or Taz single knockout mice display reduced grip strength, fragmentation of NMJs, and accumulation of synaptic nuclei. Yap1/Taz muscle-specific double knockout mice do not survive beyond birth and possess almost no NMJs, the few detectable show severely impaired morphology and are organized in widened endplate bands; and with motor nerve endings being mostly absent. Myogenic gene expression is significantly impaired in the denervated muscles of knockout mice. We found that Tead1 and Tead4 transcription rates were increased upon incubation of control primary myotubes with AGRN-conditioned medium. Reduced AGRN-dependent acetylcholine receptor clustering and synaptic gene transcription were observed in differentiated primary Tead1 and Tead4 knockout myotubes. In silico analysis of previously reported genomic occupancy sites of TEAD1/4 revealed evolutionary conserved regions of potential TEAD binding motifs in key synaptic genes, the relevance of which was functionally confirmed by reporter assays. Collectively, our data suggest a role for YAP1/TAZ-TEAD1/TEAD4 signaling, particularly through TAZ-TEAD4, in regulating synaptic gene expression and acetylcholine receptor clustering at NMJs.


Subject(s)
Gene Regulatory Networks , Transcription Factors , Mice , Animals , Transcription Factors/genetics , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Muscle Fibers, Skeletal/metabolism , Neuromuscular Junction/metabolism , Mice, Knockout , Gene Expression , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Muscle, Skeletal/metabolism
3.
Sci Adv ; 9(24): eadg0183, 2023 06 16.
Article in English | MEDLINE | ID: mdl-37327338

ABSTRACT

Junctional folds are unique membrane specializations developed progressively during the postnatal maturation of vertebrate neuromuscular junctions (NMJs), but how they are formed remains elusive. Previous studies suggested that topologically complex acetylcholine receptor (AChR) clusters in muscle cultures undergo a series of transformations, resembling the postnatal maturation of NMJs in vivo. We first demonstrated the presence of membrane infoldings at AChR clusters in cultured muscles. Live-cell super-resolution imaging further revealed that AChRs are gradually redistributed to the crest regions and spatially segregated from acetylcholinesterase along the elongating membrane infoldings over time. Mechanistically, lipid raft disruption or caveolin-3 knockdown not only inhibits membrane infolding formation at aneural AChR clusters and delays agrin-induced AChR clustering in vitro but also affects junctional fold development at NMJs in vivo. Collectively, this study demonstrated the progressive development of membrane infoldings via nerve-independent, caveolin-3-dependent mechanisms and identified their roles in AChR trafficking and redistribution during the structural maturation of NMJs.


Subject(s)
Acetylcholinesterase , Caveolin 3 , Caveolin 3/genetics , Neuromuscular Junction/physiology , Receptors, Cholinergic , Muscles
4.
Int J Mol Sci ; 24(3)2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36768425

ABSTRACT

The complexity of the cellular proteome facilitates the control of a wide range of cellular processes. Non-coding RNAs, including microRNAs and long non-coding RNAs, greatly contribute to the repertoire of tools used by cells to orchestrate various functions. Circular RNAs (circRNAs) constitute a specific class of non-coding RNAs that have recently emerged as a widely generated class of molecules produced from many eukaryotic genes that play essential roles in regulating cellular processes in health and disease. This review summarizes current knowledge about circRNAs and focuses on the functions of AMOTL1 circRNAs and AMOTL1 protein. Both products from the AMOTL1 gene have well-known functions in physiology, cancer, and other disorders. Using AMOTL1 as an example, we illustrate how focusing on both circRNAs and proteins produced from the same gene contributes to a better understanding of gene functions.


Subject(s)
MicroRNAs , RNA, Long Noncoding , RNA/genetics , RNA/metabolism , RNA, Circular/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics
5.
Front Cell Dev Biol ; 10: 838612, 2022.
Article in English | MEDLINE | ID: mdl-35372356

