Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (183)2022 05 18.
Article in English | MEDLINE | ID: mdl-35661653

ABSTRACT

The chick retina has long been an important model system in developmental neurobiology, with advantages including its large size, rapid development, and accessibility for visualization and experimental manipulations. However, its major technical limitation had been the lack of robust loss-of-function approaches for gene function analyses. This protocol describes a methodology of gene silencing in the developing chick retina that involves transgenic expression of artificial microRNAs (miRNAs) by using the Tol2 transposon system. In this approach, a Tol2 transposon plasmid that contains an expression cassette for the EmGFP (emerald green fluorescent protein) marker and artificial pre-miRNA sequences against a target gene is introduced into the embryonic chick retina with a Tol2 transposase expression construct by in ovo electroporation. In the transfected retinal cells, the transposase catalyzes the excision of the expression cassette from the transposon vector and its integration into host chromosomes, leading to the stable expression of miRNAs and the EmGFP protein. In our previous study, we have demonstrated that the expression of Nel, a glycoprotein that exerts multiple functions in neural development, can be significantly suppressed in the developing chick retina by using this technique. Our results indicate that this methodology induces a stable and robust suppression of gene expression and thus provides an efficient loss-of-function approach for studies of retinal development.


Subject(s)
MicroRNAs , Animals , Animals, Genetically Modified , Chickens/genetics , DNA Transposable Elements/genetics , Gene Transfer Techniques , MicroRNAs/genetics , MicroRNAs/metabolism , Retina/metabolism , Transposases/genetics
2.
Methods Mol Biol ; 2092: 91-108, 2020.
Article in English | MEDLINE | ID: mdl-31786784

ABSTRACT

The embryonic chick has long been a favorite model system for in vivo studies of vertebrate development. However, a major technical limitation of the chick embryo has been the lack of efficient loss-of-function approaches for analyses of gene functions. Here, we describe a methodology in which a transgene encoding artificial microRNA sequences is introduced into embryonic chick retinal cells by in ovo electroporation and integrated into the genome using the Tol2 transposon system. We show that this methodology can induce potent and stable suppression of gene expression. This technique therefore provides a rapid and robust loss-of-function approach for studies of gene function in the developing retina.


Subject(s)
Chickens/genetics , DNA Transposable Elements/genetics , Gene Expression/genetics , Retina/physiology , Animals , Chick Embryo , Electroporation/methods , Gene Expression Regulation, Developmental/genetics , Gene Transfer Techniques , Genome/genetics , MicroRNAs/genetics , Transgenes/genetics
3.
Development ; 146(4)2019 02 18.
Article in English | MEDLINE | ID: mdl-30745429

ABSTRACT

In mammals with binocular vision, retinal ganglion cell (RGC) axons from each eye project to eye-specific domains in the contralateral and ipsilateral dorsal lateral geniculate nucleus (dLGN), underpinning disparity-based stereopsis. Although domain-specific axon guidance cues that discriminate contralateral and ipsilateral RGC axons have long been postulated as a key mechanism for development of the eye-specific retinogeniculate projection, the molecular nature of such cues has remained elusive. Here, we show that the extracellular glycoprotein Nell2 (neural epidermal growth factor-like-like 2) is expressed in the dorsomedial region of the dLGN, which ipsilateral RGC axons terminate in and contralateral axons avoid. In Nell2 mutant mice, contralateral RGC axons abnormally invaded the ipsilateral domain of the dLGN, and ipsilateral axons terminated in partially fragmented patches, forming a mosaic pattern of contralateral and ipsilateral axon-termination zones. In vitro, Nell2 exerted inhibitory effects on contralateral, but not ipsilateral, RGC axons. These results provide evidence that Nell2 acts as a domain-specific positional label in the dLGN that discriminates contralateral and ipsilateral RGC axons, and that it plays essential roles in the establishment of the eye-specific retinogeniculate projection.


