Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Cell Neurosci ; 12: 195, 2018.
Article in English | MEDLINE | ID: mdl-30065631

ABSTRACT

Migration and pathfinding of neuronal growth cones during neurite extension is critically dependent on dynamic microtubules. In this study we sought to determine, which aspects of microtubule polymerization relate to growth cone morphology and migratory characteristics. We conducted a multiscale quantitative microscopy analysis using automated tracking of microtubule plus ends in migrating growth cones of cultured murine dorsal root ganglion (DRG) neurons. Notably, this comprehensive analysis failed to identify any changes in microtubule polymerization parameters that were specifically associated with spontaneous extension vs. retraction of growth cones. This suggests that microtubule dynamicity is a basic mechanism that does not determine the polarity of growth cone response but can be exploited to accommodate diverse growth cone behaviors. At the same time, we found a correlation between growth cone size and basic parameters of microtubule polymerization including the density of growing microtubule plus ends and rate and duration of microtubule growth. A similar correlation was observed in growth cones of neurons lacking the microtubule-associated protein MAP1B. However, MAP1B-null growth cones, which are deficient in growth cone migration and steering, displayed an overall reduction in microtubule dynamicity. Our results highlight the importance of taking growth cone size into account when evaluating the influence on growth cone microtubule dynamics of different substrata, guidance factors or genetic manipulations which all can change growth cone morphology and size. The type of large scale multiparametric analysis performed here can help to separate direct effects that these perturbations might have on microtubule dynamics from indirect effects resulting from perturbation-induced changes in growth cone size.

2.
Mol Cell Neurosci ; 72: 9-21, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26773468

ABSTRACT

The microtubule-associated protein MAP1B plays a key role in axon regeneration. We investigated the role of GSK3-mediated MAP1B phosphorylation in local fine-tuning of neurite branching and the underlying microtubule (MT) dynamics. In wildtype adult dorsal root ganglia (DRG) neurons, MAP1B phosphorylation is locally reduced at branching points, and branching dynamics from growth cones and distal neurite shafts is increased upon GSK3 inhibition. While map1b-/- neurites, that display increased branching, are not affected by GSK3 inhibition, transfection of map1b-/- neurons with full-length map1b-cDNA restores the wildtype branching phenotype, demonstrating that MAP1B is a key effector downstream of GSK3. Experiments in mutant mice lacking tyrosinated MTs indicate a preferential association of phospho-MAP1B with tyrosinated MTs. Interestingly, inhibition of GSK3-mediated MAP1B phosphorylation in map1b-cDNA-transfected fibroblasts protects both tyrosinated and acetylated MTs from nocodazole-induced depolymerization, while detyrosinated MTs are less abundant in the presence of MAP1B. Our data thus provide new insight into the molecular link between GSK3, MAP1B, neurite branching and MT stability regulation. We suggest that, at branching points, MAP1B undergoes a fine regulation of both its phosphorylation and sub-cellular amounts, in order to modulate the local balance between acetylated, detyrosinated, and tyrosinated microtubule pools.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neurites/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Mice , Microtubule-Associated Proteins/genetics , Neurogenesis , Phosphorylation
3.
PLoS One ; 10(10): e0140116, 2015.
Article in English | MEDLINE | ID: mdl-26448484

ABSTRACT

Podocytes are essential for the function of the kidney glomerular filter. A highly differentiated cytoskeleton is requisite for their integrity. Although much knowledge has been gained on the organization of cortical actin networks in podocyte's foot processes, less is known about the molecular organization of the microtubular cytoskeleton in primary processes and the cell body. To gain an insight into the organization of the microtubular cytoskeleton of the podocyte, we systematically analyzed the expression of microtubule associated proteins (Maps), a family of microtubules interacting proteins with known functions as regulator, scaffold and guidance proteins. We identified microtubule associated protein 1b (MAP1B) to be specifically enriched in podocytes in human and rodent kidney. Using immunogold labeling in electron microscopy, we were able to demonstrate an enrichment of MAP1B in primary processes. A similar association of MAP1B with the microtubule cytoskeleton was detected in cultured podocytes. Subcellular distribution of MAP1B HC and LC1 was analyzed using a double fluorescent reporter MAP1B fusion protein. Subsequently we analyzed mice constitutively depleted of MAP1B. Interestingly, MAP1B KO was not associated with any functional or structural alterations pointing towards a redundancy of MAP proteins in podocytes. In summary, we established MAP1B as a specific marker protein of the podocyte microtubular cytoskeleton.


