Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
J Comp Neurol ; 525(14): 2991-3009, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28560734

ABSTRACT

This study explored why lesioned retinal ganglion cell (RGC) axons regenerate successfully in the zebrafish optic nerve despite the presence of Rtn4b, the homologue of the rat neurite growth inhibitor RTN4-A/Nogo-A. Rat Nogo-A and zebrafish Rtn4b possess characteristic motifs (M1-4) in the Nogo-A-specific region, which contains delta20, the most inhibitory region of rat Nogo-A. To determine whether zebrafish M1-4 is inhibitory as rat M1-4 and Nogo-A delta20, proteins were recombinantly expressed and used as substrates for zebrafish single cell RGCs, mouse hippocampal neurons and goldfish, zebrafish and chick retinal explants. When offered as homogenous substrates, neurites of hippocampal neurons and of zebrafish single cell RGCs were inhibited by zebrafish M1-4, rat M1-4, and Nogo-A delta20. Neurite length increased when zebrafish single cell RGCs were treated with receptor-type-specific antagonists and, respectively, with morpholinos (MO) against S1PR2 and S1PR5a-which represent candidate zebrafish Nogo-A receptors. In a stripe assay, however, where M1-4 lanes alternate with polylysine-(Plys)-only lanes, RGC axons from goldfish, zebrafish, and chick retinal explants avoided rat M1-4 but freely crossed zebrafish M1-4 lanes-suggesting that zebrafish M1-4 is growth permissive and less inhibitory than rat M1-4. Moreover, immunostainings and dot blots of optic nerve and myelin showed that expression of Rtn4b is very low in tissue and myelin at 3-5 days after lesion when axons regenerate. Thus, Rtn4b seems to represent no major obstacle for axon regeneration in vivo because it is less inhibitory for RGC axons from retina explants, and because of its low abundance.


Subject(s)
Axons/physiology , Myelin Proteins/metabolism , Nerve Regeneration , Nogo Proteins/metabolism , Optic Nerve Injuries/physiopathology , Optic Nerve/physiology , Retinal Ganglion Cells/physiology , Zebrafish Proteins/metabolism , Amino Acid Motifs , Animals , Cells, Cultured , Chick Embryo , Goldfish , Hippocampus/pathology , Hippocampus/physiopathology , Mice, Inbred C57BL , Myelin Proteins/chemistry , Myelin Sheath/metabolism , Neuronal Outgrowth/physiology , Nogo Proteins/chemistry , Nogo Receptors/antagonists & inhibitors , Nogo Receptors/metabolism , Optic Nerve/pathology , Optic Nerve Injuries/pathology , Rats , Retina/pathology , Retina/physiopathology , Tissue Culture Techniques , Tissue Scaffolds , Zebrafish , Zebrafish Proteins/chemistry
3.
Exp Neurol ; 289: 31-45, 2017 03.
Article in English | MEDLINE | ID: mdl-27993509

ABSTRACT

Reggie-1 and -2 (flotillins) reside at recycling vesicles and promote jointly with Rab11a the targeted delivery of cargo. Recycling is essential for synapse formation suggesting that reggies and Rab11a may regulate the development of spine synapses. Recycling vesicles provide cargo for dendritic growth and recycle surface glutamate receptors (AMPAR, GluA) for long-term potentiation (LTP) induced surface exposure. Here, we show reduced number of spine synapses and impairment of an in vitro correlate of LTP in hippocampal neurons from reggie-1 k.o. (Flot2-/-) mice maturating in culture. These defects apparently result from reduced trafficking of PSD-95 revealed by live imaging of 10 div reggie-1 k.o. (Flot2-/-) neurons and likely impairs co-transport of cargo destined for spines: N-cadherin and the glutamate receptors GluA1 and GluN1. Impaired cargo trafficking and fewer synapses also emerged in reggie-1 siRNA, reggie-2 siRNA, and reggie-1 and -2 siRNA-treated neurons and was in siRNA and k.o. neurons rescued by reggie-1-EGFP and CA-Rab11a-EGFP. While correlative expressional changes of specific synapse proteins were observed in reggie-1 k.o. (Flot2-/-) brains in vivo, this did not occur in neurons maturating in vitro. Our work suggests that reggie-1 and reggie-2 function at Rab11a recycling containers in the transport of PSD-95, N-cadherin, GluA1 and GluN1, and promote (together with significant signaling molecules) spine-directed trafficking, spine synapse formation and the in vitro correlate of LTP.


