Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
2.
Neuromuscul Disord ; 13(3): 207-15, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12609502

ABSTRACT

Deficiency of laminin alpha2 is the cause of one of the most severe muscular dystrophies in humans and other species. It is not yet clear how particular mutations in the laminin alpha2 chain gene affect protein expression, and how abnormal levels or structure of the protein affect disease. Animal models may be valuable for such genotype-phenotype analysis and for determining mechanism of disease as well as function of laminin. Here, we have analyzed protein expression in three lines of mice with mutations in the laminin alpha2 chain gene and in two lines of transgenic mice overexpressing the human laminin alpha2 chain gene in skeletal muscle. The dy(3K)/dy(3K) experimental mutant mice are completely deficient in laminin alpha2; the dy/dy spontaneous mutant mice have small amounts of apparently normal laminin; and the dy(W)/dy(W) mice express even smaller amounts of a truncated laminin alpha2, lacking domain VI. Interestingly, all mutants lack laminin alpha2 in peripheral nerve. We have demonstrated previously, that overexpression of the human laminin alpha2 in skeletal muscle in dy(2J)/dy(2J) and dy(W)/dy(W) mice under the control of a striated muscle-specific creatine kinase promoter substantially prevented the muscular dystrophy in these mice. However, dy(W)/dy(W) mice, expressing the human laminin alpha2 under the control of the striated muscle-specific portion of the desmin promoter, still developed muscular dystrophy. This failure to rescue is apparently because of insufficient production of laminin alpha2. This study provides additional evidence that the amount of laminin alpha2 is most critical for the prevention of muscular dystrophy. These data may thus be of significance for attempts to treat congenital muscular dystrophy in human patients.


Subject(s)
Genotype , Laminin/metabolism , Muscular Dystrophies/metabolism , Phenotype , Animals , DNA Mutational Analysis , Desmin/genetics , Disease Models, Animal , Fluorescent Antibody Technique/methods , Gene Expression , Humans , Immunoblotting/methods , Laminin/chemistry , Laminin/deficiency , Laminin/genetics , Mice , Mice, Mutant Strains/genetics , Mice, Mutant Strains/metabolism , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Peripheral Nerves/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary/physiology , Protein Subunits/immunology , Protein Subunits/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology
3.
Nature ; 413(6853): 302-7, 2001 Sep 20.
Article in English | MEDLINE | ID: mdl-11565031

ABSTRACT

Congenital muscular dystrophy is a heterogeneous and severe, progressive muscle-wasting disease that frequently leads to death in early childhood. Most cases of congenital muscular dystrophy are caused by mutations in LAMA2, the gene encoding the alpha2 chain of the main laminin isoforms expressed by muscle fibres. Muscle fibre deterioration in this disease is thought to be caused by the failure to form the primary laminin scaffold, which is necessary for basement membrane structure, and the missing interaction between muscle basement membrane and the dystrophin-glycoprotein complex (DGC) or the integrins. With the aim to restore muscle function in a mouse model for this disease, we have designed a minigene of agrin, a protein known for its role in the formation of the neuromuscular junction. Here we show that this mini-agrin-which binds to basement membrane and to alpha-dystroglycan, a member of the DGC-amends muscle pathology by a mechanism that includes agrin-mediated stabilization of alpha-dystroglycan and the laminin alpha5 chain. Our data provides in vivo evidence that a non-homologous protein in combination with rational protein design can be used to devise therapeutic tools that may restore muscle function in human muscular dystrophies.