Subject(s)
Glomerular Filtration Barrier/metabolism , Podocytes/metabolism , Animals , Biomarkers , Cells, Cultured , Female , Glomerular Filtration Rate , Male , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins , Microtubules/metabolism , Microtubules/ultrastructure , Organ Specificity , Podocytes/ultrastructure
4.
PLoS One ; 10(3): e0119524, 2015.
Article in English | MEDLINE | ID: mdl-25775433

ABSTRACT

Draxin is an important axon guidance cue necessary for the formation of forebrain commissures including the corpus callosum, but the molecular details of draxin signaling are unknown. To unravel how draxin signals are propagated we used murine cortical neurons and genetic and pharmacological approaches. We found that draxin-induced growth cone collapse critically depends on draxin receptors (deleted in colorectal cancer, DCC), inhibition of protein kinase B/Akt, activation of GSK-3ß (glycogen synthase kinase-3ß) and the presence of microtubule-associated protein MAP1B. This study, for the first time elucidates molecular events in draxin repulsion, links draxin and DCC to MAP1B and identifies a novel MAP1B-depenent GSK-3ß pathway essential for chemo-repulsive axon guidance cue signaling.


Subject(s)
Axons/physiology , Glycogen Synthase Kinase 3/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cells, Cultured , DCC Receptor , Female , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3 beta , Male , Mice , Neurons/physiology , Receptors, Cell Surface/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism
5.
Neuron ; 82(2): 334-49, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24656932

ABSTRACT

The ocular motility disorder "Congenital fibrosis of the extraocular muscles type 1" (CFEOM1) results from heterozygous mutations altering the motor and third coiled-coil stalk of the anterograde kinesin, KIF21A. We demonstrate that Kif21a knockin mice harboring the most common human mutation develop CFEOM. The developing axons of the oculomotor nerve's superior division stall in the proximal nerve; the growth cones enlarge, extend excessive filopodia, and assume random trajectories. Inferior division axons reach the orbit but branch ectopically. We establish a gain-of-function mechanism and find that human motor or stalk mutations attenuate Kif21a autoinhibition, providing in vivo evidence for mammalian kinesin autoregulation. We identify Map1b as a Kif21a-interacting protein and report that Map1b⁻/⁻ mice develop CFEOM. The interaction between Kif21a and Map1b is likely to play a critical role in the pathogenesis of CFEOM1 and highlights a selective vulnerability of the developing oculomotor nerve to perturbations of the axon cytoskeleton.


Subject(s)
Axons/pathology , Eye Diseases, Hereditary/genetics , Fibrosis/genetics , Kinesins/genetics , Kinesins/metabolism , Mutation/genetics , Ocular Motility Disorders/genetics , Oculomotor Nerve/pathology , Age Factors , Animals , Animals, Newborn , Axons/ultrastructure , Cell Count , Disease Models, Animal , Embryo, Mammalian , Eye Diseases, Hereditary/pathology , Eye Diseases, Hereditary/physiopathology , Eye Movements/genetics , Eye Movements/physiology , Fibrosis/pathology , Fibrosis/physiopathology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/ultrastructure , Ocular Motility Disorders/pathology , Ocular Motility Disorders/physiopathology , Oculomotor Nerve/ultrastructure
6.
Biomol NMR Assign ; 8(1): 123-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23339032

ABSTRACT

Microtubule-associated protein 1B (MAP1B) is a classical high molecular mass microtubule-associated protein expressed at high levels in the brain. It confers specific properties to neuronal microtubules and is essential for neuronal differentiation, brain development and synapse maturation. Misexpression of the protein contributes to the development of brain disorders in humans. However, despite numerous reports demonstrating the importance of MAP1B in regulation of the neuronal cytoskeleton during neurite extension and axon guidance, its mechanism of action is still elusive. Here we focus on the intrinsically disordered microtubule binding domain of the light chain of MAP1B. In order to obtain more detailed structural information about this domain we assigned NMR chemical shifts of backbone and aliphatic side chain atoms.