ABSTRACT

Lynx1 is a glycosylphosphatidylinositol (GPI)-linked protein shown to affect synaptic plasticity through modulation of nicotinic acetylcholine receptor (nAChR) subtypes in the brain. Because of this function and structural similarity to α-bungarotoxin, which binds muscle-specific nAChRs with high affinity, Lynx1 is a promising candidate for modulating nAChRs in skeletal muscles. However, little is known about the expression and roles of Lynx1 in skeletal muscles and neuromuscular junctions (NMJs). Here, we show that Lynx1 is expressed in skeletal muscles, increases during development, and concentrates at NMJs. We also demonstrate that Lynx1 interacts with muscle-specific nAChR subunits. Additionally, we present data indicating that Lynx1 deletion alters the response of skeletal muscles to cholinergic transmission and their contractile properties. Based on these findings, we asked if Lynx1 deletion affects developing and adult NMJs. Loss of Lynx1 had no effect on NMJs at postnatal day 9 (P9) and moderately increased their size at P21. Thus, Lynx1 plays a minor role in the structural development of NMJs. In 7- and 12-month-old mice lacking Lynx1, there is a marked increase in the incidence of NMJs with age- and disease-associated morphological alterations. The loss of Lynx1 also reduced the size of adult muscle fibers. Despite these effects, Lynx1 deletion did not alter the rate of NMJ reinnervation and stability following motor axon injury. These findings suggest that Lynx1 is not required during fast remodeling of the NMJ, as is the case during reformation following crushing of motor axons and development. Instead, these data indicate that the primary role of Lynx1 may be to maintain the structure and function of adult and aging NMJs.

6.
Int J Mol Sci ; 22(17)2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34502296

ABSTRACT

Proper muscle function depends on the neuromuscular junctions (NMJs), which mature postnatally to complex "pretzel-like" structures, allowing for effective synaptic transmission. Postsynaptic acetylcholine receptors (AChRs) at NMJs are anchored in the actin cytoskeleton and clustered by the scaffold protein rapsyn, recruiting various actin-organizing proteins. Mechanisms driving the maturation of the postsynaptic machinery and regulating rapsyn interactions with the cytoskeleton are still poorly understood. Drebrin is an actin and microtubule cross-linker essential for the functioning of the synapses in the brain, but its role at NMJs remains elusive. We used immunohistochemistry, RNA interference, drebrin inhibitor 3,5-bis-trifluoromethyl pyrazole (BTP2) and co-immunopreciptation to explore the role of this protein at the postsynaptic machinery. We identify drebrin as a postsynaptic protein colocalizing with the AChRs both in vitro and in vivo. We also show that drebrin is enriched at synaptic podosomes. Downregulation of drebrin or blocking its interaction with actin in cultured myotubes impairs the organization of AChR clusters and the cluster-associated microtubule network. Finally, we demonstrate that drebrin interacts with rapsyn and a drebrin interactor, plus-end-tracking protein EB3. Our results reveal an interplay between drebrin and cluster-stabilizing machinery involving rapsyn, actin cytoskeleton, and microtubules.


Subject(s)
Acetylcholine/metabolism , Microtubules/physiology , Myoblasts/physiology , Neuromuscular Junction/physiology , Neuropeptides/pharmacology , Receptors, Cholinergic/metabolism , Synapses/physiology , Actin Cytoskeleton/metabolism , Animals , Cells, Cultured , Mice , Microtubules/drug effects , Myoblasts/cytology , Myoblasts/drug effects , Neuromuscular Junction/drug effects , Receptors, Cholinergic/genetics , Synaptic Transmission
7.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228026

ABSTRACT

Caveolae are the cholesterol-rich small invaginations of the plasma membrane present in many cell types including adipocytes, endothelial cells, epithelial cells, fibroblasts, smooth muscles, skeletal muscles and cardiac muscles. They serve as specialized platforms for many signaling molecules and regulate important cellular processes like energy metabolism, lipid metabolism, mitochondria homeostasis, and mechano-transduction. Caveolae can be internalized together with associated cargo. The caveolae-dependent endocytic pathway plays a role in the withdrawal of many plasma membrane components that can be sent for degradation or recycled back to the cell surface. Caveolae are formed by oligomerization of caveolin proteins. Caveolin-3 is a muscle-specific isoform, whose malfunction is associated with several diseases including diabetes, cancer, atherosclerosis, and cardiovascular diseases. Mutations in Caveolin-3 are known to cause muscular dystrophies that are collectively called caveolinopathies. Altered expression of Caveolin-3 is also observed in Duchenne's muscular dystrophy, which is likely a part of the pathological process leading to muscle weakness. This review summarizes the major functions of Caveolin-3 in skeletal muscles and discusses its involvement in the pathology of muscular dystrophies.