Subject(s)
Agrin/genetics , Agrin/therapeutic use , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/therapy , Agrin/physiology , Animals , Basement Membrane/metabolism , Chickens , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Dystroglycans , Laminin/deficiency , Laminin/metabolism , Membrane Glycoproteins/metabolism , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/congenital , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Protein Binding
5.
Nat Struct Biol ; 8(8): 705-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11473262

ABSTRACT

Agrin is the key organizer of postsynaptic differentiation at the neuromuscular junction. This organization activity requires the binding of agrin to the synaptic basal lamina. Binding is conferred by the N-terminal agrin (NtA) domain, which mediates a high-affinity interaction with the coiled coil domain of laminins. Here, we report the crystal structure of chicken NtA at 1.6 A resolution. The structure reveals that NtA harbors an oligosaccharide/oligonucleotide-binding fold with several possible sites for the interaction with different ligands. A high structural similarity of NtA with the protease inhibition domain in tissue inhibitor of metalloproteinases-1 (TIMP-1) supports the idea of additional functions of agrin besides synaptogenic activity.


Subject(s)
Agrin/chemistry , Agrin/metabolism , Chickens , Laminin/metabolism , Tissue Inhibitor of Metalloproteinase-1/chemistry , Amino Acid Sequence , Animals , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Humans , Laminin/chemistry , Ligands , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Static Electricity
6.
Matrix Biol ; 20(1): 3-12, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11245999

ABSTRACT

Synapses are highly specialized structures designed to guarantee precise and efficient communication between neurons and their target cells. Molecules of the extracellular matrix have an instructive role in the formation of the neuromuscular junction, the best-characterized synapse. In this review, the molecular mechanisms underlying these instructive signals will be discussed with particular emphasis on the receptors involved. Additionally, recent evidence for the involvement of specific adhesion complexes in the formation and modulation of synapses in the central nervous system will be reviewed. Synapses are specialized junctions between neurons and their target cells where information is transferred from the pre- to the postsynaptic cell. At most vertebrate synapses, this transfer is accomplished by the release of a specific neurotransmitter from the presynaptic nerve terminal. The release of neurotransmitter is initiated by the action potential and the subsequent influx of Ca(2+) into the presynaptic nerve terminal. This results in the rapid fusion of vesicles with the nerve membrane and the release of the neurotransmitter into the synaptic cleft. The neurotransmitter then diffuses across the cleft and binds to specific postsynaptic receptors, resulting in a change in the membrane potential of the postsynaptic cell. This can result in the generation of an action potential. The high precision of synaptic transmission requires that pre- and postsynaptic structures are both highly organized and in juxtaposition to each other. In addition, alterations in synaptic transmission are the basis of learning and memory and are likely to be accompanied by the remodeling of synaptic structures (Toni et al., 1999). Thus, the study of how synapses are formed during development is also of relevance for the understanding of the cellular and molecular processes involved in learning and memory. This review focuses on the molecular mechanisms involved in the formation and the function of synapses.


Subject(s)
Extracellular Matrix Proteins/physiology , Neuromuscular Junction/physiology , Synaptic Transmission/physiology , Animals , Brain/physiology , Humans , Muscles/innervation , Neurons/physiology , Synapses/physiology
7.
Mol Cell Neurosci ; 17(1): 208-25, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11161480

ABSTRACT

Agrin is a basal lamina-associated heparansulfate proteoglycan that is a key molecule in the formation of the vertebrate neuromuscular junction. The carboxy-terminal part of agrin is involved in its synaptogenic activity. The amino-terminal end of chick agrin consists of a signal sequence, required for the targeting of the protein to the secretory pathway, and the amino-terminal agrin (NtA) domain that binds to basal lamina-associated laminins. The cDNA encoding rat agrin lacks this NtA domain and instead codes for a shorter amino-terminal end. While the NtA domain is conserved in several species, including human, sequences homologous to the amino-terminus of rat agrin have not been described. In this work, we have characterized these amino-terminal sequences in mouse and chick. We show that they all serve as a noncleaved signal anchor that immobilizes the protein in a N(cyto)/C(exo) orientation in the plasma membrane. Like the secreted form, this transmembrane form of agrin is highly glycosylated indicative of a heparansulfate proteoglycan. The structure of the 5' end of the mouse agrin gene suggests that a distinct promoter drives expression of the transmembrane form. Agrin transcripts encoding this form are enriched in the embryonic brain, particularly in neurons. To our knowledge, this is the first example of a molecule that is synthesized both as a basal lamina and a plasma membrane protein.