Subject(s)
Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nuclear Magnetic Resonance, Biomolecular , Amino Acid Sequence , Animals , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Rats
7.
PLoS One ; 8(6): e68168, 2013.
Article in English | MEDLINE | ID: mdl-23840827

ABSTRACT

Protein S-nitrosylation, the covalent attachment of a nitroso moiety to thiol groups of specific cysteine residues, is one of the major pathways of nitric oxide signaling. Hundreds of proteins are subject to this transient post-translational modification and for some the functional consequences have been identified. Biochemical assays for the analysis of protein S-nitrosylation have been established and can be used to study if and under what conditions a given protein is S-nitrosylated. In contrast, the equally desirable subcellular localization of specific S-nitrosylated protein isoforms has not been achieved to date. In the current study we attempted to specifically localize S-nitrosylated α- and ß-tubulin isoforms in primary neurons after fixation. The approach was based on in situ replacement of the labile cysteine nitroso modification with a stable tag and the subsequent use of antibodies which recognize the tag in the context of the tubulin polypeptide sequence flanking the cysteine residue of interest. We established a procedure for tagging S-nitrosylated proteins in cultured primary neurons and obtained polyclonal anti-tag antibodies capable of specifically detecting tagged proteins on immunoblots and in fixed cells. However, the antibodies were not specific for tubulin isoforms. We suggest that different tagging strategies or alternative methods such as fluorescence resonance energy transfer techniques might be more successful.


Subject(s)
Antibodies/metabolism , Immunohistochemistry/methods , S-Nitrosothiols/metabolism , Tubulin/metabolism , Animals , Cells, Cultured , Cysteine/metabolism , Mice , Neurons/metabolism , Nitric Oxide/metabolism , Protein Isoforms/metabolism , Sulfhydryl Compounds/metabolism
8.
PLoS One ; 7(11): e49722, 2012.
Article in English | MEDLINE | ID: mdl-23152929

ABSTRACT

Microtubule-associated proteins of the MAP1 family (MAP1A, MAP1B, and MAP1S) share, among other features, a highly conserved COOH-terminal domain approximately 125 amino acids in length. We conducted a yeast 2-hybrid screen to search for proteins interacting with this domain and identified α1-syntrophin, a member of a multigene family of adapter proteins involved in signal transduction. We further demonstrate that the interaction between the conserved COOH-terminal 125-amino acid domain (which is located in the light chains of MAP1A, MAP1B, and MAP1S) and α1-syntrophin is direct and occurs through the pleckstrin homology domain 2 (PH2) and the postsynaptic density protein 95/disk large/zonula occludens-1 protein homology domain (PDZ) of α1-syntrophin. We confirmed the interaction of MAP1B and α1-syntrophin by co-localization of the two proteins in transfected cells and by co-immunoprecipitation experiments from mouse brain. In addition, we show that MAP1B and α1-syntrophin partially co-localize in Schwann cells of the murine sciatic nerve during postnatal development and in the adult. However, intracellular localization of α1-syntrophin and other Schwann cell proteins such as ezrin and dystrophin-related protein 2 (DRP2) and the localization of the axonal node of Ranvier-associated protein Caspr1/paranodin were not affected in MAP1B null mice. Our findings add to a growing body of evidence that classical MAPs are likely to be involved in signal transduction not only by directly modulating microtubule function, but also through their interaction with signal transduction proteins.


