Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Rep ; 29(11): 3605-3619.e10, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31825839

ABSTRACT

Ribosome assembly occurs mainly in the nucleolus, yet recent studies have revealed robust enrichment and translation of mRNAs encoding many ribosomal proteins (RPs) in axons, far away from neuronal cell bodies. Here, we report a physical and functional interaction between locally synthesized RPs and ribosomes in the axon. We show that axonal RP translation is regulated through a sequence motif, CUIC, that forms an RNA-loop structure in the region immediately upstream of the initiation codon. Using imaging and subcellular proteomics techniques, we show that RPs synthesized in axons join axonal ribosomes in a nucleolus-independent fashion. Inhibition of axonal CUIC-regulated RP translation decreases local translation activity and reduces axon branching in the developing brain, revealing the physiological relevance of axonal RP synthesis in vivo. These results suggest that axonal translation supplies cytoplasmic RPs to maintain/modify local ribosomal function far from the nucleolus in neurons.


Subject(s)
Axons/metabolism , Neurogenesis , Ribosomal Proteins/genetics , Ribosomes/metabolism , Animals , Axons/ultrastructure , Brain/cytology , Brain/growth & development , Brain/metabolism , Cells, Cultured , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Ribonucleic Acid , Ribosomal Proteins/metabolism , Ribosomes/genetics , Xenopus laevis
2.
Elife ; 82019 11 20.
Article in English | MEDLINE | ID: mdl-31746735

ABSTRACT

Extrinsic cues trigger the local translation of specific mRNAs in growing axons via cell surface receptors. The coupling of ribosomes to receptors has been proposed as a mechanism linking signals to local translation but it is not known how broadly this mechanism operates, nor whether it can selectively regulate mRNA translation. We report that receptor-ribosome coupling is employed by multiple guidance cue receptors and this interaction is mRNA-dependent. We find that different receptors associate with distinct sets of mRNAs and RNA-binding proteins. Cue stimulation of growing Xenopus retinal ganglion cell axons induces rapid dissociation of ribosomes from receptors and the selective translation of receptor-specific mRNAs. Further, we show that receptor-ribosome dissociation and cue-induced selective translation are inhibited by co-exposure to translation-repressive cues, suggesting a novel mode of signal integration. Our findings reveal receptor-specific interactomes and suggest a generalizable model for cue-selective control of the local proteome.


Subject(s)
Axons/physiology , RNA, Messenger/genetics , Receptors, Cell Surface/genetics , Xenopus laevis/genetics , Animals , Axons/metabolism , Protein Biosynthesis/genetics , Proteome/genetics , RNA-Binding Proteins/genetics , Retinal Ganglion Cells/metabolism , Ribosomes/genetics , Signal Transduction , Xenopus laevis/growth & development
3.
Nat Neurosci ; 19(12): 1592-1598, 2016 12.
Article in English | MEDLINE | ID: mdl-27643431

ABSTRACT

During nervous system development, neurons extend axons along well-defined pathways. The current understanding of axon pathfinding is based mainly on chemical signaling. However, growing neurons interact not only chemically but also mechanically with their environment. Here we identify mechanical signals as important regulators of axon pathfinding. In vitro, substrate stiffness determined growth patterns of Xenopus retinal ganglion cell axons. In vivo atomic force microscopy revealed a noticeable pattern of stiffness gradients in the embryonic brain. Retinal ganglion cell axons grew toward softer tissue, which was reproduced in vitro in the absence of chemical gradients. To test the importance of mechanical signals for axon growth in vivo, we altered brain stiffness, blocked mechanotransduction pharmacologically and knocked down the mechanosensitive ion channel piezo1. All treatments resulted in aberrant axonal growth and pathfinding errors, suggesting that local tissue stiffness, read out by mechanosensitive ion channels, is critically involved in instructing neuronal growth in vivo.


Subject(s)
Axons/metabolism , Brain/growth & development , Mechanotransduction, Cellular/physiology , Neurogenesis/physiology , Retina/metabolism , Visual Pathways/growth & development , Animals , Axons/pathology , Retinal Ganglion Cells , Xenopus laevis , Zebrafish
4.
Open Biol ; 6(4): 150218, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27248654