Subject(s)
Arrhythmias, Cardiac/genetics , Cardiomegaly/genetics , Caveolin 3/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Neuromuscular Junction/genetics , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Caveolae/metabolism , Caveolin 3/chemistry , Caveolin 3/metabolism , Dystrophin/genetics , Dystrophin/metabolism , Endocytosis , Gene Expression Regulation , Humans , Mechanotransduction, Cellular , Mice , Mice, Inbred mdx , Muscle, Skeletal/physiopathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/physiopathology , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Neuromuscular Junction/physiopathology , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism
8.
Stem Cell Res Ther ; 11(1): 238, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32552916

ABSTRACT

BACKGROUND: Pluripotent stem cells present the ability to self-renew and undergo differentiation into any cell type building an organism. Importantly, a lot of evidence on embryonic stem cell (ESC) differentiation comes from in vitro studies. However, ESCs cultured in vitro do not necessarily behave as cells differentiating in vivo. For this reason, we used teratomas to study early and advanced stages of in vivo ESC myogenic differentiation and the role of Pax7 in this process. Pax7 transcription factor plays a crucial role in the formation and differentiation of skeletal muscle precursor cells during embryonic development. It controls the expression of other myogenic regulators and also acts as an anti-apoptotic factor. It is also involved in the formation and maintenance of satellite cell population. METHODS: In vivo approach we used involved generation and analysis of pluripotent stem cell-derived teratomas. Such model allows to analyze early and also terminal stages of tissue differentiation, for example, terminal stages of myogenesis, including the formation of innervated and vascularized mature myofibers. RESULTS: We determined how the lack of Pax7 function affects the generation of different myofiber types. In Pax7-/- teratomas, the skeletal muscle tissue occupied significantly smaller area, as compared to Pax7+/+ ones. The proportion of myofibers expressing Myh3 and Myh2b did not differ between Pax7+/+ and Pax7-/- teratomas. However, the area of Myh7 and Myh2a myofibers was significantly lower in Pax7-/- ones. Molecular characteristic of skeletal muscles revealed that the levels of mRNAs coding Myh isoforms were significantly lower in Pax7-/- teratomas. The level of mRNAs encoding Pax3 was significantly higher, while the expression of Nfix, Eno3, Mck, Mef2a, and Itga7 was significantly lower in Pax7-/- teratomas, as compared to Pax7+/+ ones. We proved that the number of satellite cells in Pax7-/- teratomas was significantly reduced. Finally, analysis of neuromuscular junction localization in samples prepared with the iDISCO method confirmed that the organization of neuromuscular junctions in Pax7-/- teratomas was impaired. CONCLUSIONS: Pax7-/- ESCs differentiate in vivo to embryonic myoblasts more readily than Pax7+/+ cells. In the absence of functional Pax7, initiation of myogenic differentiation is facilitated, and as a result, the expression of mesoderm embryonic myoblast markers is upregulated. However, in the absence of functional Pax7 neuromuscular junctions, formation is abnormal, what results in lower differentiation potential of Pax7-/- ESCs during advanced stages of myogenesis.


Subject(s)
Satellite Cells, Skeletal Muscle , Teratoma , Animals , Cell Differentiation , Mice , Mouse Embryonic Stem Cells , Muscle Development/genetics , Muscle, Skeletal , NFI Transcription Factors , PAX7 Transcription Factor/genetics , Teratoma/genetics
9.
Front Mol Neurosci ; 13: 104, 2020.
Article in English | MEDLINE | ID: mdl-32587503

ABSTRACT

The neuromuscular junctions (NMJs) connect muscle fibers with motor neurons and enable the coordinated contraction of skeletal muscles. The dystrophin-associated glycoprotein complex (DGC) is an essential component of the postsynaptic machinery of the NMJ and is important for the maintenance of NMJ structural integrity. To identify novel proteins that are important for NMJ organization, we performed a mass spectrometry-based screen for interactors of α-dystrobrevin 1 (aDB1), one of the components of the DGC. The guanidine nucleotide exchange factor (GEF) Arhgef5 was found to be one of the aDB1 binding partners that is recruited to Tyr-713 in a phospho-dependent manner. We show here that Arhgef5 localizes to the NMJ and that its genetic depletion in the muscle causes the fragmentation of the synapses in conditional knockout mice. Arhgef5 loss in vivo is associated with a reduction in the levels of active GTP-bound RhoA and Cdc42 GTPases, highlighting the importance of actin dynamics regulation for the maintenance of NMJ integrity.