Subject(s)
Calcium-Binding Proteins/metabolism , Central Nervous System/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Muscle Proteins/metabolism , Peripheral Nervous System/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Central Nervous System/cytology , Cytoskeletal Proteins/metabolism , Mice , Microtubules/metabolism , Peripheral Nervous System/cytology , Protein Binding , Protein Transport , Schwann Cells/metabolism
9.
J Neurosci ; 30(23): 7804-16, 2010 Jun 09.
Article in English | MEDLINE | ID: mdl-20534829

ABSTRACT

c-Jun N-terminal kinases (JNKs) (comprising JNK1-3 isoforms) are members of the MAPK (mitogen-activated protein kinase) family, activated in response to various stimuli including growth factors and inflammatory cytokines. Their activation is facilitated by scaffold proteins, notably JNK-interacting protein-1 (JIP1). Originally considered to be mediators of neuronal degeneration in response to stress and injury, recent studies support a role of JNKs in early stages of neurite outgrowth, including adult axonal regeneration. However, the function of individual JNK isoforms, and their potential effector molecules, remained unknown. Here, we analyzed the role of JNK signaling during axonal regeneration from adult mouse dorsal root ganglion (DRG) neurons, combining pharmacological JNK inhibition and mice deficient for each JNK isoform and for JIP1. We demonstrate that neuritogenesis is delayed by lack of JNK2 and JNK3, but not JNK1. JNK signaling is further required for sustained neurite elongation, as pharmacological JNK inhibition resulted in massive neurite retraction. This function relies on JNK1 and JNK2. Neurite regeneration of jip1(-/-) DRG neurons is affected at both initiation and extension stages. Interestingly, activated JNKs (phospho-JNKs), as well as JIP1, are also present in the cytoplasm of sprouting or regenerating axons, suggesting a local action on cytoskeleton proteins. Indeed, we have shown that JNK1 and JNK2 regulate the phosphorylation state of microtubule-associated protein MAP1B, whose role in axonal regeneration was previously characterized. Moreover, lack of MAP1B prevents neurite retraction induced by JNK inhibition. Thus, signaling by individual JNKs is differentially implicated in the reorganization of the cytoskeleton, and neurite regeneration.


Subject(s)
Ganglia, Spinal/cytology , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Nerve Regeneration/physiology , Neurites/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Cells, Cultured , Female , Genotype , Isoenzymes , JNK Mitogen-Activated Protein Kinases/deficiency , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Phosphorylation , Polymerase Chain Reaction
10.
J Biol Chem ; 284(39): 26502-9, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19625254

ABSTRACT

Cytolinker proteins stabilize cells mechanically, regulate cytoskeleton dynamics, and provide scaffolds for signaling molecules. For plectin, the prototype of these proteins, an unusual diversity of isoforms has been reported, which show distinct expression patterns, subcellular localizations, and functions. Plectin has been shown to have important functions in skin and muscle, but little is known about its role in neural cells. To address this issue, we generated two knock-out mouse lines, one which was selectively lacking plectin 1c (P1c), the major isoform expressed in neural cells, and another in which plectin was conditionally deleted in neuronal precursor cells. Using isoform-specific antibodies, we found P1c to be expressed late in development and to associate with postsynaptic dendrites of central nervous system neurons, motorneurons of spinal cord, sciatic nerve axons, and Schwann cells. Motor nerve conduction velocity was found significantly reduced in sciatic nerve from P1c-deficient as well as from conditional knock-out mice. This defect was traceable to an increased number of motor nerve fibers with small cross-sectional areas; the thicknesses of axons and of myelin sheaths were unaffected. This is the first report demonstrating an important role of plectin in a major nerve function.


