Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Neuron ; 76(6): 1091-107, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23259946

ABSTRACT

Neurites are the characteristic structural element of neurons that will initiate brain connectivity and elaborate information. Early in development, neurons are spherical cells but this symmetry is broken through the initial formation of neurites. This fundamental step is thought to rely on actin and microtubule dynamics. However, it is unclear which aspects of the complex actin behavior control neuritogenesis and which molecular mechanisms are involved. Here, we demonstrate that augmented actin retrograde flow and protrusion dynamics facilitate neurite formation. Our data indicate that a single family of actin regulatory proteins, ADF/Cofilin, provides the required control of actin retrograde flow and dynamics to form neurites. In particular, the F-actin severing activity of ADF/Cofilin organizes space for the protrusion and bundling of microtubules, the backbone of neurites. Our data reveal how ADF/Cofilin organizes the cytoskeleton to drive actin retrograde flow and thus break the spherical shape of neurons.


Subject(s)
Actin Depolymerizing Factors/physiology , Actins/metabolism , Cell Shape/physiology , Cerebral Cortex/embryology , Destrin/physiology , Growth Cones/metabolism , Neurites/metabolism , Animals , Biological Transport , Cell Growth Processes/physiology , Cells, Cultured , Cerebral Cortex/cytology , Hippocampus/cytology , Hippocampus/embryology , In Vitro Techniques , Mice , Mice, Knockout , Microtubules/physiology , Neurogenesis/physiology
2.
Semin Cell Dev Biol ; 22(8): 825-33, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21884814

ABSTRACT

Neuronal polarization, the formation of one long axon and several short dendrites, is an obligatory process to integrate and propagate information within the brain. Axon formation is the key event during neuronal polarization and is based on tightly regulated rearrangements of the cytoskeleton. Here, we discuss how the cytoskeleton drives neuronal polarization. First, we convey the role of the actin cytoskeleton and microtubules during axon formation. Second, we discuss different cytoskeletal binding and regulating proteins, which are essential to specify the axon. Finally, we outline plus end tracking proteins (+TIPs) as important regulators for neuronal polarization by mediating the interaction between the actin cytoskeleton and microtubules and compare this function to other polarity processes.


Subject(s)
Cell Polarity , Cytoskeleton/metabolism , Neurons/cytology
3.
J Neurosci ; 31(4): 1528-38, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21273437

ABSTRACT

Axon formation is a hallmark of initial neuronal polarization. This process is thought to be regulated by enhanced microtubule stability in the subsequent axon and changes in actin dynamics in the future axonal growth cone. Here, we show that the microtubule end-binding proteins cytoplasmic linker protein (CLIP)-115 and CLIP-170 were enriched in the axonal growth cone and extended into the actin-rich domain of the growth cone. CLIPs were necessary for axon formation and sufficient to induce an axon. The regulation of axonal microtubule stabilization by CLIPs enabled the protrusion of microtubules into the leading edge of the axonal growth cone. Moreover, CLIPs positively regulated growth cone dynamics and restrained actin arc formation, which was necessary for axon growth. In fact, in neurons without CLIP activity, axon formation was restored by actin destabilization or myosin II inhibition. Together, our data suggest that CLIPs enable neuronal polarization by controlling the stabilization of microtubules and growth cone dynamics.


Subject(s)
Growth Cones/physiology , Microtubule-Associated Proteins/physiology , Microtubules/physiology , Neoplasm Proteins/physiology , Actins/physiology , Actins/ultrastructure , Animals , Axons/physiology , Axons/ultrastructure , Cells, Cultured , Growth Cones/ultrastructure , Hippocampus/cytology , Rats
4.
J Neurosci ; 30(20): 6930-43, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20484635