10.
Semin Cell Dev Biol ; 104: 31-38, 2020 08.
Article in English | MEDLINE | ID: mdl-32147379

ABSTRACT

Neuromuscular junctions (NMJs) are specialized synapses in the peripheral nervous system that allow the transmission of neuronal impulses to skeletal muscles for their contraction. Due to its size and accessibility, the NMJ is a commonly used model for studying basic principles of synapse organization and function. Similar to synapses in the central nervous system, NMJs are composed of presynaptic axonal terminals, the postsynaptic machinery formed at the membrane of the muscle fibers, and the synapse-associated glial cells. The special glial cells at the NMJs are called terminal Schwann cells or perisynaptic Schwann cells (PSCs). Decades of studies on the NMJ, as well as the most recent discoveries, have revealed multiple functions for PSCs at different stages of synaptic formation, maintenance, and disassembly. This review summarizes major observations in the field.


Subject(s)
Neuromuscular Junction/metabolism , Presynaptic Terminals/metabolism , Schwann Cells/metabolism , Animals , Models, Biological , Neuromuscular Junction/cytology , Schwann Cells/cytology
11.
Sci Rep ; 10(1): 4524, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32161296

ABSTRACT

Motor neurons form specialized synapses with skeletal muscle fibers, called neuromuscular junctions (NMJs). Cultured myotubes are used as a simplified in vitro system to study the postsynaptic specialization of muscles. The stimulation of myotubes with the glycoprotein agrin or laminin-111 induces the clustering of postsynaptic machinery that contains acetylcholine receptors (AChRs). When myotubes are grown on laminin-coated surfaces, AChR clusters undergo developmental remodeling to form topologically complex structures that resemble mature NMJs. Needing further exploration are the molecular processes that govern AChR cluster assembly and its developmental maturation. Here, we describe an improved protocol for culturing muscle cells to promote the formation of complex AChR clusters. We screened various laminin isoforms and showed that laminin-221 was the most potent for inducing AChR clusters, whereas laminin-121, laminin-211, and laminin-221 afforded the highest percentages of topologically complex assemblies. Human primary myotubes that were formed by myoblasts obtained from patient biopsies also assembled AChR clusters that underwent remodeling in vitro. Collectively, these results demonstrate an advancement of culturing myotubes that can facilitate high-throughput screening for potential therapeutic targets for neuromuscular disorders.


Subject(s)
Cell Culture Techniques , Laminin , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Post-Synaptic Density , Animals , Cell Line , Cells, Cultured , Fluorescent Antibody Technique , Laminin/chemistry , Mice , Models, Biological , Myoblasts/cytology , Myoblasts/physiology , Neuromuscular Junction , Receptors, Cholinergic/metabolism
12.
PLoS Biol ; 17(5): e3000253, 2019 05.
Article in English | MEDLINE | ID: mdl-31042703

ABSTRACT

The angiomotin (Amot)-Yes-associated protein 1 (Yap1) complex plays a major role in regulating the inhibition of cell contact, cellular polarity, and cell growth in many cell types. However, the function of Amot and the Hippo pathway transcription coactivator Yap1 in the central nervous system remains unclear. We found that Amot is a critical mediator of dendritic morphogenesis in cultured hippocampal cells and Purkinje cells in the brain. Amot function in developing neurons depends on interactions with Yap1, which is also indispensable for dendrite growth and arborization in vitro. The conditional deletion of Amot and Yap1 in neurons led to a decrease in the complexity of Purkinje cell dendritic trees, abnormal cerebellar morphology, and impairments in motor coordination. Our results indicate that the function of Amot and Yap1 in dendrite growth does not rely on interactions with TEA domain (TEAD) transcription factors or the expression of Hippo pathway-dependent genes. Instead, Amot and Yap1 regulate dendrite development by affecting the phosphorylation of S6 kinase and its target S6 ribosomal protein.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Dendrites/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Locomotion/physiology , Microfilament Proteins/metabolism , Angiomotins , Animals , Hippocampus/cytology , Integrases/metabolism , Mice, Inbred C57BL , Morphogenesis , Motor Activity , Phosphorylation , Protein Binding , Purkinje Cells/metabolism , Rats, Wistar , Ribosomal Protein S6/metabolism , YAP-Signaling Proteins
13.
Proc Natl Acad Sci U S A ; 116(17): 8397-8402, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30962377