Subject(s)
Gene Targeting/methods , Motor Neurons/physiology , Neural Conduction/physiology , Plectin/metabolism , Animals , Central Nervous System/metabolism , Female , Genotype , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Microscopy, Fluorescence , Plectin/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Ranvier's Nodes/ultrastructure , Sciatic Nerve/metabolism , Sciatic Nerve/physiology , Spinal Cord/metabolism , Spinal Nerve Roots/ultrastructure
11.
Mol Cell Neurosci ; 36(2): 235-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17764972

ABSTRACT

We previously described the function of MAP1B in both turning and branching of regenerating neurites. Our results suggested implication of MAP1B in coupling of actin and microtubule movements, a hypothesis investigated here using DRG neurons and Schwann cells (SCs), which also transiently express MAP1B. Cell motility and cytoskeletal rearrangements were assessed before and after addition of lysophosphatidic acid (LPA), an extracellular signaling phospholipid triggering changes in actin distribution and cell morphology. First, we show that MAP1B is required for SC migration in vitro, extending our previous work on its function in growth cone motility. Second, LPA stimulation induces drastic retraction of processes from MAP1B-expressing cells in a two-step process: actin contraction, which is followed by microtubule backfolding. More importantly, we provide evidence that MAP1B is required for microtubule backfolding, thereby unravelling an important molecular mechanism implicated in coupling the movements of actin and microtubules during process retraction of neural cells.


Subject(s)
Actin Cytoskeleton/physiology , Ganglia, Spinal/cytology , Growth Cones/metabolism , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , Neurons/cytology , Schwann Cells/cytology , Analysis of Variance , Animals , Cell Movement/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Growth Cones/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Models, Biological , Neurons/drug effects , Schwann Cells/drug effects , Video Recording/methods
12.
Nat Cell Biol ; 9(9): 1035-45, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17704770

ABSTRACT

Treatment of cultured vertebrate neurons with nitric oxide leads to growth-cone collapse, axon retraction and the reconfiguration of axonal microtubules. We show that the light chain of microtubule-associated protein (MAP) 1B is a substrate for S-nitrosylation in vivo, in cultured cells and in vitro. S-nitrosylation occurs at Cys 2457 in the COOH terminus. Nitrosylation of MAP1B leads to enhanced interaction with microtubules and correlates with the inhibition of neuroblastoma cell differentiation. We further show, in dorsal root ganglion neurons, that MAP1B is necessary for neuronal nitric oxide synthase control of growth-cone size, growth-cone collapse and axon retraction. These results reveal an S-nitrosylation-dependent signal-transduction pathway that is involved in regulation of the axonal cytoskeleton and identify MAP1B as a major component of this pathway. We propose that MAP1B acts by inhibiting a microtubule- and dynein-based mechanism that normally prevents axon retraction.


Subject(s)
Axons/metabolism , Microtubule-Associated Proteins/metabolism , Nitric Oxide/metabolism , Signal Transduction/physiology , Animals , Axons/ultrastructure , Cells, Cultured , Cysteine/metabolism , Ganglia, Spinal/cytology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Neurons/cytology , Neurons/metabolism , Nitroso Compounds , Protein Conformation , Rats
13.
Exp Cell Res ; 313(8): 1602-14, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17391669

ABSTRACT

We characterized chicken erythrocyte and human platelet ferritin by biochemical studies and immunofluorescence. Erythrocyte ferritin was found to be a homopolymer of H-ferritin subunits, resistant to proteinase K digestion, heat stable, and contained iron. In mature chicken erythrocytes and human platelets, ferritin was localized at the marginal band, a ring-shaped peripheral microtubule bundle, and displayed properties of bona fide microtubule-associated proteins such as tau. Red blood cell ferritin association with the marginal band was confirmed by temperature-induced disassembly-reassembly of microtubules. During erythrocyte differentiation, ferritin co-localized with coalescing microtubules during marginal band formation. In addition, ferritin was found in the nuclei of mature erythrocytes, but was not detectable in those of bone marrow erythrocyte precursors. These results suggest that ferritin has a function in marginal band formation and possibly in protection of the marginal band from damaging effects of reactive oxygen species by sequestering iron in the mature erythrocyte. Moreover, our data suggest that ferritin and syncolin, a previously identified erythrocyte microtubule-associated protein, are identical. Nuclear ferritin might contribute to transcriptional silencing or, alternatively, constitute a ferritin reservoir.