Subject(s)
Nerve Tissue Proteins/physiology , Vision, Ocular , Visual Pathways/physiology , Animals , Axon Guidance , Axons/metabolism , Geniculate Bodies/physiology , Genotype , Mice , Mice, Transgenic , Mutation , Retina/physiology , Retinal Ganglion Cells/physiology , Time Factors
4.
Development ; 145(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29439133

ABSTRACT

Genetic factors underlying the human limb abnormality congenital talipes equinovarus ('clubfoot') remain incompletely understood. The spontaneous autosomal recessive mouse 'peroneal muscular atrophy' mutant (PMA) is a faithful morphological model of human clubfoot. In PMA mice, the dorsal (peroneal) branches of the sciatic nerves are absent. In this study, the primary developmental defect was identified as a reduced growth of sciatic nerve lateral motor column (LMC) neurons leading to failure to project to dorsal (peroneal) lower limb muscle blocks. The pma mutation was mapped and a candidate gene encoding LIM-domain kinase 1 (Limk1) identified, which is upregulated in mutant lateral LMC motor neurons. Genetic and molecular analyses showed that the mutation acts in the EphA4-Limk1-Cfl1/cofilin-actin pathway to modulate growth cone extension/collapse. In the chicken, both experimental upregulation of Limk1 by electroporation and pharmacological inhibition of actin turnover led to defects in hindlimb spinal motor neuron growth and pathfinding, and mimicked the clubfoot phenotype. The data support a neuromuscular aetiology for clubfoot and provide a mechanistic framework to understand clubfoot in humans.


Subject(s)
Charcot-Marie-Tooth Disease/embryology , Clubfoot/embryology , Clubfoot/genetics , Lim Kinases/genetics , Mutation , Animals , Axons , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Chick Embryo , Chromosome Mapping , Clubfoot/pathology , Disease Models, Animal , Female , Hindlimb/abnormalities , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Motor Neurons/pathology , Muscle, Skeletal/abnormalities , Muscle, Skeletal/innervation , Peroneal Nerve/abnormalities , Phenotype , Pregnancy , Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Sciatic Nerve/abnormalities , Up-Regulation
5.
Cancer Sci ; 106(5): 656-64, 2015 May.
Article in English | MEDLINE | ID: mdl-25726761

ABSTRACT

Neural epidermal growth factor-like like (NELL) 1 and 2 constitute a family of multimeric and multimodular extracellular glycoproteins. Although the osteogenic effects of NELL1 and functions of NELL2 in neural development have been reported, their expression and functions in cancer are largely unknown. In this study, we examined expression of NELL1 and NELL2 in renal cell carcinoma (RCC) using clinical specimens and cell lines. We show that, whereas NELL1 and NELL2 proteins are strongly expressed in renal tubules in non-cancerous areas of RCC specimens, their expression is significantly downregulated in cancerous areas. Silencing of NELL1 and NELL2 mRNA expression was also detected in RCC cell lines. Analysis of NELL1/2 promoter methylation status indicated that the CpG islands in the NELL1 and NELL2 genes are hypermethylated in RCC cell lines. NELL1 and NELL2 bind to RCC cells, suggesting that these cells express a receptor for NELL1 and NELL2 that can transduce signals. Furthermore, we found that both NELL1 and NELL2 inhibit RCC cell migration, and NELL1 further inhibits RCC cell adhesion. These results suggest that silencing of NELL gene expression by promoter hypermethylation plays roles in RCC progression by affecting cancer cell behavior.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Nerve Tissue Proteins/metabolism , Aged , Azacitidine/pharmacology , Calcium-Binding Proteins , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor/drug effects , Cell Movement , CpG Islands , DNA Methylation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/metabolism , Male , Middle Aged , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic
6.
Mol Biol Cell ; 25(2): 234-44, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24258025

ABSTRACT

For correct functioning of the nervous system, the appropriate number and complement of neuronal cell types must be produced during development. However, the molecular mechanisms that regulate the production of individual classes of neurons are poorly understood. In this study, we investigate the function of the thrombospondin-1-like glycoprotein, Nel (neural epidermal growth factor [EGF]-like), in the generation of retinal ganglion cells (RGCs) in chicks. During eye development, Nel is strongly expressed in the presumptive retinal pigment epithelium and RGCs. Nel overexpression in the developing retina by in ovo electroporation increases the number of RGCs, whereas the number of displaced amacrine cells decreases. Conversely, knockdown of Nel expression by transposon-mediated introduction of RNA interference constructs results in decrease in RGC number and increase in the number of displaced amacrine cells. Modifications of Nel expression levels do not appear to affect proliferation of retinal progenitor cells, but they significantly alter the progression rate of RGC differentiation from the central retina to the periphery. Furthermore, Nel protects RGCs from apoptosis during retinal development. These results indicate that Nel positively regulates RGC production by promoting their differentiation and survival during development.