Subject(s)
Agrin/metabolism , Cell Membrane/metabolism , Central Nervous System/metabolism , Membrane Proteins/biosynthesis , Protein Sorting Signals/physiology , Agrin/genetics , Animals , Cell Line , Chick Embryo , Conserved Sequence/physiology , Glycosylation , Humans , Mice , Neuromuscular Junction/metabolism , Protein Processing, Post-Translational/genetics , Protein Sorting Signals/genetics , Rats , Receptor Aggregation/physiology , Receptors, Cholinergic/metabolism , Sequence Homology, Amino Acid , Species Specificity , Transfection
8.
Exp Gerontol ; 35(9-10): 1389-403, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11113617

ABSTRACT

The discovery of two missense mutations (A53T and A30P) in the gene encoding the presynaptic protein alpha-synuclein (alphaSN) that are genetically linked to rare familial forms of Parkinson's disease and its accumulation in Lewy bodies and Lewy neurites has triggered several attempts to generate transgenic mice overexpressing human alphaSN. Analogous to a successful strategy for the production of transgenic animal models for Alzheimer's disease we generated mice expressing wildtype and the A53T mutant of human alphaSN in the nervous system under control of mouse Thy1 regulatory sequences. These animals develop neuronal alpha-synucleinopathy, striking features of Lewy pathology, neuronal degeneration and motor defects. Neurons in brainstem and motor neurons appeared particularly vulnerable. Motor neuron pathology included axonal damage and denervation of neuromuscular junctions, suggesting that alphaSN may interfere with a universal mechanism of synapse maintenance. Thy1-transgene expression of wildtype human alphaSN resulted in comparable pathological changes thus supporting a central role for mutant and wildtype alphaSN in familial and idiopathic forms of diseases with neuronal alpha-synucleinopathy and Lewy pathology. The mouse models provide means to address fundamental aspects of alpha-synucleinopathy and to test therapeutic strategies.


Subject(s)
Lewy Bodies/pathology , Nerve Tissue Proteins/genetics , Parkinson Disease/pathology , Amino Acid Substitution , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Gene Expression , Genotype , Humans , Lewy Bodies/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Mutation , Parkinson Disease/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synucleins , Transgenes/genetics , alpha-Synuclein
9.
J Neurosci ; 20(16): 5989-96, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10934247

ABSTRACT

At chemical synapses, neurotransmitter receptors are concentrated in the postsynaptic membrane. During the development of the neuromuscular junction, motor neurons induce aggregation of acetylcholine receptors (AChRs) underneath the nerve terminal by the redistribution of existing AChRs and preferential transcription of the AChR subunit genes in subsynaptic myonuclei. Neural agrin, when expressed in nonsynaptic regions of muscle fibers in vivo, activates both mechanisms resulting in the assembly of a fully functional postsynaptic apparatus. Several lines of evidence indicate that synaptic transcription of AChR genes is primarily dependent on a promoter element called N-box. The Ets-related transcription factor growth-associated binding protein (GABP) binds to this motif and has thus been suggested to regulate synaptic gene expression. Here, we assessed the role of GABP in synaptic gene expression and in the formation of postsynaptic specializations in vivo by perturbing its function during postsynaptic differentiation induced by neural agrin. We find that neural agrin-mediated activation of the AChR epsilon subunit promoter is abolished by the inhibition of GABP function. Importantly, the number of AChR aggregates formed in response to neural agrin was strongly reduced. Moreover, aggregates of acetylcholine esterase and utrophin, two additional components of the postsynaptic apparatus, were also reduced. Together, these results are the first direct in vivo evidence that GABP regulates synapse-specific gene expression at the neuromuscular junction and that GABP is required for the formation of a functional postsynaptic apparatus.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Neuromuscular Junction/embryology , Receptors, Cholinergic/metabolism , Synaptic Membranes/enzymology , Transcription Factors/metabolism , Acetylcholinesterase/metabolism , Agrin/genetics , Agrin/metabolism , Animals , COS Cells , Cell Differentiation/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , GA-Binding Protein Transcription Factor , Membrane Proteins/metabolism , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/ultrastructure , Mutation/genetics , Neuromuscular Junction/enzymology , Neuromuscular Junction/ultrastructure , Promoter Regions, Genetic/genetics , Rats , Receptors, Cholinergic/genetics , Synaptic Membranes/ultrastructure , Transcription Factors/genetics , Utrophin
10.
J Neurosci ; 20(16): 6021-9, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10934251