Subject(s)
Blood Platelets/cytology , Erythrocytes/cytology , Ferritins/metabolism , Microtubules/metabolism , Amino Acid Sequence , Animals , Apoferritins/genetics , Apoferritins/metabolism , Blood Platelets/metabolism , Cell Differentiation , Chick Embryo , Chickens , Erythrocytes/metabolism , Erythropoiesis , Ferritins/genetics , Gene Library , Gene Silencing , Humans , Molecular Sequence Data , Reactive Oxygen Species/metabolism
14.
Biochim Biophys Acta ; 1763(10): 1011-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16996626

ABSTRACT

The microtubule-associated proteins MAP1A and MAP1B are related but distinct multi-subunit protein complexes that consist of heavy and light chains. The predominant forms of these complexes are homotypic, i.e. they consist of a MAP1A heavy chain associated with MAP1A light chains or a MAP1B heavy chain associated with MAP1B light chains, respectively. In addition, MAP1A and MAP1B can exchange subunits and form heterotypic complexes consisting of a MAP1A heavy chain associated with MAP1B light chains which might play a role in a transition period of neuronal differentiation. Here we extend previous findings by confirming that heterotypic MAP1B heavy chain-MAP1A light chain complexes also exist in the developing murine brain. We show that these complexes form through interaction of homologous domains conserved in heavy and light chains of MAP1A and MAP1B. Likewise, conserved domains of the MAP1A and MAP1B light chains account for formation of light chain heterodimers. By yeast 2-hybrid analysis we located the light chain binding domain on the heavy chain to amino acids 211-508, thereby defining a new functional subdomain.


Subject(s)
Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Subunits/chemistry , Animals , Cell Line , Conserved Sequence , Dimerization , Protein Binding , Rats , Structure-Activity Relationship , Transfection
15.
Proc Natl Acad Sci U S A ; 102(51): 18449-54, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16344482

ABSTRACT

Plectin, a typical cytolinker protein, is essential for skin and skeletal muscle integrity. It stabilizes cells mechanically, regulates cytoskeleton dynamics, and serves as a scaffolding platform for signaling molecules. A variety of isoforms expressed in different tissues and cell types account for this versatility. To uncover the role of plectin 1, the major isoform expressed in tissues of mesenchymal origin, against the background of all other variants, we raised plectin isoform 1-specific antibodies and generated isoform-deficient mice. In contrast to plectin-null mice (lacking all plectin isoforms), which die shortly after birth because of severe skin blistering, plectin isoform 1-deficient mice were viable at birth, had a normal lifespan, and did not display the skin blistering phenotype. However, dermal fibroblasts isolated from plectin 1-deficient mice exhibited abnormalities in their actin cytoskeleton and impaired migration potential. Similarly, plectin 1-deficient T cells isolated from nymph nodes showed diminished chemotactic migration in vitro. Most strikingly, in vivo we found that leukocyte infiltration during wound healing was reduced in the mutant mice. These data show a specific role of a cytolinker protein in immune cell motility. Single isoform-deficient mice thus represent a powerful tool to unravel highly specific functions of plectin variants.


Subject(s)
Cytosol/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Plectin/deficiency , Actins/metabolism , Animals , Cell Adhesion , Cell Line , Cell Movement , Cell Polarity , Connective Tissue/metabolism , Cytoskeleton/metabolism , Fibroblasts , Leukocytes/cytology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Plectin/genetics , Plectin/metabolism , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Skin Abnormalities/metabolism , Skin Abnormalities/pathology
16.
Behav Brain Res ; 164(2): 188-96, 2005 Nov 07.
Article in English | MEDLINE | ID: mdl-16102853

ABSTRACT

We investigated mice deficient for the microtubule-associated protein MAP1B, a cytoskeletal element highly expressed in the developing nervous system, for altered performance in behavior, learning, and memory. Using the multiple T-maze, the open field and the Morris water maze we found that mice homozygous for a deletion of the MAP1B gene demonstrate impaired locomotor activity most likely correlated to a lack of physical endurance in general. In contrast, there were no significant differences in cognitive function and memory retention. In addition, we performed electroretinography and observed a reduction of the a-wave amplitude in response to single flash, white light stimulation. Taken together, these data provide further evidence for an important role of MAP1B in synaptic neurotransmission.