ABSTRACT

Endocytosis and local protein synthesis (LPS) act coordinately to mediate the chemotropic responses of axons, but the link between these two processes is poorly understood. The endosomal sorting complex required for transport (ESCRT) is a key regulator of cargo sorting in the endocytic pathway, and here we have investigated the role of ESCRT-II, a critical ESCRT component, in Xenopus retinal ganglion cell (RGC) axons. We show that ESCRT-II is present in RGC axonal growth cones (GCs) where it co-localizes with endocytic vesicle GTPases and, unexpectedly, with the Netrin-1 receptor, deleted in colorectal cancer (DCC). ESCRT-II knockdown (KD) decreases endocytosis and, strikingly, reduces DCC in GCs and leads to axon growth and guidance defects. ESCRT-II-depleted axons fail to turn in response to a Netrin-1 gradient in vitro and many axons fail to exit the eye in vivo These defects, similar to Netrin-1/DCC loss-of-function phenotypes, can be rescued in whole (in vitro) or in part (in vivo) by expressing DCC. In addition, ESCRT-II KD impairs LPS in GCs and live imaging reveals that ESCRT-II transports mRNAs in axons. Collectively, our results show that the ESCRT-II-mediated endocytic pathway regulates both DCC and LPS in the axonal compartment and suggest that ESCRT-II aids gradient sensing in GCs by coupling endocytosis to LPS.


Subject(s)
Axons/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Protein Biosynthesis , Receptors, Cell Surface/metabolism , Retina/metabolism , Xenopus Proteins/metabolism , Animals , Axons/drug effects , DCC Receptor , Endocytosis/drug effects , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/drug effects , Endosomes/metabolism , Gene Knockdown Techniques , Growth Cones/drug effects , Growth Cones/metabolism , Nerve Growth Factors/pharmacology , Netrin-1 , Phenotype , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Tumor Suppressor Proteins/pharmacology , Xenopus Proteins/genetics , Xenopus laevis
5.
Nat Neurosci ; 16(2): 166-73, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23292679

ABSTRACT

Cell adhesion molecules and diffusible cues both regulate axon pathfinding, yet how these two modes of signaling interact is poorly understood. The homophilic cell adhesion molecule NF-protocadherin (NFPC) is expressed in the mid-dorsal optic tract neuroepithelium and in the axons of developing retinal ganglion cells (RGC) in Xenopus laevis. Here we report that targeted disruption of NFPC function in RGC axons or the optic tract neuroepithelium results in unexpectedly localized pathfinding defects at the caudal turn in the mid-optic tract. Semaphorin 3A (Sema3A), which lies adjacent to this turn, stimulates rapid, protein synthesis-dependent increases in growth cone NFPC and its cofactor, TAF1, in vitro. In vivo, growth cones exhibit marked increases in NFPC translation reporter activity in this mid-optic tract region that are attenuated by blocking neuropilin-1 function. Our results suggest that translation-linked coupling between regionally localized diffusible cues and cell adhesion can help axons navigate discrete segments of the pathway.


Subject(s)
Axons/physiology , Cadherins/physiology , Cues , Protein Biosynthesis , Retinal Ganglion Cells/cytology , Signal Transduction/physiology , Visual Pathways/physiology , Xenopus Proteins/physiology , Analysis of Variance , Animals , Anisomycin/pharmacology , COS Cells , Cadherins/genetics , Chlorocebus aethiops , Electroporation , Embryo, Nonmammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Confocal , Neuroepithelial Cells/physiology , Organ Culture Techniques , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Protein Synthesis Inhibitors/pharmacology , Protocadherins , Retina/cytology , Semaphorin-3A/pharmacology , Signal Transduction/drug effects , Time Factors , Transcription Factors/metabolism , Transfection , Xenopus Proteins/genetics , Xenopus laevis
6.
Cell ; 148(4): 752-64, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22341447

ABSTRACT

Local protein synthesis plays a key role in regulating stimulus-induced responses in dendrites and axons. Recent genome-wide studies have revealed that thousands of different transcripts reside in these distal neuronal compartments, but identifying those with functionally significant roles presents a challenge. We performed an unbiased screen to look for stimulus-induced, protein synthesis-dependent changes in the proteome of Xenopus retinal ganglion cell (RGC) axons. The intermediate filament protein lamin B2 (LB2), normally associated with the nuclear membrane, was identified as an unexpected major target. Axonal ribosome immunoprecipitation confirmed translation of lb2 mRNA in vivo. Inhibition of lb2 mRNA translation in axons in vivo does not affect guidance but causes axonal degeneration. Axonal LB2 associates with mitochondria, and LB2-deficient axons exhibit mitochondrial dysfunction and defects in axonal transport. Our results thus suggest that axonally synthesized lamin B plays a crucial role in axon maintenance by promoting mitochondrial function.