ABSTRACT

The presynaptic protein alpha-synuclein is a prime suspect for contributing to Lewy pathology and clinical aspects of diseases, including Parkinson's disease, dementia with Lewy bodies, and a Lewy body variant of Alzheimer's disease. alpha-Synuclein accumulates in Lewy bodies and Lewy neurites, and two missense mutations (A53T and A30P) in the alpha-synuclein gene are genetically linked to rare familial forms of Parkinson's disease. Under control of mouse Thy1 regulatory sequences, expression of A53T mutant human alpha-synuclein in the nervous system of transgenic mice generated animals with neuronal alpha-synucleinopathy, features strikingly similar to those observed in human brains with Lewy pathology, neuronal degeneration, and motor defects, despite a lack of transgene expression in dopaminergic neurons of the substantia nigra pars compacta. Neurons in brainstem and motor neurons appeared particularly vulnerable. Motor neuron pathology included axonal damage and denervation of neuromuscular junctions in several muscles examined, suggesting that alpha-synuclein interfered with a universal mechanism of synapse maintenance. Thy1 transgene expression of wild-type human alpha-synuclein resulted in similar pathological changes, thus supporting a central role for mutant and wild-type alpha-synuclein in familial and idiotypic forms of diseases with neuronal alpha-synucleinopathy and Lewy pathology. These mouse models provide a means to address fundamental aspects of alpha-synucleinopathy and test therapeutic strategies.


Subject(s)
Central Nervous System/pathology , Gene Expression Regulation/physiology , Lewy Bodies/metabolism , Mutation/physiology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/physiopathology , Animals , Central Nervous System/metabolism , Central Nervous System/physiopathology , Humans , Lewy Bodies/genetics , Mice , Mice, Transgenic , Motor Activity/physiology , Motor Neurons/metabolism , Motor Neurons/pathology , Motor Neurons/ultrastructure , Movement Disorders/genetics , Movement Disorders/pathology , Movement Disorders/physiopathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Psychomotor Performance/physiology , Synucleins , alpha-Synuclein
11.
EMBO J ; 18(23): 6762-70, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10581249

ABSTRACT

Coiled-coil domains are found in a wide variety of proteins, where they typically specify subunit oligomerization. Recently, we have demonstrated that agrin, a multidomain heparan sulfate proteoglycan with a crucial role in the development of the nerve-muscle synapse, binds to the three-stranded coiled-coil domain of laminin-1. The interaction with laminin mediates the integration of agrin into basement membranes. Here we characterize the binding site within the laminin-1 coiled coil in detail. Binding assays with individual laminin-1 full-length chains and fragments revealed that agrin specifically interacts with the gamma1 subunit of laminin-1, whereas no binding to alpha1 and beta1 chains was detected. By using recombinant gamma1 chain fragments, we mapped the binding site to a sequence of 20 residues. Furthermore, we demonstrate that a coiled-coil conformation of this binding site is required for its interaction with agrin. The finding that recombinant gamma1 fragments bound at least 10-fold less than native laminin-1 indicates that the structure of the three-stranded coiled-coil domain of laminin is required for high-affinity agrin binding. Interestingly, no binding to a chimeric gamma2 fragment was observed, indicating that the interaction of agrin with laminin is isoform specific.