Subject(s)
Exploratory Behavior/physiology , Microtubule-Associated Proteins/physiology , Motor Activity/physiology , Physical Endurance/physiology , Retina/physiology , Vision, Ocular/physiology , Animals , Electroretinography , Female , Homozygote , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Photic Stimulation , Stress, Physiological/physiopathology
17.
J Biol Chem ; 280(36): 31981-90, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16014633

ABSTRACT

In eukaryotes, the nuclear export of mRNA is mediated by nuclear export factor 1 (NXF1) receptors. Metazoans encode additional NXF1-related proteins of unknown function, which share homology and domain organization with NXF1. Some mammalian NXF1-related genes are expressed preferentially in the brain and are thought to participate in neuronal mRNA metabolism. To address the roles of NXF1-related factors, we studied the two mouse NXF1 homologues, mNXF2 and mNXF7. In neuronal cells, mNXF2, but not mNXF7, exhibited mRNA export activity similar to that of Tip-associated protein/NXF1. Surprisingly, mNXF7 incorporated into mobile particles in the neurites that contained poly(A) and ribosomal RNA and colocalized with Staufen1-containing transport granules, indicating a role in neuronal mRNA trafficking. Yeast two-hybrid interaction, coimmunoprecipitation, and in vitro binding studies showed that NXF proteins bound to brain-specific microtubule-associated proteins (MAP) such as MAP1B and the WD repeat protein Unrip. Both in vitro and in vivo, MAP1B also bound to NXF export cofactor U2AF as well as to Staufen1 and Unrip. These findings revealed a network of interactions likely coupling the export and cytoplasmic trafficking of mRNA. We propose a model in which MAP1B tethers the NXF-associated mRNA to microtubules and facilitates their translocation along dendrites while Unrip provides a scaffold for the assembly of these transport intermediates.


Subject(s)
Cytoplasm/metabolism , Nucleocytoplasmic Transport Proteins/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/physiology , Active Transport, Cell Nucleus/genetics , Animals , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cytoplasm/genetics , Genes, Reporter , Humans , Lipoproteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , RNA, Ribosomal/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Tumor Cells, Cultured , Two-Hybrid System Techniques
18.
J Biol Chem ; 280(3): 2257-65, 2005 Jan 21.
Article in English | MEDLINE | ID: mdl-15528209

ABSTRACT

The related high molecular mass microtubule-associated proteins (MAPs) MAP1A and MAP1B are predominantly expressed in the nervous system and are involved in axon guidance and synaptic function. MAP1B is implicated in fragile X mental retardation, giant axonal neuropathy, and ataxia type 1. We report the functional characterization of a novel member of the microtubule-associated protein 1 family, which we termed MAP1S (corresponding to sequence data bank entries for VCY2IP1 and C19ORF5). MAP1S contains the three hallmark domains of the microtubule-associated protein 1 family but hardly any additional sequences. It decorates neuronal microtubules and copurifies with tubulin from brain. MAP1S is synthesized as a precursor protein that is partially cleaved into heavy and light chains in a tissue-specific manner. Heavy and light chains interact to form the MAP1S complex. The light chain binds, bundles, and stabilizes microtubules and binds to actin. The heavy chain appears to regulate light chain activity. In contrast to MAP1A and MAP1B, MAP1S is expressed in a wide range of tissues in addition to neurons and represents the non-neuronal counterpart of this cytolinker family.