Subject(s)
Axons/metabolism , Lamin Type B/metabolism , Mitochondria/metabolism , Retinal Ganglion Cells/metabolism , Xenopus laevis/embryology , Animals , Axonal Transport , Embryo, Nonmammalian/metabolism , Protein Biosynthesis , Xenopus laevis/metabolism
7.
Neuron ; 65(3): 341-57, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20159448

ABSTRACT

Regulated protein degradation via the ubiquitin-proteasome system (UPS) plays a central role in building synaptic connections, yet little is known about either which specific UPS components are involved or UPS targets in neurons. We report that inhibiting the UPS in developing Xenopus retinal ganglion cells (RGCs) with a dominant-negative ubiquitin mutant decreases terminal branching in the tectum but does not affect long-range navigation to the tectum. We identify Nedd4 as a prominently expressed E3 ligase in RGC axon growth cones and show that disrupting its function severely inhibits terminal branching. We further demonstrate that PTEN, a negative regulator of the PI3K pathway, is a key downstream target of Nedd4: not only does Nedd4 regulate PTEN levels in RGC growth cones, but also, the decrease of PTEN rescues the branching defect caused by Nedd4 inhibition. Together our data suggest that Nedd4-regulated PTEN is a key regulator of terminal arborization in vivo.


Subject(s)
Axons/physiology , Down-Regulation/physiology , Endosomal Sorting Complexes Required for Transport/metabolism , Growth Cones/physiology , PTEN Phosphohydrolase/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Arginine/genetics , Cell Line, Transformed , Down-Regulation/genetics , Electroporation/methods , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Humans , Immunoprecipitation/methods , Lysine/genetics , Microscopy, Confocal/methods , Mutation/genetics , Nedd4 Ubiquitin Protein Ligases , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Retina/cytology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Superior Colliculi/cytology , Tissue Culture Techniques , Transduction, Genetic/methods , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics , Xenopus , Xenopus Proteins
8.
Neuron ; 64(3): 355-366, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19914184

ABSTRACT

Engrailed transcription factors regulate the expression of guidance cues that pattern retinal axon terminals in the dorsal midbrain. They also act directly to guide axon growth in vitro. We show here that an extracellular En gradient exists in the tectum along the anterior-posterior axis. Neutralizing extracellular Engrailed in vivo with antibodies expressed in the tectum causes temporal axons to map aberrantly to the posterior tectum in chick and Xenopus. Furthermore, posterior membranes from wild-type tecta incubated with anti-Engrailed antibodies or posterior membranes from Engrailed-1 knockout mice exhibit diminished repulsive activity for temporal axons. Since EphrinAs play a major role in anterior-posterior mapping, we tested whether Engrailed cooperates with EphrinA5 in vitro. We find that Engrailed restores full repulsion to axons given subthreshold doses of EphrinA5. Collectively, our results indicate that extracellular Engrailed contributes to retinotectal mapping in vivo by modulating the sensitivity of growth cones to EphrinA.


Subject(s)
Axons/physiology , Chemotaxis/physiology , Extracellular Space/metabolism , Homeodomain Proteins/metabolism , Retina/physiology , Superior Colliculi/physiology , Animals , Chick Embryo , Growth Cones/physiology , Homeodomain Proteins/genetics , In Vitro Techniques , Mice , Mice, Knockout , Receptors, Eph Family/metabolism , Retina/embryology , Retina/growth & development , Retinal Ganglion Cells/physiology , Superior Colliculi/embryology , Superior Colliculi/growth & development , Visual Pathways/embryology , Visual Pathways/growth & development , Visual Pathways/physiology , Xenopus
9.
J Neurosci ; 28(1): 100-5, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18171927

ABSTRACT

NF-protocadherin (NFPC)-mediated cell-cell adhesion plays a critical role in vertebrate neural tube formation. NFPC is also expressed during the period of axon tract formation, but little is known about its function in axonogenesis. Here we have tested the role of NFPC and its cytosolic cofactor template-activating factor 1 (TAF1) in the emergence of the Xenopus retinotectal projection. NFPC is expressed in the developing retina and optic pathway and is abundant in growing retinal axons. Inhibition of NFPC function in developing retinal ganglion cells (RGCs) severely reduces axon initiation and elongation and suppresses dendrite genesis. Furthermore, an identical phenotype occurs when TAF1 function is blocked. These data provide evidence that NFPC regulates axon initiation and elongation and indicate a conserved role for TAF1, a transcriptional regulator, as a downstream cytosolic effector of NFPC in RGCs.