Subject(s)
Agrin/chemistry , Agrin/metabolism , Laminin/chemistry , Laminin/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Circular Dichroism , DNA, Complementary/metabolism , Escherichia coli/metabolism , Gene Deletion , Laminin/genetics , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Temperature , Transfection , Ultracentrifugation
12.
Eur J Neurosci ; 10(10): 3141-52, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9786208

ABSTRACT

The extracellular matrix molecule agrin is both necessary and sufficient for inducing the formation of postsynaptic specializations at the neuromuscular junction (NMJ). At the mature NMJ, agrin is stably incorporated in synaptic basal lamina. The postsynapse-inducing activity of chick agrin, as assayed by its capability of causing aggregation of acetylcholine receptors (AChRs) on cultured muscle cells, maps to a 21 kDa, C-terminal domain. Binding of chick agrin to muscle basal lamina is mediated by the laminins and maps to a 25 kDa, N-terminal fragment of agrin. Here we show that an expression construct encoding a 'mini'-agrin, in which the laminin-binding fragment was fused to the AChR-clustering domain, is sufficient to induce postsynaptic differentiation in vivo when injected into non-synaptic sites of rat soleus muscle. As shown for ectopic postsynaptic differentiation induced by full-length neural agrin, myonuclei underneath the ectopic sites expressed the gene for the AChR epsilon-subunit. Altogether, our data show that a 'mini'-agrin construct encoding only a small fraction of the entire agrin protein is sufficient to induce postsynapse-like structures that are reminiscent of those induced by full-length neural agrin or innervation by motor neurons.


Subject(s)
Agrin/genetics , Genes , Muscle Fibers, Skeletal/drug effects , Protein Structure, Tertiary , Recombinant Fusion Proteins/pharmacology , Agrin/physiology , Animals , COS Cells , Cell Differentiation/drug effects , Cells, Cultured , Chick Embryo , Extracellular Matrix , Gene Expression Regulation/drug effects , Laminin/metabolism , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Nerve Tissue Proteins , Neuromuscular Junction , Protein Binding/genetics , Rats , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Synapses/physiology , Transcription, Genetic , Transfection
13.
J Cell Sci ; 111 ( Pt 17): 2595-605, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9701558

ABSTRACT

Dystrophin-related and associated proteins are important for the formation and maintenance of the mammalian neuromuscular junction. Initial studies in the electric organ of Torpedo californica showed that the dystrophin-related protein dystrobrevin (87K) co-purifies with the acetylcholine receptors and other postsynaptic proteins. Dystrobrevin is also a major phosphotyrosine-containing protein in the postsynaptic membrane. Since inhibitors of tyrosine protein phosphorylation block acetylcholine receptor clustering in cultured muscle cells, we examined the role of alpha-dystrobrevin during synapse formation and in response to agrin. Using specific antibodies, we show that C2 myoblasts and early myotubes only produce alpha-dystrobrevin-1, the mammalian orthologue of Torpedo dystrobrevin, whereas mature skeletal muscle expresses three distinct alpha-dystrobrevin isoforms. In myotubes, alpha-dystrobrevin-1 is found on the cell surface and also in acetylcholine receptor-rich domains. Following agrin stimulation, alpha-dystrobrevin-1 becomes re-localised beneath the cell surface into macroclusters that contain acetylcholine receptors and another dystrophin-related protein, utrophin. This redistribution is not associated with tyrosine phosphorylation of alpha-dystrobrevin-1 by agrin. Furthermore, we show that alpha-dystrobrevin-1 is associated with both utrophin in C2 cells and dystrophin in mature skeletal muscle. Thus alpha-dystrobrevin-1 is a component of two protein complexes in muscle, one with utrophin at the neuromuscular junction and the other with dystrophin at the sarcolemma. These results indicate that alpha-dystrobrevin-1 is not involved in the phosphorylation-dependent, early stages of receptor clustering, but rather in the stabilisation and maturation of clusters, possibly via an interaction with utrophin.