Subject(s)
Hippocampus/cytology , Long-Term Potentiation/physiology , Membrane Proteins/metabolism , Neurons/physiology , Receptors, AMPA/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Animals, Newborn , Cadherins/metabolism , Cells, Cultured , Disks Large Homolog 4 Protein , Endocytosis/drug effects , Endocytosis/genetics , Female , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Long-Term Potentiation/drug effects , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Protein Transport/genetics , Pyridinium Compounds/pharmacokinetics , Quaternary Ammonium Compounds/pharmacokinetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors , rab GTP-Binding Proteins/genetics
4.
Neurobiol Dis ; 51: 168-76, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23174179

ABSTRACT

The ability of fish retinal ganglion cells (RGCs) to regenerate their axons was shown to require the re-expression and function of the two proteins reggie-1 and -2. RGCs in mammals fail to upregulate reggie expression and to regenerate axons after lesion suggesting the possibility that induced upregulation might promote regeneration. In the present study, RGCs in adult rats were induced to express reggie-1 by intravitreal injection of adeno-associated viral vectors (AAV2/1) expressing reggie-1 (AAV.R1-EGFP) 14d prior to optic nerve crush. Four weeks later, GAP-43-positive regenerating axons had crossed the lesion and grown into the nerve at significantly higher numbers and length (up to 5mm) than the control transduced with AAV.EGFP. Consistently, after transduction with AAV.R1-EGFP as opposed to AAV.EGFP, primary RGCs in vitro grew long axons on chondroitin sulfate proteoglycan (CSPG) and Nogo-A, both glial cell-derived inhibitors of neurite growth, suggesting that reggie-1 can provide neurons with the ability to override inhibitors of neurite growth. This reggie-1-mediated enhancement of growth was reproduced in mouse hippocampal and N2a neurons which generated axons 40-60% longer than their control counterparts. This correlates with the reggie-1-dependent activation of Src and PI3 kinase (PI3K), of the Rho family GTPase Rac1 and downstream effectors such as cofilin. This increased growth also depends on TC10, the GTPase involved in cargo delivery to the growth cone. Thus, the upregulation of reggie-1 in mammalian neurons provides nerve cells with neuron-intrinsic properties required for axon growth and successful regeneration in the adult mammalian CNS.


Subject(s)
Axons/metabolism , Membrane Proteins/biosynthesis , Nerve Regeneration/physiology , Neurites/metabolism , Optic Nerve/metabolism , Animals , Blotting, Western , Mice , Rats , Rats, Wistar , Signal Transduction/physiology , Transduction, Genetic , Up-Regulation
5.
J Neurosci ; 31(49): 18013-25, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22159115

ABSTRACT

The role of prion protein (PrP) is insufficiently understood partially because PrP-deficient (-/-) neurons from C57BL/6J mice seem to differentiate normally and are functionally mildly impaired. Here, we reassessed this notion and, unexpectedly, discovered that PrP(-/-) hippocampal growth cones were abnormally small and poor in filopodia and cargo-containing vesicles. Based on our findings that PrP-PrP trans-interaction recruits E-cadherin to cell contact sites and reggie microdomains, and that reggies are essential for growth by regulating membrane trafficking, we reasoned that PrP and reggie might promote cargo (N-cadherin) delivery via PrP-reggie-connected signaling upon PrP activation (by PrP-Fc-induced trans-interaction). In wild-type but not PrP(-/-) neurons, PrP activation led to (1) enhanced PrP-reggie cocluster formation, (2) reggie-associated fyn and MAP kinase activation, (3) Exo70 and N-cadherin (cargo) recruitment to reggie, (4) the preference of the growth cone for PrP-Fc as substrate, and (5) longer neurites. Conversely, PrP-reggie-induced N-cadherin recruitment was blocked by mutant TC10, the GTPase downstream of reggie, triggering exocyst-assisted cargo delivery. This implies that PrP functions in reggie-mediated signaling and cargo trafficking, thus promoting growth cone complexity and vitality and thereby growth cone elongation.