ABSTRACT

Actin filaments (F-actin) are key components of sarcomeres, the basic contractile units of skeletal muscle myofibrils. A crucial step during myofibril differentiation is the sequential exchange of α-actin isoforms from smooth muscle (α-SMA) and cardiac (α-CAA) to skeletal muscle α-actin (α-SKA) that, in mice, occurs during early postnatal life. This "α-actin switch" requires the coordinated activity of actin regulators because it is vital that sarcomere structure and function are maintained during differentiation. The molecular machinery that controls the α-actin switch, however, remains enigmatic. Cyclase-associated proteins (CAP) are a family of actin regulators with largely unknown physiological functions. We here report a function for CAP2 in regulating the α-actin exchange during myofibril differentiation. This α-actin switch was delayed in systemic CAP2 mutant mice, and myofibrils remained in an undifferentiated stage at the onset of the often excessive voluntary movements in postnatal mice. The delay in the α-actin switch coincided with the onset of motor function deficits and histopathological changes including a high frequency of type IIB ring fibers. Our data suggest that subtle disturbances of postnatal F-actin remodeling are sufficient for predisposing muscle fibers to form ring fibers. Cofilin2, a putative CAP2 interaction partner, has been recently implicated in myofibril actin cytoskeleton differentiation, and the myopathies in cofilin2 and CAP2 mutant mice showed striking similarities. We therefore propose a model in which CAP2 and cofilin2 cooperate in actin regulation during myofibril differentiation.


Subject(s)
Actin Cytoskeleton/physiology , Carrier Proteins , Cell Differentiation , Muscle, Skeletal , Myofibrils/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Knockout , Muscle Development/genetics , Muscle Development/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
Ann N Y Acad Sci ; 1412(1): 62-72, 2018 01.
Article in English | MEDLINE | ID: mdl-29068540

ABSTRACT

The proper function of skeletal muscles relies on their ability to process signals derived from motor neurons, transmit stimuli along the muscle fibers, contract, and regenerate efficiently after injury. The dystrophin-glycoprotein complex (DGC; also called the dystrophin-associated protein complex) plays a central role in all of these processes. It acts as a transmembrane platform that anchors the extracellular matrix (ECM) to the intracellular cytoskeleton and makes muscle fibers more resistant to injury. The DGC also contributes to the transmission of contraction-evoked force from the sarcomere to the ECM. The dysfunction of DGC-associated proteins can lead to myopathies, including Duchenne's muscular dystrophy, manifested by progressive muscle damage and impairments in regeneration. The DGC also plays a pivotal role in the organization of neuromuscular junctions (NMJs), where it stabilizes postsynaptic machinery, including receptors for the neurotransmitter acetylcholine (AChRs). Here, we focus on the role of the DGC complex in NMJ and skeletal muscle physiology and discuss the novel components that are associated with the complex.


Subject(s)
Dystrophin/metabolism , Glycoproteins/metabolism , Neuromuscular Junction/metabolism , Animals , Dystrophin-Associated Proteins/metabolism , Humans , Models, Neurological , Multiprotein Complexes/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/etiology , Muscular Dystrophy, Duchenne/metabolism , Receptors, Cholinergic/metabolism
15.
Sci Rep ; 7(1): 9116, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831123

ABSTRACT

Neuromuscular junctions (NMJs) are specialized synapses that connect motor neurons to skeletal muscle fibers and orchestrate proper signal transmission from the nervous system to muscles. The efficient formation and maintenance of the postsynaptic machinery that contains acetylcholine receptors (AChR) are indispensable for proper NMJ function. Abnormalities in the organization of synaptic components often cause severe neuromuscular disorders, such as muscular dystrophy. The dystrophin-associated glycoprotein complex (DGC) was shown to play an important role in NMJ development. We recently identified liprin-α-1 as a novel binding partner for one of the cytoplasmic DGC components, α-dystrobrevin-1. In the present study, we performed a detailed analysis of localization and function of liprin-α-1 at the murine NMJ. We showed that liprin-α-1 localizes to both pre- and postsynaptic compartments at the NMJ, and its synaptic enrichment depends on the presence of the nerve. Using cultured muscle cells, we found that liprin-α-1 plays an important role in AChR clustering and the organization of cortical microtubules. Our studies provide novel insights into the function of liprin-α-1 at vertebrate neuromuscular synapses.