Subject(s)
Dystrophin-Associated Proteins , Muscle Proteins/chemistry , Muscle, Skeletal/chemistry , Neuropeptides/chemistry , Agrin/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Brain , Cell Line , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Fetus , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred mdx , Molecular Sequence Data , Muscle Proteins/genetics , Muscle, Skeletal/cytology , Neuropeptides/genetics , Phosphotyrosine/metabolism , Protein Isoforms/chemistry , Synapses/chemistry , Synapses/physiology , Utrophin , Vanadates/pharmacology
14.
Mol Cell Neurosci ; 11(4): 206-16, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9675052

ABSTRACT

The accumulation of nicotinic acetylcholine receptors (AChRs) at neuromuscular synapses is triggered by agrin, a protein that is synthesized by both nerve and muscle. Nerve-derived agrin, which contains an amino acid insert at a conserved splice site in the carboxy-terminal part of the protein, induces AChR aggregation and causes tyrosine phosphorylation of the AChR beta subunit. In contrast, agrin isoforms synthesized by muscle cells lack such an insert and have no effect on AChR distribution. In order to identify possible functional roles of muscle-derived agrin we have analyzed further the effect of various fragments of recombinant agrin on AChR phosphorylation. A carboxy-terminal fragment of muscle agrin, c95A0B0, reduced AChR gamma and delta subunit phosphorylation when added to C2C12 myotubes in culture. Although c95A0B0 had no effect on AChR beta subunit phosphorylation when added alone, it inhibited AChR beta subunit phosphorylation and AChR aggregation by the nerve-specific agrin isoform c95A4B8. We conclude that muscle-derived agrin can influence, both directly and indirectly, AChR phosphorylation. Such changes may play a role in the formation, maintenance, or function of the neuromuscular junction.


Subject(s)
Agrin/pharmacology , Muscle Proteins/pharmacology , Muscle, Skeletal/drug effects , Protein Processing, Post-Translational/drug effects , Receptors, Cholinergic/metabolism , Agrin/genetics , Agrin/physiology , Animals , Cells, Cultured , Chickens , Cytoskeletal Proteins/metabolism , Dystroglycans , Laminin/pharmacology , Membrane Glycoproteins/metabolism , Muscle Proteins/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/pharmacology , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Receptors, Cholinergic/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Structure-Activity Relationship
15.
Trends Neurosci ; 21(1): 22-7, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9464682

ABSTRACT

The synapse is a key structure that is involved in perception, learning and memory. Understanding the sequence of steps that is involved in establishing synapses during development might also help to understand mechanisms that cause changes in synapses during learning and memory. For practical reasons, most of our current knowledge of synapse development is derived from studies of the vertebrate neuromuscular junction (NMJ). Several lines of evidence strongly suggest that motor axons release the molecule agrin to induce the formation of the postsynaptic apparatus in muscle fibers. Recent advances implicate proteins such as dystroglycan, MuSK, and rapsyn in the transduction of agrin signals. Recently, additional functions of agrin have been discovered, including the upregulation of gene transcription in myonuclei and the control of presynaptic differentiation. Agrin therefore appears to play a unique role in controlling synaptic differentiation on both sides of the NMJ.