Subject(s)
Axons/physiology , Cadherins/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Retinal Ganglion Cells/cytology , Xenopus Proteins/physiology , Animals , Embryo, Nonmammalian , Green Fluorescent Proteins/metabolism , Mutation/genetics , Organ Culture Techniques , Protocadherins , Retina/cytology , Transfection/methods , Xenopus
10.
Development ; 135(2): 333-42, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18077591

ABSTRACT

Sphingosine 1-phosphate (S1P), a lysophospholipid, plays an important chemotactic role in the migration of lymphocytes and germ cells, and is known to regulate aspects of central nervous system development such as neurogenesis and neuronal migration. Its role in axon guidance, however, has not been examined. We show that sphingosine kinase 1, an enzyme that generates S1P, is expressed in areas surrounding the Xenopus retinal axon pathway, and that gain or loss of S1P function in vivo causes errors in axon navigation. Chemotropic assays reveal that S1P elicits fast repulsive responses in retinal growth cones. These responses require heparan sulfate, are sensitive to inhibitors of proteasomal degradation, and involve RhoA and LIM kinase activation. Together, the data identify downstream components that mediate S1P-induced growth cone responses and implicate S1P signalling in axon guidance.


Subject(s)
Axons/metabolism , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Visual Pathways/embryology , Visual Pathways/metabolism , Xenopus laevis/embryology , Animals , Axons/drug effects , Axons/enzymology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/enzymology , Enzyme Activation/drug effects , Gene Expression Regulation, Developmental/drug effects , Growth Cones/drug effects , Growth Cones/enzymology , Growth Cones/pathology , Heparitin Sulfate/metabolism , Humans , Lim Kinases/metabolism , Lysophospholipids/pharmacology , Mice , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proteasome Inhibitors , Receptors, Lysosphingolipid/metabolism , Retina/drug effects , Retina/enzymology , Retina/pathology , Signal Transduction/drug effects , Sphingosine/metabolism , Sphingosine/pharmacology , Visual Pathways/drug effects , Visual Pathways/enzymology , rhoA GTP-Binding Protein/metabolism
11.
BMC Dev Biol ; 7: 107, 2007 Sep 27.
Article in English | MEDLINE | ID: mdl-17900342

ABSTRACT

BACKGROUND: Blastomere injection of mRNA or antisense oligonucleotides has proven effective in analyzing early gene function in Xenopus. However, functional analysis of genes involved in neuronal differentiation and axon pathfinding by this method is often hampered by earlier function of these genes during development. Therefore, fine spatio-temporal control of over-expression or knock-down approaches is required to specifically address the role of a given gene in these processes. RESULTS: We describe here an electroporation procedure that can be used with high efficiency and low toxicity for targeting DNA and antisense morpholino oligonucleotides (MOs) into spatially restricted regions of the Xenopus CNS at a critical time-window of development (22-50 hour post-fertilization) when axonal tracts are first forming. The approach relies on the design of "electroporation chambers" that enable reproducible positioning of fixed-spaced electrodes coupled with accurate DNA/MO injection. Simple adjustments can be made to the electroporation chamber to suit the shape of different aged embryos and to alter the size and location of the targeted region. This procedure can be used to electroporate separate regions of the CNS in the same embryo allowing separate manipulation of growing axons and their intermediate and final targets in the brain. CONCLUSION: Our study demonstrates that electroporation can be used as a versatile tool to investigate molecular pathways involved in axon extension during Xenopus embryogenesis. Electroporation enables gain or loss of function studies to be performed with easy monitoring of electroporated cells. Double-targeted transfection provides a unique opportunity to monitor axon-target interaction in vivo. Finally, electroporated embryos represent a valuable source of MO-loaded or DNA transfected cells for in vitro analysis. The technique has broad applications as it can be tailored easily to other developing organ systems and to other organisms by making simple adjustments to the electroporation chamber.


Subject(s)
DNA, Complementary/genetics , Electroporation , Gene Expression Regulation, Developmental , Oligonucleotides, Antisense , Xenopus laevis/embryology , Zebrafish Proteins/genetics , Animals , Body Patterning/genetics , Central Nervous System/embryology , Embryo, Nonmammalian , Green Fluorescent Proteins , Transfection , Xenopus laevis/genetics
12.
Development ; 134(11): 2137-46, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17507414