Subject(s)
Avian Proteins/genetics , Cell Differentiation/genetics , Glycoproteins/genetics , Retina/growth & development , Thrombospondins/metabolism , Animals , Apoptosis/genetics , Cell Survival/genetics , Chickens , Gene Expression Regulation, Developmental , Retina/metabolism , Retinal Ganglion Cells , Stem Cells/cytology , Stem Cells/metabolism , Thrombospondins/genetics
7.
J Biol Chem ; 287(5): 3282-91, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22157752

ABSTRACT

Nel (neural epidermal growth factor (EGF)-like molecule) is a multimeric, multimodular extracellular glycoprotein with heparin-binding activity and structural similarities to thrombospondin-1. Nel is predominantly expressed in the nervous system and has been implicated in neuronal proliferation and differentiation, retinal axon guidance, synaptic functions, and spatial learning. The Nel protein contains an N-terminal thrombospondin-1 (TSP-N) domain, five cysteine-rich domains, and six EGF-like domains. However, little is known about the functions of specific domains of the Nel protein. In this study, we have performed structure-function analysis of Nel, by using a series of expression constructs for different regions of the Nel protein. Our studies demonstrate that the TSP-N domain is responsible for homo-multimer formation of Nel and its heparin-binding activity. In vivo, Nel and related Nell1 are expressed in several regions of the mouse central nervous system with partly overlapping patterns. When they are expressed in the same cells in vitro, Nel and Nell1 can form hetero-multimers through the TSP-N domain, but they do not hetero-oligomerize with thrombospondin-1. Whereas both the TSP-N domain and cysteine-rich domains can bind to retinal axons in vivo, only the latter causes growth cone collapse in cultured retinal axons, suggesting that cysteine-rich domains interact with and activate an inhibitory axon guidance receptor. These results suggest that Nel interacts with a range of molecules through its different domains and exerts distinct functions.


Subject(s)
Central Nervous System/embryology , Gene Expression Regulation, Developmental/physiology , Growth Cones/metabolism , Nerve Tissue Proteins/biosynthesis , Protein Multimerization/physiology , Thrombospondin 1 , Animals , Chick Embryo , HEK293 Cells , Humans , Mice , Nerve Tissue Proteins/genetics , Protein Structure, Tertiary
8.
Mol Cell Neurosci ; 41(2): 113-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19249368

ABSTRACT

Nel is a glycoprotein containing five chordin-like and six epidermal growth factor-like domains and is strongly expressed in the nervous system. In this study, we have examined expression patterns and in vitro functions of Nel in the chicken retinotectal system. We have found that in the developing tectum, expression of Nel is localized in specific laminae that retinal axons normally do not enter, including the border between the retinorecipient and non-retinorecipient laminae. Nel-binding activity is detected on retinal axons both in vivo and in vitro, suggesting that retinal axons express a receptor for Nel. In vitro, Nel inhibits retinal axon outgrowth and induces growth cone collapse and axon retraction. These results indicate that Nel acts as an inhibitory guidance cue for retinal axons, and suggest its roles in the establishment of the lamina-specificity in the retinotectal projection.


Subject(s)
Axons/metabolism , Cell Movement/physiology , Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Retinal Ganglion Cells , Tectum Mesencephali/cytology , Animals , Axons/ultrastructure , Chick Embryo , Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Signal Transduction/physiology , Tissue Culture Techniques
9.
J Neurosci ; 26(23): 6269-81, 2006 Jun 07.
Article in English | MEDLINE | ID: mdl-16763034