Subject(s)
Agrin/physiology , Cell Differentiation/physiology , Neuromuscular Junction/physiology , Neurons/physiology , Synapses/physiology , Agrin/metabolism , Animals , Humans , Neuromuscular Junction/metabolism , Synapses/metabolism
16.
J Biol Chem ; 273(1): 600-5, 1998 Jan 02.
Article in English | MEDLINE | ID: mdl-9417121

ABSTRACT

Agrin is a basement membrane-associated proteoglycan that induces the formation of postsynaptic specializations at the neuromuscular junction. This activity is modulated by alternative splicing and is thought to be mediated by receptors expressed in muscle fibers. An isoform of agrin that does not induce postsynaptic specializations binds with high affinity to dystroglycan, a component of the dystrophin-glycoprotein complex. Transcripts encoding this agrin isoform are expressed in a variety of non-muscle tissues. Here, we analyzed the tissue distribution of agrin and dystroglycan on the protein level and determined their binding affinities. We found that agrin is most abundant in lung, kidney, and brain. Only a little agrin was detected in skeletal muscle, and no agrin was found in liver. Dystroglycan was highly expressed in all tissues examined except in liver. In a solid-phase radioligand binding assay, agrin bound to dystroglycan from lung, kidney, and skeletal muscle with a dissociation constant between 1.8 and 2.2 nM, while the affinity to brain-derived dystroglycan was 4.6 nM. In adult kidney and lung, agrin co-purified and co-immunoprecipitated with dystroglycan, and both molecules were co-localized in embryonic tissue. These data show that the agrin isoform expressed in non-muscle tissue is a high-affinity binding partner of dystroglycan and they suggest that this interaction, like that between laminin and dystroglycan, may be important for the mechanical integrity of the tissue.


Subject(s)
Agrin/metabolism , Cytoskeletal Proteins/metabolism , Membrane Glycoproteins/metabolism , Animals , COS Cells , Chickens , Dystroglycans , Kidney/metabolism , Lung/metabolism , Muscle, Skeletal/metabolism , Protein Binding
17.
EMBO J ; 17(2): 335-43, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9430625

ABSTRACT

Agrin is a large, multidomain heparan sulfate proteoglycan that is associated with basement membranes of several tissues. Particular splice variants of agrin are essential for the formation of synaptic structures at the neuromuscular junction. The binding of agrin to laminin appears to be required for its localization to synaptic basal lamina and other basement membranes. Here, electron microscopy was used to determine the structure of agrin and to localize its binding site in laminin-1. Agrin appears as an approximately 95 nm long particle that consists of a globular, N-terminal laminin-binding domain, a central rod predominantly formed by the follistatin-like domains and three globular, C-terminal laminin G-like domains. In a few cases, heparan sulfate glycosaminoglycan chains were seen emerging from the central portion of the core protein. Moreover, we show that agrin binds to the central region of the three-stranded, coiled-coil oligomerization domain in the long arm of laminin-1, which mediates subunit assembly of the native laminin molecule. In summary, our data show for the first time a protein-protein interaction of the extracellular matrix that involves a coiled-coil domain, and they assign a novel role to this domain of laminin-1. Based on this, we propose that agrin associates with basal lamina in a polarized way.


Subject(s)
Agrin/chemistry , Agrin/ultrastructure , Laminin/metabolism , Agrin/genetics , Agrin/metabolism , Animals , Binding Sites , COS Cells , Cell Line , Chickens , Humans , Laminin/chemistry , Laminin/ultrastructure , Microscopy, Electron , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Protein Structure, Tertiary , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
18.
Neurology ; 50(1): 54-61, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9443457

ABSTRACT

We studied two families with five affected members suffering from ptosis and slowly progressive limb-girdle muscle weakness. All patients had abnormal decremental response on low-frequency nerve stimulation, but there were no repetitive responses to single stimuli. The patients improved on anti-acetylcholinesterase drugs. Intercostal muscle was obtained for special studies from one patient of each family. In vitro microelectrode studies were done in Patient 1. Miniature end-plate potentials were of low amplitude, and the quantal content of the evoked end-plate potentials was normal. Light microscopy revealed a marked type 1 fiber predominance. Acetylcholinesterase reactivity was dispersed over increased length of individual fibers in Patient 2. On morphometry of the end-plate ultrastructure, the number of secondary synaptic clefts per neuromuscular junction and the expansion of the postsynaptic area were markedly reduced. In Patient 1, but not in Patient 2, the envelopment of the nerve terminal by Schwann cell was increased. Acetylcholine-receptor (AChR) density was reduced as judged by the reduced immunoreactivity to antibodies against different receptor subunits. Immunohistochemical analysis of proteins known to be involved in orchestrating the end-plate structure showed deficiency of the AChR-associated protein utrophin. These patients appear to have a defect in the development or maintenance of the postsynaptic clefts; whether this defect results from or causes a reduced expression of utrophin or AChR is unclear.