Subject(s)
Microtubule-Associated Proteins/metabolism , Actins/metabolism , Animals , Base Sequence , Brain/growth & development , DNA Primers , DNA, Complementary , Genome , Humans , Immunohistochemistry , Mice , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Molecular Weight , Purkinje Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
19.
J Neurochem ; 91(4): 1007-17, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15525354

ABSTRACT

G-protein-coupled receptors (GPCRs) are the largest group of cell surface molecules involved in signal transduction and are receptors for a wide variety of stimuli ranging from light, calcium and odourants to biogenic amines and peptides. It is assumed that systematic genomic data-mining has identified the overwhelming majority of all remaining GPCRs in the genome. Here we report the cloning of a novel orphan GPCR which was identified in a search for erythropoietin-induced genes in the brain as a strongly up-regulated gene. This unknown gene coded for a protein which had a seven-transmembrane topology and key features typical of GPCRs of the A family but a low overall identity to all known GPCRs. The protein, coded ee3, has an unusually high evolutionary conservation and is expressed in neurons in diverse areas of the CNS with relation to integrative functions or motor tasks. A yeast two-hybrid screen for interacting proteins revealed binding to the microtubule-associated protein (MAP) 1b. Coupling to MAP1a has been described for another cognate GPCR, the 5-hydroxytryptamine (5HT) 2a receptor. Surprisingly, we found complete colocalization of ee3 and the 5HT2a receptor. The interaction with MAP1b proved to be critical for the stability or folding of ee3 as in mice lacking MAP1b the ee3 protein was undetectable by immunohistochemistry, although messenger RNA levels remained unchanged. We propose that ee3 is a highly interesting new orphan GPCR with potential connections to erythropoietin and 5HT2a receptor signalling.


Subject(s)
Erythropoietin/pharmacology , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Brain/drug effects , Brain/metabolism , Cloning, Molecular , Humans , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Multigene Family , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Organ Specificity , Rats , Sequence Homology, Amino Acid , Two-Hybrid System Techniques , Up-Regulation/drug effects , Up-Regulation/genetics
20.
J Neurosci ; 24(32): 7204-13, 2004 Aug 11.
Article in English | MEDLINE | ID: mdl-15306655

ABSTRACT

During development, microtubule-associated protein 1B (MAP1B) is one of the earliest MAPs, preferentially localized in axons and growth cones, and plays a role in axonal outgrowth. Although generally downregulated in the adult, we have shown that MAP1B is constitutively highly expressed in adult dorsal root ganglia (DRGs) and associated with central sprouting and peripheral regeneration of these neurons. Mutant mice with a complete MAP1B null allele that survive until adulthood exhibit a reduced myelin sheath diameter and conductance velocity of peripheral axons and lack of the corpus callosum. Here, to determine the function of MAP1B in axonal regeneration, we used cultures of adult DRG explants and/or dissociated neurons derived from this map1b-/- mouse line. Whereas the overall length of regenerating neurites lacking MAP1B was similar to wild-type controls, our analysis revealed two main defects. First, map1b-/- neurites exhibited significantly (twofold) higher terminal and collateral branching. Second, the turning capacity of growth cones (i.e., "choice" of a proper orientation) was impaired. In addition, lack of MAP1B may affect the post-translational modification of tubulin polymers: quantitative analysis showed a reduced amount of acetylated microtubules within growth cones, whereas the distribution of tyrosinated or detyrosinated microtubules was normal. Both growth cone turning and axonal branch formation are known to involve local regulation of the microtubule network. Our results demonstrate that MAP1B plays a role in these processes during plastic changes in the adult. In particular, the data suggest MAP1B implication in the locally coordinated assembly of cytoskeletal components required for branching and straight directional axon growth.


Subject(s)
Axons/physiology , Ganglia, Spinal/cytology , Microtubule-Associated Proteins/physiology , Nerve Regeneration , Neurons/physiology , Animals , Axons/ultrastructure , Cells, Cultured , Ganglia, Spinal/ultrastructure , Growth Cones/physiology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubules/ultrastructure , Myelin Sheath/pathology , Neurites/ultrastructure , Neuronal Plasticity , Neurons/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...