ABSTRACT

Neuronal migration and axon growth, key events during neuronal development, require distinct changes in the cytoskeleton. Although many molecular regulators of polarity have been identified and characterized, relatively little is known about their physiological role in this process. To study the physiological function of Rac1 in neuronal development, we have generated a conditional knock-out mouse, in which Rac1 is ablated in the whole brain. Rac1-deficient cerebellar granule neurons, which do not express other Rac isoforms, showed impaired neuronal migration and axon formation both in vivo and in vitro. In addition, Rac1 ablation disrupts lamellipodia formation in growth cones. The analysis of Rac1 effectors revealed the absence of the Wiskott-Aldrich syndrome protein (WASP) family verprolin-homologous protein (WAVE) complex from the plasma membrane of knock-out growth cones. Loss of WAVE function inhibited axon growth, whereas overexpression of a membrane-tethered WAVE mutant partially rescued axon growth in Rac1-knock-out neurons. In addition, pharmacological inhibition of the WAVE complex effector Arp2/3 also reduced axon growth. We propose that Rac1 recruits the WAVE complex to the plasma membrane to enable actin remodeling necessary for axon growth.


Subject(s)
Cell Movement/physiology , Neurons/physiology , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac1 GTP-Binding Protein/metabolism , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Axons/drug effects , Axons/metabolism , Bromodeoxyuridine/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cerebellum/cytology , Cerebellum/growth & development , Cofilin 1/metabolism , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Growth Cones/drug effects , Growth Cones/metabolism , Ki-67 Antigen/metabolism , Luminescent Proteins/genetics , Mice , Mice, Knockout , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Organ Culture Techniques/methods , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Transfection/methods , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/metabolism
5.
Virol J ; 6: 213, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19951438

ABSTRACT

BACKGROUND: Hepatitis C virus (HCV) associated liver diseases may be related to apoptotic processes. Thus, we investigated the role of different HCV proteins in apoptosis induction as well as their potency to interact with different apoptosis inducing agents. METHODS AND RESULTS: The use of a tightly adjustable tetracycline (Tet)-dependent HCV protein expression cell system with the founder osteosarcoma cell line U-2 OS allowed switch-off and on of the endogenous production of HCV proteins. Analyzed were cell lines expressing the HCV polyprotein, the core protein, protein complexes of the core, envelope proteins E1, E2 and p7, and non-structural proteins NS3 and NS4A, NS4B or NS5A and NS5B. Apoptosis was measured mainly by the detection of hypodiploid apoptotic nuclei in the absence or presence of mitomycin C, etoposide, TRAIL and an agonistic anti-CD95 antibody. To further characterize cell death induction, a variety of different methods like fluorescence microscopy, TUNEL (terminal deoxynucleotidyl transferase (TdT)-catalyzed deoxyuridinephosphate (dUTP)-nick end labeling) assay, Annexin V staining, Western blot and caspase activation assays were included into our analysis.Two cell lines expressing the core protein but not the total polyprotein exerted a strong apoptotic effect, while the other cell lines did not induce any or only a slight effect by measuring the hypodiploid nuclei. Cell death induction was caspase-independent since it could not be blocked by zVAD-fmk. Moreover, caspase activity was absent in Western blot analysis and fluorometric assays while typical apoptosis-associated morphological features like the membrane blebbing and nuclei condensation and fragmentation could be clearly observed by microscopy. None of the HCV proteins influenced the apoptotic effect mediated via the mitochondrial apoptosis pathway while only the core protein enhanced death-receptor-mediated apoptosis. CONCLUSION: Our data showed a caspase-independent apoptosis-like effect of the core protein, which seems to be inhibited in the presence of further HCV proteins like the non structural (NS) proteins. This observation could be of relevance for the viral spread since induction of an apoptosis-like cell death by the core protein may have some impact on the release of the HCV particles from the host cell.


Subject(s)
Apoptosis , Hepacivirus/metabolism , Viral Core Proteins/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Diploidy , Enzyme Activation , Hepacivirus/genetics , Humans , Mitochondria/metabolism , Receptors, Death Domain/metabolism , Viral Core Proteins/genetics
6.
Nat Methods ; 5(7): 605-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18536722

ABSTRACT

Live imaging of the actin cytoskeleton is crucial for the study of many fundamental biological processes, but current approaches to visualize actin have several limitations. Here we describe Lifeact, a 17-amino-acid peptide, which stained filamentous actin (F-actin) structures in eukaryotic cells and tissues. Lifeact did not interfere with actin dynamics in vitro and in vivo and in its chemically modified peptide form allowed visualization of actin dynamics in nontransfectable cells.