ABSTRACT

Neuronal pentraxins (NPs) define a family of proteins that are homologous to C-reactive and acute-phase proteins in the immune system and have been hypothesized to be involved in activity-dependent synaptic plasticity. To investigate the role of NPs in vivo, we generated mice that lack one, two, or all three NPs. NP1/2 knock-out mice exhibited defects in the segregation of eye-specific retinal ganglion cell (RGC) projections to the dorsal lateral geniculate nucleus, a process that involves activity-dependent synapse formation and elimination. Retinas from mice lacking NP1 and NP2 had cholinergically driven waves of activity that occurred at a frequency similar to that of wild-type mice, but several other parameters of retinal activity were altered. RGCs cultured from these mice exhibited a significant delay in functional maturation of glutamatergic synapses. Other developmental processes, such as pathfinding of RGCs at the optic chiasm and hippocampal long-term potentiation and long-term depression, appeared normal in NP-deficient mice. These data indicate that NPs are necessary for early synaptic refinements in the mammalian retina and dorsal lateral geniculate nucleus. We speculate that NPs exert their effects through mechanisms that parallel the known role of short pentraxins outside the CNS.


Subject(s)
C-Reactive Protein/physiology , Geniculate Bodies/physiology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Retina/physiology , Synapses/physiology , Visual Pathways/physiology , Animals , Cells, Cultured , Geniculate Bodies/growth & development , Glutamic Acid/metabolism , Hippocampus/physiology , Mice , Mice, Knockout , Neuronal Plasticity , Retina/cytology , Retina/growth & development , Retinal Ganglion Cells/physiology , Synapses/metabolism , Synaptic Transmission/physiology , Visual Pathways/growth & development
10.
J Biol Chem ; 280(32): 29355-63, 2005 Aug 12.
Article in English | MEDLINE | ID: mdl-15955811

ABSTRACT

EphB6 is a unique member in the Eph family of receptor tyrosine kinases in that its kinase domain contains several alterations in conserved amino acids and is catalytically inactive. Although EphB6 is expressed both in a variety of embryonic and adult tissues, biological functions of this receptor are largely unknown. In the present study, we examined the function of EphB6 in cell adhesion and migration. We demonstrated that EphB6 exerted biphasic effects in response to different concentrations of the ephrin-B2 ligand; EphB6 promoted cell adhesion and migration when stimulated with low concentrations of ephrin-B2, whereas it induced repulsion and inhibited migration upon stimulation with high concentrations of ephrin-B2. A truncated EphB6 receptor lacking the cytoplasmic domain showed monophasic-positive effects on cell adhesion and migration, indicating that the cytoplasmic domain is essential for the negative effects. EphB6 is constitutively associated with the Src family kinase Fyn. High concentrations of ephrin-B2 induced tyrosine phosphorylation of EphB6 through an Src family kinase activity. These results indicate that EphB6 can both positively and negatively regulate cell adhesion and migration, and suggest that tyrosine phosphorylation of the receptor by an Src family kinase acts as the molecular switch for the functional transition.


Subject(s)
Receptor, EphB6/physiology , Animals , CHO Cells , Catalysis , Cell Adhesion , Cell Line , Cell Movement , Cricetinae , Cytoplasm/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Ephrin-B2/metabolism , Humans , Immunoblotting , Immunoprecipitation , Ligands , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn , Receptor, EphB6/chemistry , Time Factors , Transfection , Tyrosine/chemistry , src-Family Kinases/metabolism
11.
J Neurosci ; 24(10): 2542-50, 2004 Mar 10.
Article in English | MEDLINE | ID: mdl-15014130

ABSTRACT

EphA tyrosine kinases are thought to act as topographically specific receptors in the well-characterized projection map from the retina to the tectum. Here, we describe a loss-of-function analysis of EphA receptors in retinotectal mapping. Expressing patches of a cytoplasmically truncated EphA3 receptor in chick retina caused temporal axons to have reduced responsiveness to posterior tectal repellent activity in vitro and to shift more posteriorly within the map in vivo. A gene disruption of mouse EphA5, replacing the intracellular domain with beta-galactosidase, reduced in vitro responsiveness of temporal axons to posterior target membranes. It also caused map abnormalities in vivo, with temporal axons shifted posteriorly and nasal axons anteriorly, but with the entire target still filled by retinal axons. The anterior shift of nasal axons was not accompanied by increased responsiveness to tectal repellent activity, in contrast to the comparable anterior shift in ephrin-A knock-outs, helping to resolve a previous ambiguity in interpreting the ephrin gene knock-outs. The results show the functional requirement for endogenous EphA receptors in retinotectal mapping, show that the receptor intracellular domain is required for a forward signaling response to topographic cues, and provide new evidence for a role of axon competition in topographic mapping.


Subject(s)
Receptors, Eph Family/physiology , Retinal Ganglion Cells/metabolism , Superior Colliculi/metabolism , Visual Pathways/metabolism , Animals , Axons/metabolism , Axons/physiology , Chick Embryo , Gene Targeting , Genes, Reporter , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Mutant Strains , Receptor, EphA3/biosynthesis , Receptor, EphA3/genetics , Receptor, EphA3/physiology , Receptor, EphA5/biosynthesis , Receptor, EphA5/genetics , Receptor, EphA5/physiology , Receptors, Eph Family/deficiency , Receptors, Eph Family/genetics , Retinal Ganglion Cells/cytology , Sequence Deletion , Superior Colliculi/cytology , Visual Pathways/cytology
12.
Development ; 129(24): 5647-58, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12421705

ABSTRACT

Neural maps in the vertebrate central nervous system often show discontinuously segregated, domain-to-domain patterns. However, the molecular mechanism that establishes such maps is not well understood. Here we show that in the chicken olivocerebellar system, EphA receptors and ephrin-As are expressed with distinct levels and combinations in mapping domains. When ephrin-A2 is retrovirally overexpressed in the cerebellum, the olivocerebellar map is disrupted, excluding axons with high receptor activity from ectopic expression domains. Conversely, overexpression of a truncated EphA3 receptor in the cerebellum reduces endogenous ligand activity to undetectable levels and causes aberrant mapping, with high receptor axons invading high ligand domains. In vitro, ephrin-A2 inhibits outgrowth of inferior olive axons in a region-specific manner. These results suggest that Eph receptors and ephrins constitute domain-specific positional information, and the spatially accurate receptor-ligand interaction is essential to guide inferior olive axons to their correct target domains.


Subject(s)
Receptor, EphA2/metabolism , Receptor, EphA3/metabolism , Receptors, Eph Family/metabolism , Animals , Axons , Cell Line , Cell Membrane , Central Nervous System/embryology , Cerebellum/metabolism , Chickens , Humans , In Situ Hybridization , Ligands , Models, Biological , Protein Binding , Protein Structure, Tertiary , Purkinje Cells/metabolism , Receptor, EphA3/biosynthesis , Retroviridae/genetics , Time Factors
13.
Microsc Res Tech ; 59(1): 58-67, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12242697

ABSTRACT

The Eph family of receptor tyrosine kinases and their cell-presented ligands, the ephrins, are frequently overexpressed in a wide variety of cancers, including breast, small-cell lung and gastrointestinal cancers, melanomas, and neuroblastomas. In particular, one Eph family member, EphA2, is overexpressed in many cancers, including 40% of breast cancers. EphA2 can also transform breast epithelial cells in vitro to display properties commonly associated with the development of metastasis. Remarkably, the oncogenic properties of EphA2 contravene traditional dogma with regard to the oncogenic properties of a growth factor and its receptor tyrosine kinase: while stimulation of EphA2 by its ligand (ephrin-A1) results in EphA2 autophosphorylation, the stimulation reverses the oncogenic transformation. As will be discussed in this review, the apparent dependence of oncogenicity on the dephosphorylated state of EphA2 most probably reflects the unique nature of Eph signaling. In particular, oncogenecity may depend on the capacity of unactivated EphA2 to interact with a variety of signaling molecules. As well as acting in oncogenic transformation, a growing body of evidence supports the importance of the concerted actions of ephrins and Eph molecules in tumor angiogenesis. Genetic studies, using targeted mutagenesis in mice, reveal that ephrin-B1, ephrin-B2, and EphB4 are essential for the normal morphogenesis of the embryonic vasculature into a sophisticated network of arteries, veins, and capillaries. Initial studies indicate that these molecules are also angiogenic in tumors, and as such represent important new targets for the development of chemotherapeutic treatments.


Subject(s)
Ephrins/metabolism , Neoplasms/physiopathology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Transformation, Neoplastic , Female , Humans , Mice , Neovascularization, Pathologic
SELECTION OF CITATIONS
SEARCH DETAIL
...