ABSTRACT

The Enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) family of proteins is required for filopodia formation in growth cones and plays a crucial role in guidance cue-induced remodeling of the actin cytoskeleton. In vivo studies with pharmacological inhibitors of actin polymerization have previously provided evidence for the view that filopodia are needed for growth cone navigation in the developing visual pathway. Here we have re-examined this issue using an alternative strategy to generate growth cones without filopodia in vivo by artificially targeting Xena/XVASP (Xenopus homologs of Ena/VASP) proteins to mitochondria in retinal ganglion cells (RGCs). We used the specific binding of the EVH1 domain of the Ena/VASP family of proteins with the ligand motif FP4 to sequester the protein at the mitochondria surface. RGCs with reduced function of Xena/XVASP proteins extended fewer axons out of the eye and possessed dynamic lamellipodial growth cones missing filopodia that advanced slowly in the optic tract. Surprisingly, despite lacking filopodia, the axons navigated along the optic pathway without obvious guidance errors, indicating that the Xena/XVASP family of proteins and filopodial protrusions are non-essential for pathfinding in retinal axons. However, depletion of Xena/XVASP proteins severely impaired the ability of growth cones to form branches within the optic tectum, suggesting that this protein family, and probably filopodia, plays a key role in establishing terminal arborizations.


Subject(s)
Cell Adhesion Molecules/metabolism , Growth Cones/physiology , Microfilament Proteins/metabolism , Mitochondria/metabolism , Phosphoproteins/metabolism , Pseudopodia/physiology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Xenopus/embryology , Animals , Cells, Cultured , Green Fluorescent Proteins , Growth Cones/metabolism , Microscopy, Video
13.
Neuron ; 49(2): 215-28, 2006 Jan 19.
Article in English | MEDLINE | ID: mdl-16423696

ABSTRACT

Slits mediate multiple axon guidance decisions, but the mechanisms underlying the responses of growth cones to these cues remain poorly defined. We show here that collapse induced by Slit2-conditioned medium (Slit2-CM) in Xenopus retinal growth cones requires local protein synthesis (PS) and endocytosis. Slit2-CM elicits rapid activation of translation regulators and MAP kinases in growth cones, and inhibition of MAPKs or disruption of heparan sulfate blocks Slit2-CM-induced PS and repulsion. Interestingly, Slit2-CM causes a fast PS-dependent decrease in cytoskeletal F-actin concomitant with a PS-dependent increase in the actin-depolymerizing protein cofilin. Our findings reveal an unexpected link between Slit2 and cofilin in growth cones and suggest that local translation of actin regulatory proteins contributes to repulsion.


Subject(s)
Growth Cones/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Retina/physiology , Signal Transduction/physiology , Actin Depolymerizing Factors/physiology , Actins/metabolism , Animals , Axons/physiology , Endocytosis/drug effects , Endocytosis/physiology , Gene Expression Regulation/physiology , Growth Cones/ultrastructure , Heparitin Sulfate/physiology , Immunoprecipitation , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Leucine/metabolism , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/drug effects , Organ Culture Techniques , Protein Kinases/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Retina/ultrastructure , TOR Serine-Threonine Kinases , Xenopus laevis
14.
Nat Neurosci ; 8(10): 1301-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16172602

ABSTRACT

Axon growth is governed by the ability of growth cones to interpret attractive and repulsive guidance cues. Recent studies have shown that secreted signaling molecules known as morphogens can also act as axon guidance cues. Of the large family of Wnt signaling components, only Wnt4 and Wnt5 seem to participate directly in axon guidance. Here we show that secreted Frizzled-related protein 1 (SFRP1), a proposed Wnt signaling inhibitor, can directly modify and reorient the growth of chick and Xenopus laevis retinal ganglion cell axons. This activity does not require Wnt inhibition and is modulated by extracellular matrix molecules. Intracellularly, SFRP1 function requires G(alpha) protein activation, protein synthesis and degradation, and it is modulated by cyclic nucleotide levels. Because SFRP1 interacts with Frizzled-2 (Fz2) and interference with Fz2 expression abolishes growth cone responses to SFRP1, we propose a previously unknown function for this molecule: the ability to guide growth cone movement via the Fz2 receptor.


Subject(s)
Axons/physiology , Proteins/pharmacology , Receptors, G-Protein-Coupled/physiology , Retina/cytology , Retinal Ganglion Cells/cytology , Animals , Animals, Genetically Modified , Axons/drug effects , Brain/embryology , Brain/metabolism , Cells, Cultured , Chick Embryo , Dose-Response Relationship, Drug , Drug Interactions , Fibronectins/pharmacology , Immunohistochemistry/methods , Immunoprecipitation/methods , In Situ Hybridization/methods , Intracellular Signaling Peptides and Proteins , Laminin/pharmacology , Morpholines/pharmacology , Neurites/drug effects , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Proteins/genetics , Proteins/metabolism , Retinal Ganglion Cells/physiology , Time Factors , Visual Pathways/embryology , Visual Pathways/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...