Subject(s)
Cytoskeletal Proteins/deficiency , Membrane Proteins/deficiency , Motor Endplate/chemistry , Myasthenia Gravis/congenital , Myasthenia Gravis/genetics , Receptors, Cholinergic/deficiency , Adult , Animals , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/genetics , Female , Humans , Male , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Mice, Knockout , Microscopy, Electron , Motor Endplate/ultrastructure , Myasthenia Gravis/pathology , Pedigree , Receptors, Cholinergic/analysis , Receptors, Cholinergic/genetics , Synaptic Vesicles/ultrastructure , Utrophin
19.
J Histochem Cytochem ; 46(1): 19-27, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9405491

ABSTRACT

Agrin is a heparan sulfate proteoglycan (HSPG) that is highly concentrated in the synaptic basal lamina at the neuromuscular junction (NMJ). Agrin-like immunoreactivity is also detected outside the NMJ. Here we show that agrin is a major HSPG component of the human glomerular basement membrane (GBM). This is in addition to perlecan, a previously characterized HSPG of basement membranes. Antibodies against agrin and against an unidentified GBM HSPG produced a strong staining of the GBM and the NMJ, different from that observed with anti-perlecan antibodies. In addition, anti-agrin antisera recognized purified GBM HSPG and competed with an anti-GBM HSPG monoclonal antibody in ELISA. Furthermore, both antibodies recognized a molecule that migrated in SDS-PAGE as a smear and had a molecular mass of approximately 200-210 kD after deglycosylation. In immunoelectron microscopy, agrin showed a linear distribution along the GBM and was present throughout the width of the GBM. This was again different from perlecan, which was exclusively present on the endothelial side of the GBM and was distributed in a nonlinear manner. Quantitative ELISA showed that, compared with perlecan, the agrin-like GBM HSPG showed a sixfold higher molarity in crude glomerular extract. These results show that agrin is a major component of the GBM, indicating that it may play a role in renal ultrafiltration and cell matrix interaction. (J Histochem Cytochem 46:19-27, 1998)


Subject(s)
Agrin/biosynthesis , Basement Membrane/metabolism , Heparan Sulfate Proteoglycans/metabolism , Kidney Glomerulus/metabolism , Adult , Agrin/immunology , Animals , Antibodies, Monoclonal , Basement Membrane/ultrastructure , Bungarotoxins/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Heparitin Sulfate/metabolism , Humans , Immune Sera/metabolism , Kidney Cortex/cytology , Kidney Cortex/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/ultrastructure , Microscopy, Fluorescence , Microscopy, Immunoelectron , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Proteoglycans/metabolism , Rats
20.
Cell Tissue Res ; 290(2): 357-65, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9321698

ABSTRACT

Upon arrival of a motor axon at the muscle fiber, signals released from its growth cone initiate the formation of a synapse. This process consists of two stages: arrest of axon growth at the target area and differentiation of pre- and postsynaptic cells at the site of nerve-muscle contact. Studies of regenerating neuromuscular junctions in vertebrates have revealed that important signals for the formation of this synapse are located in the synaptic basal lamina, and attempts to identify these signals have led to the isolation of agrin and other components. In this review, we discuss the evidence for the involvement of these molecules and their potential functional role in the formation and maintenance of the neuromuscular junction, with emphasis on agrin.


Subject(s)
Agrin/physiology , Neuromuscular Junction/physiology , Synapses/physiology , Animals , Humans , Nerve Tissue Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...