Subject(s)
Neuromuscular Junction/metabolism , Proteins/metabolism , Synaptic Transmission , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Gene Expression , Mice , Microtubules/metabolism , Muscles/metabolism , Protein Transport , Proteins/genetics , Receptors, Cholinergic/metabolism
16.
ASN Neuro ; 9(3): 1759091417708720, 2017.
Article in English | MEDLINE | ID: mdl-28523979

ABSTRACT

New neurons are born throughout the life of mammals in germinal zones of the brain known as neurogenic niches: the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus of the hippocampus. These niches contain a subpopulation of cells known as adult neural progenitor cells (aNPCs), which self-renew and give rise to new neurons and glia. aNPCs are regulated by many factors present in the niche, including the extracellular matrix (ECM). We show that the neuropeptide PACAP (pituitary adenylate cyclase-activating polypeptide) affects subventricular zone-derived aNPCs by increasing their surface adhesion. Gene array and reconstitution assays indicate that this effect can be attributed to the regulation of ECM components and ECM-modifying enzymes in aNPCs by PACAP. Our work suggests that PACAP regulates a bidirectional interaction between the aNPCs and their niche: PACAP modifies ECM production and remodeling, in turn the ECM regulates progenitor cell adherence. We speculate that PACAP may in this manner help restrict adult neural progenitors to the stem cell niche in vivo, with potential significance for aNPC function in physiological and pathological states.


Subject(s)
Adult Stem Cells/metabolism , Cell Adhesion/physiology , Neural Stem Cells/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/deficiency , Adult Stem Cells/cytology , Animals , Cells, Cultured , Culture Media, Conditioned , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Matrix/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Stem Cell Niche/physiology
17.
J Cell Sci ; 129(5): 898-911, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26769899

ABSTRACT

Neuromuscular junctions (NMJs), the synapses made by motor neurons on muscle fibers, form during embryonic development but undergo substantial remodeling postnatally. Several lines of evidence suggest that α-dystrobrevin, a component of the dystrophin-associated glycoprotein complex (DGC), is a crucial regulator of the remodeling process and that tyrosine phosphorylation of one isoform, α-dystrobrevin-1, is required for its function at synapses. We identified a functionally important phosphorylation site on α-dystrobrevin-1, generated phosphorylation-specific antibodies to it and used them to demonstrate dramatic increases in phosphorylation during the remodeling period, as well as in nerve-dependent regulation in adults. We then identified proteins that bind to this site in a phosphorylation-dependent manner and others that bind to α-dystrobrevin-1 in a phosphorylation-independent manner. They include multiple members of the DGC, as well as α-catulin, liprin-α1, Usp9x, PI3K, Arhgef5 and Grb2. Finally, we show that two interactors, α-catulin (phosphorylation independent) and Grb2 (phosphorylation dependent) are localized to NMJs in vivo, and that they are required for proper organization of neurotransmitter receptors on myotubes.


Subject(s)
Dystrophin-Associated Proteins/metabolism , GRB2 Adaptor Protein/metabolism , Neuromuscular Junction/metabolism , Neuropeptides/metabolism , Receptors, Cholinergic/metabolism , alpha Catenin/metabolism , Animals , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Interaction Maps , Protein Processing, Post-Translational , Protein Transport , Synaptic Transmission
18.
Histochem Cell Biol ; 144(1): 21-38, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25896210