Subject(s)
Cadherins/metabolism , Growth Cones/drug effects , Membrane Proteins/metabolism , Neurons/cytology , Prions/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Cells, Cultured , Exocytosis/drug effects , Exocytosis/genetics , Hippocampus/cytology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/drug effects , Neurites/physiology , Peptides/pharmacology , Prions/genetics , Protein Transport/drug effects , Protein Transport/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection/methods , Vesicular Transport Proteins/metabolism
6.
J Neurosci ; 29(49): 15489-98, 2009 Dec 09.
Article in English | MEDLINE | ID: mdl-20007473

ABSTRACT

In contrast to mammals, lesioned axons in the zebrafish (ZF) optic nerve regenerate and restore vision. This correlates with the absence of the NogoA-specific N-terminal domains from the ZF nogo/rtn-4 (reticulon-4) gene that inhibits regeneration in mammals. However, mammalian nogo/rtn-4 carries a second inhibitory C-terminal domain, Nogo-66, being 70% identical with ZF-Nogo66. The present study examines, (1) whether ZF-Nogo66 is inhibitory and effecting similar signaling pathways upon Nogo66-binding to the Nogo66 receptor NgR and its coreceptors, and (2) whether Rat-Nogo66 on fish, and ZF-Nogo66 on mouse neurons, cause inhibition via NgR. Our results from "outgrowth, collapse and contact assays" suggest, surprisingly, that ZF-Nogo66 is growth-permissive for ZF and mouse neurons, quite in contrast to its Rat-Nogo66 homolog which inhibits growth. The opposite effects of ZF- and Rat-Nogo66 are, in both fish and mouse, transmitted by GPI (glycosylphosphatidylinositol)-anchored receptors, including NgR. The high degree of sequence homology in the predicted binding site is consistent with the ability of ZF- and mammalian-Nogo66 to bind to NgRs of both species. Yet, Rat-Nogo66 elicits phosphorylation of the downstream effector cofilin whereas ZF-Nogo66 has no influence on cofilin phosphorylation--probably because of significantly different Rat- versus ZF-Nogo66 sequences outside of the receptor-binding region effecting, by speculation, recruitment of a different set of coreceptors or microdomain association of NgR. Thus, not only was the NogoA-specific domain lost in fish, but Nogo66, the second inhibitory domain in mammals, and its signaling upon binding to NgR, was modified so that ZF-Nogo/RTN-4 does not impair axon regeneration.


Subject(s)
Axons/physiology , Myelin Proteins/metabolism , Nerve Regeneration/physiology , Optic Nerve/physiology , Receptors, Cell Surface/metabolism , Zebrafish Proteins/metabolism , Actin Depolymerizing Factors/metabolism , Animals , Glycosylphosphatidylinositols/metabolism , Growth Cones/physiology , HeLa Cells , Hippocampus/physiology , Humans , In Vitro Techniques , Mice , Myelin Proteins/genetics , Neurites/physiology , Neurons/physiology , Nogo Proteins , Rats , Retina/physiology , Retinal Ganglion Cells/physiology , Signal Transduction , Species Specificity , Zebrafish
7.
J Neurosci ; 29(20): 6607-15, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19458231

ABSTRACT

The reggies/flotillins--proteins upregulated during axon regeneration in retinal ganglion cells (RGCs)--are scaffolding proteins of microdomains and involved in neuronal differentiation. Here, we show that reggies regulate axon regeneration in zebrafish (ZF) after optic nerve section (ONS) in vivo as well as axon/neurite extension in hippocampal and N2a neurons in vitro through signal transduction molecules modulating actin dynamics. ZF reggie-1a, -2a, and -2b downregulation by reggie-specific morpholino (Mo) antisense oligonucleotides directly after ONS significantly reduced ZF RGC axon regeneration: RGC axons from reggie Mo retinas were markedly reduced. Moreover, the number of axon-regenerating RGCs, identified by insertion of A488-coupled dextran, decreased by 69% in retinas 7 d after Mo application. At 10 and 14 d, RGCs decreased by 53 and 33%, respectively, in correlation with the gradual inactivation of the Mos. siRNA-mediated knockdown of reggie-1 and -2 inhibited the differentiation and axon/neurite extension in hippocampal and N2a neurons. N2a cells had significantly shorter filopodia, more cells had lamellipodia and fewer neurites, defects which were rescued by a reggie-1 construct without siRNA-binding sites. Furthermore, reggie knockdown strongly perturbed the balanced activation of the Rho family GTPases Rac1, RhoA, and cdc42, influenced the phosphorylation of cortactin and cofilin, the formation of the N-WASP, cortactin and Arp3 complex, and affected p38, Ras, ERK1/2 (extracellular signal-regulated kinases 1 and 2), and focal adhesion kinase activation. Thus, as suggested by their prominent re-expression after lesion, the reggies represent neuron-intrinsic factors for axon outgrowth and regeneration, being crucial for the coordinated assembly of signaling complexes regulating cytoskeletal remodeling.