Subject(s)
Actins/metabolism , Coloring Agents/chemistry , Peptides/chemistry , Staining and Labeling/methods , Animals , Cells, Cultured , Cytoskeleton/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Microscopy, Fluorescence , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
7.
J Cell Biol ; 180(3): 619-32, 2008 Feb 11.
Article in English | MEDLINE | ID: mdl-18268107

ABSTRACT

Axon formation is the initial step in establishing neuronal polarity. We examine here the role of microtubule dynamics in neuronal polarization using hippocampal neurons in culture. We see increased microtubule stability along the shaft in a single neurite before axon formation and in the axon of morphologically polarized cells. Loss of polarity or formation of multiple axons after manipulation of neuronal polarity regulators, synapses of amphids defective (SAD) kinases, and glycogen synthase kinase-3beta correlates with characteristic changes in microtubule turnover. Consistently, changing the microtubule dynamics is sufficient to alter neuronal polarization. Application of low doses of the microtubule-destabilizing drug nocodazole selectively reduces the formation of future dendrites. Conversely, low doses of the microtubule-stabilizing drug taxol shift polymerizing microtubules from neurite shafts to process tips and lead to the formation of multiple axons. Finally, local stabilization of microtubules using a photoactivatable analogue of taxol induces axon formation from the activated area. Thus, local microtubule stabilization in one neurite is a physiological signal specifying neuronal polarization.


Subject(s)
Cell Differentiation/physiology , Cell Polarity/physiology , Central Nervous System/embryology , Central Nervous System/metabolism , Microtubules/metabolism , Neurons/metabolism , Animals , Cell Differentiation/drug effects , Cell Polarity/drug effects , Cells, Cultured , Central Nervous System/ultrastructure , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Growth Cones/drug effects , Growth Cones/metabolism , Growth Cones/ultrastructure , Hippocampus/embryology , Hippocampus/metabolism , Hippocampus/ultrastructure , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Microtubules/drug effects , Microtubules/ultrastructure , Neurites/drug effects , Neurites/metabolism , Neurites/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Nocodazole/pharmacology , Paclitaxel/analogs & derivatives , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Tubulin Modulators/pharmacology
8.
J Neurosci ; 27(48): 13117-29, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-18045906

ABSTRACT

The establishment of polarity is an essential process in early neuronal development. Although a number of molecules controlling neuronal polarity have been identified, genetic evidence about their physiological roles in this process is mostly lacking. We analyzed the consequences of loss of Cdc42, a central regulator of polarity in multiple systems, on the polarization of mammalian neurons. Genetic ablation of Cdc42 in the brain led to multiple abnormalities, including striking defects in the formation of axonal tracts. Neurons from the Cdc42 null animals sprouted neurites but had a strongly suppressed ability to form axons both in vivo and in culture. This was accompanied by disrupted cytoskeletal organization, enlargement of the growth cones, and inhibition of filopodial dynamics. Axon formation in the knock-out neurons was rescued by manipulation of the actin cytoskeleton, indicating that the effects of Cdc42 ablation are exerted through modulation of actin dynamics. In addition, the knock-outs showed a specific increase in the phosphorylation (inactivation) of the Cdc42 effector cofilin. Furthermore, the active, nonphosphorylated form of cofilin was enriched in the axonal growth cones of wild-type, but not of mutant, neurons. Importantly, cofilin knockdown resulted in polarity defects quantitatively analogous to the ones seen after Cdc42 ablation. We conclude that Cdc42 is a key regulator of axon specification, and that cofilin is a physiological downstream effector of Cdc42 in this process.


Subject(s)
Actin Depolymerizing Factors/metabolism , Neurons/physiology , cdc42 GTP-Binding Protein/physiology , Actins/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/physiology , Axons/physiology , Cell Polarity/drug effects , Cell Polarity/physiology , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Embryo, Mammalian , Gene Expression Regulation/physiology , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Mice , Mice, Knockout , Microtubule-Associated Proteins , Phosphorylation , RNA Interference/physiology , cdc42 GTP-Binding Protein/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...