ABSTRACT

The important role of unconventional myosin VI (MVI) in skeletal and cardiac muscle has been recently postulated (Karolczak et al. in Histochem Cell Biol 139:873-885, 2013). Here, we addressed for the first time a role for this unique myosin motor in myogenic cells as well as during their differentiation into myotubes. During myoblast differentiation, the isoform expression pattern of MVI and its subcellular localization underwent changes. In undifferentiated myoblasts, MVI-stained puncti were seen throughout the cytoplasm and were in close proximity to actin filaments, Golgi apparatus, vinculin-, and talin-rich focal adhesion as well as endoplasmic reticulum. Colocalization of MVI with endoplasmic reticulum was enhanced during myotube formation, and differentiation-dependent association was also seen in sarcoplasmic reticulum of neonatal rat cardiomyocytes (NRCs). Moreover, we observed enrichment of MVI in myotube regions containing acetylcholine receptor-rich clusters, suggesting its involvement in the organization of the muscle postsynaptic machinery. Overexpression of the H246R MVI mutant (associated with hypertrophic cardiomyopathy) in myoblasts and NRCs caused the formation of abnormally large intracellular vesicles. MVI knockdown caused changes in myoblast morphology and inhibition of their migration. On the subcellular level, MVI-depleted myoblasts exhibited aberrations in the organization of actin cytoskeleton and adhesive structures as well as in integrity of Golgi apparatus and endoplasmic reticulum. Also, MVI depletion or overexpression of H246R mutant caused the formation of significantly wider or aberrant myotubes, respectively, indicative of involvement of MVI in myoblast differentiation. The presented results suggest an important role for MVI in myogenic cells and possibly in myoblast differentiation.


Subject(s)
Muscle Development , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myoblasts/physiology , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cell Adhesion , Cell Differentiation , Cell Line , Cell Movement , Cell Shape , Cytoplasm/metabolism , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/ultrastructure , Mice , Myoblasts/ultrastructure , Myocytes, Cardiac/ultrastructure , Myosin Heavy Chains/chemistry , Rats , Sarcoplasmic Reticulum/metabolism
19.
Eur J Cell Biol ; 93(10-12): 478-85, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25012928

ABSTRACT

Podosomes are adhesive, matrix remodeling organelles that have been described in numerous cell types, including all three vertebrate muscle cell lineages. Podosomes have been intensively studied in smooth muscle cells, but they have also been described in cardiac myocytes and skeletal muscle cells where they are proposed to play a role in developmental remodeling of neuromuscular junction postsynaptic machinery. In this review, we summarize the current state of knowledge of podosomes in muscle cells, with a focus on their potential function at the maturing synapse.


Subject(s)
Cell Surface Extensions/metabolism , Muscle Fibers, Skeletal/metabolism , Myocytes, Smooth Muscle/metabolism , Neuromuscular Junction/metabolism , Animals , Cell Fusion , Cell Surface Extensions/ultrastructure , Humans , Muscle Fibers, Skeletal/ultrastructure , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Neuromuscular Junction/ultrastructure
20.
J Cell Sci ; 126(Pt 10): 2225-35, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23525008

ABSTRACT

Neuromuscular junctions (NMJs) in mammalian skeletal muscle undergo a postnatal topological transformation from a simple oval plaque to a complex branched structure. We previously showed that podosomes, actin-rich adhesive organelles, promote the remodeling process, and demonstrated a key role for one podosome component, LL5ß. To further investigate molecular mechanisms of postsynaptic maturation, we purified LL5ß-associated proteins from myotubes and showed that three regulators of the actin cytoskeleton--Amotl2, Asef2 and Flii--interact with LL5ß. These and other LL5ß-interacting proteins are associated with conventional podosomes in macrophages and podosome-like invadopodia in fibroblasts, strengthening the close relationship between synaptic and non-synaptic podosomes. We then focused on Amotl2, showing that it is associated with synaptic podosomes in cultured myotubes and with NMJs in vivo. Depletion of Amotl2 in myotubes leads to increased size of synaptic podosomes and corresponding alterations in postsynaptic topology. Depletion of Amotl2 from fibroblasts disrupts invadopodia in these cells. These results demonstrate a role for Amotl2 in synaptic maturation and support the involvement of podosomes in this process.


Subject(s)
Carrier Proteins/metabolism , Muscle, Skeletal/physiology , Myoblasts/physiology , Neuromuscular Junction , Actin Cytoskeleton/metabolism , Animals , Cell Differentiation , Cell Surface Extensions/ultrastructure , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Mice , Microfilament Proteins/metabolism , Myoblasts/ultrastructure , Protein Binding , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Synaptic Potentials , Trans-Activators
SELECTION OF CITATIONS
SEARCH DETAIL
...