Subject(s)
Cell Differentiation/physiology , Hippocampus/cytology , Membrane Proteins/metabolism , Nerve Regeneration/physiology , Neurons/physiology , Optic Nerve Injuries/physiopathology , Retina/pathology , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Dextrans , Down-Regulation/drug effects , Green Fluorescent Proteins/genetics , Immunoprecipitation , Insulin-Like Growth Factor I/pharmacology , Membrane Proteins/genetics , Mice , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Neuroblastoma , Neurons/drug effects , Oligodeoxyribonucleotides, Antisense/pharmacology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Organ Preservation Solutions , RNA, Small Interfering/metabolism , Retina/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection/methods , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Zebrafish , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism
8.
J Cell Biol ; 182(6): 1185-200, 2008 Sep 22.
Article in English | MEDLINE | ID: mdl-18809727

ABSTRACT

Receptor protein tyrosine phosphatase alpha (RPTPalpha) phosphatase activity is required for intracellular signaling cascades that are activated in motile cells and growing neurites. Little is known, however, about mechanisms that coordinate RPTPalpha activity with cell behavior. We show that clustering of neural cell adhesion molecule (NCAM) at the cell surface is coupled to an increase in serine phosphorylation and phosphatase activity of RPTPalpha. NCAM associates with T- and L-type voltage-dependent Ca(2+) channels, and NCAM clustering at the cell surface results in Ca(2+) influx via these channels and activation of NCAM-associated calmodulin-dependent protein kinase IIalpha (CaMKIIalpha). Clustering of NCAM promotes its redistribution to lipid rafts and the formation of a NCAM-RPTPalpha-CaMKIIalpha complex, resulting in serine phosphorylation of RPTPalpha by CaMKIIalpha. Overexpression of RPTPalpha with mutated Ser180 and Ser204 interferes with NCAM-induced neurite outgrowth, which indicates that neurite extension depends on NCAM-induced up-regulation of RPTPalpha activity. Thus, we reveal a novel function for a cell adhesion molecule in coordination of cell behavior with intracellular phosphatase activity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurites/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Animals , Calcium Channels/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Enzyme Activation , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , Neural Cell Adhesion Molecules/genetics , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/metabolism , Signal Transduction/physiology
9.
J Cell Biol ; 168(1): 127-39, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15623578

ABSTRACT

The neural cell adhesion molecule (NCAM) forms a complex with p59fyn kinase and activates it via a mechanism that has remained unknown. We show that the NCAM140 isoform directly interacts with the intracellular domain of the receptor-like protein tyrosine phosphatase RPTPalpha, a known activator of p59fyn. Whereas this direct interaction is Ca2+ independent, formation of the complex is enhanced by Ca2+-dependent spectrin cytoskeleton-mediated cross-linking of NCAM and RPTPalpha in response to NCAM activation and is accompanied by redistribution of the complex to lipid rafts. Association between NCAM and p59fyn is lost in RPTPalpha-deficient brains and is disrupted by dominant-negative RPTPalpha mutants, demonstrating that RPTPalpha is a link between NCAM and p59fyn. NCAM-mediated p59fyn activation is abolished in RPTPalpha-deficient neurons, and disruption of the NCAM-p59fyn complex in RPTPalpha-deficient neurons or with dominant-negative RPTPalpha mutants blocks NCAM-dependent neurite outgrowth, implicating RPTPalpha as a major phosphatase involved in NCAM-mediated signaling.


Subject(s)
Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurites/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins/metabolism , src-Family Kinases/metabolism , Animals , Animals, Newborn , CHO Cells , Calcium/metabolism , Cricetinae , Enzyme Activation , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Multiprotein Complexes , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/genetics , Neurites/ultrastructure , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fyn , Receptor-Like Protein Tyrosine Phosphatases, Class 4 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/physiology , Spectrin/metabolism , src-Family Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL