Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Res Int ; 2015: 863845, 2015.
Article in English | MEDLINE | ID: mdl-26697494

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a potent multifunctional protein that inhibits angiogenesis and has neurogenic and neuroprotective properties. Since the wet form of age-related macular degeneration is characterized by choroidal neovascularization (CNV), PEDF would be an ideal candidate to inhibit CNV and support retinal pigment epithelial (RPE) cells. However, its short half-life has precluded its clinical use. To deliver PEDF to the subretinal space, we transfected RPE cells with the PEDF gene using the Sleeping Beauty transposon system. Transfected cells expressed and secreted biologically active recombinant PEDF (rPEDF). In cultures of human umbilical vein endothelial cells, rPEDF reduced VEGF-induced cumulative sprouting by ≥47%, decreased migration by 77%, and increased rate of apoptosis at least 3.4 times. rPEDF induced neurite outgrowth in neuroblastoma cells and protected ganglion and photoreceptor cells in organotypic retinal cultures. In a rat model of CNV, subretinal transplantation of PEDF-transfected cells led to a reduction of the CNV area by 48% 14 days after transplantation and decreased clinical significant lesions by 55% and 40% after 7 and 14 days, respectively. We showed that transplantation of pigment epithelial cells overexpressing PEDF can restore a permissive subretinal environment for RPE and photoreceptor maintenance, while inhibiting choroidal blood vessel growth.


Subject(s)
Choroidal Neovascularization/genetics , Eye Proteins/genetics , Human Umbilical Vein Endothelial Cells/transplantation , Macular Degeneration/genetics , Nerve Growth Factors/genetics , Recombinant Proteins/genetics , Serpins/genetics , Animals , Apoptosis/genetics , Choroidal Neovascularization/pathology , Choroidal Neovascularization/therapy , DNA Transposable Elements/genetics , Eye Proteins/administration & dosage , Ganglion Cysts/genetics , Ganglion Cysts/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macular Degeneration/pathology , Macular Degeneration/therapy , Nerve Growth Factors/administration & dosage , Neurites/metabolism , Neurites/pathology , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Rats , Recombinant Proteins/administration & dosage , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Serpins/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
EMBO Rep ; 12(10): 1055-61, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21909076

ABSTRACT

R-spondins are secreted Wnt signalling agonists, which regulate embryonic patterning and stem cell proliferation, but whose mechanism of action is poorly understood. Here we show that R-spondins bind to the orphan G-protein-coupled receptors LGR4 and LGR5 by their Furin domains. Gain- and loss-of-function experiments in mammalian cells and Xenopus embryos indicate that LGR4 and LGR5 promote R-spondin-mediated Wnt/ß-catenin and Wnt/PCP signalling. R-spondin-triggered ß-catenin signalling requires Clathrin, while Wnt3a-mediated ß-catenin signalling requires Caveolin-mediated endocytosis, suggesting that internalization has a mechanistic role in R-spondin signalling.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Thrombospondins/metabolism , Wnt Signaling Pathway , Xenopus Proteins/metabolism , Animals , Cell Line , Clathrin/metabolism , Endocytosis , Gene Expression Regulation , HEK293 Cells , Hep G2 Cells , Humans , Mice , Protein Binding , Receptors, G-Protein-Coupled/genetics , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/genetics
3.
Invest Ophthalmol Vis Sci ; 52(8): 5400-7, 2011 Jul 23.
Article in English | MEDLINE | ID: mdl-21508100

ABSTRACT

PURPOSE: Genetic modification of cells before transplantation may allow the delivery of neuroprotective and other functional molecules to patients with neurodegenerative diseases. To avoid complications associated with virally transfected cells, we have explored the use of nonviral methods to insert genetic material into RPE cells. METHODS: After transfection with plasmids encoding different pigment epithelium-derived factor (PEDF) fusion proteins, transfected cells were established and passaged up to 100 times. Gene expression of PEDF, ZnT3, ZIP2, CRALBP, CATD, and ZO-1 was determined by RT-PCR. Secretion dynamics were analyzed using ELISA and a spheroid-based assay was used to confirm the anti-angiogenic activity of the recombinant PEDF. RESULTS: Transfection efficiency reached up to 98.7% with a plasmid encoding PEDF and enhanced green fluorescent protein (EGFP) separately and 87.2% with a plasmid encoding an EGFP-PEDF fusion. Immunoblotting revealed that transfected RPE cells express the appropriate PEDF or EGFP-PEDF. Expression of recombinant PEDF is stable, as shown by its secretion for the 2 years and the 100 passages the cells have been followed. PEDF expression was overexpressed and the transfected cells exhibited increased proliferation, up-regulation of ZnT3 and ZIP2, and inhibited sprouting in human umbilical vein endothelial cell spheroids. CONCLUSIONS: Genetic in vitro modification of pigment epithelial cells using nonviral transfection protocols should improve the potential therapeutic treatment of neurodegenerative diseases by transplantation of genetically modified cells without the disadvantages of virally mediated transfection. Here we have shown that genetically modified RPE cells overexpress a functional human recombinant PEDF, as evidenced by the autogenic regulation of proliferation, up-regulation of two distinct zinc transporters, and in vitro inhibition of endothelial cell sprouting.


Subject(s)
Cation Transport Proteins/metabolism , Eye Proteins/genetics , Gene Expression Regulation/physiology , Nerve Growth Factors/genetics , Retinal Pigment Epithelium/metabolism , Serpins/genetics , Transfection , Zinc/metabolism , Animals , Blotting, Western , Carrier Proteins , Cell Proliferation , Cells, Cultured , Electroporation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Plasmids , Rabbits , Recombinant Fusion Proteins/metabolism , Retinal Pigment Epithelium/cytology , Reverse Transcriptase Polymerase Chain Reaction
4.
Development ; 135(22): 3655-64, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18842812

ABSTRACT

The vertebrate embryonic vasculature develops from angioblasts, which are specified from mesodermal precursors and develop in close association with blood cells. The signals that regulate embryonic vasculogenesis and angiogenesis are incompletely understood. Here, we show that R-spondin 3 (Rspo3), a member of a novel family of secreted proteins in vertebrates that activate Wnt/beta-catenin signaling, plays a key role in these processes. In Xenopus embryos, morpholino antisense knockdown of Rspo3 induces vascular defects because Rspo3 is essential for regulating the balance between angioblast and blood cell specification. In mice, targeted disruption of Rspo3 leads to embryonic lethality caused by vascular defects. Specifically in the placenta, remodeling of the vascular plexus is impaired. In human endothelial cells, R-spondin signaling promotes proliferation and sprouting angiogenesis in vitro, indicating that Rspo3 can regulate endothelial cells directly. We show that vascular endothelial growth factor is an immediate early response gene and a mediator of R-spondin signaling. The results identify Rspo3 as a novel, evolutionarily conserved angiogenic factor in embryogenesis.


Subject(s)
Blood Cells/cytology , Cell Differentiation , Neovascularization, Physiologic , Signal Transduction , Thrombospondins/metabolism , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endothelial Cells/cytology , Female , Gene Expression Regulation, Developmental , Hematopoiesis , Mice , Placenta/metabolism , Thrombospondins/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Wnt Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism , beta Catenin/metabolism
5.
Genes Dev ; 21(4): 465-80, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17322405

ABSTRACT

Dickkopf-1 (Dkk1) is a secreted protein that negatively modulates the Wnt/beta catenin pathway. Lack of Dkk1 function affects head formation in frog and mice, supporting the idea that Dkk1 acts as a "head inducer" during gastrulation. We show here that lack of Dkk1 function accelerates internalization and rostral progression of the mesendoderm and that gain of function slows down both internalization and convergence extension, indicating a novel role for Dkk1 in modulating these movements. The motility phenotype found in the morphants is not observed in embryos in which the Wnt/beta catenin pathway is overactivated, and that dominant-negative Wnt proteins are not able to rescue the gastrulation movement defect induced by absence of Dkk1. These data strongly suggest that Dkk1 is acting in a beta catenin independent fashion when modulating gastrulation movements. We demonstrate that the glypican 4/6 homolog Knypek (Kny) binds to Dkk1 and that they are able to functionally interact in vivo. Moreover, Dkk1 regulation of gastrulation movements is kny dependent. Kny is a component of the Wnt/planar cell polarity (PCP) pathway. We found that indeed Dkk1 is able to activate this pathway in both Xenopus and zebrafish. Furthermore, concomitant alteration of the beta catenin and PCP activities is able to mimic the morphant accelerated cell motility phenotype. Our data therefore indicate that Dkk1 regulates gastrulation movement through interaction with LRP5/6 and Kny and coordinated modulations of Wnt/beta catenin and Wnt/PCP pathways.


Subject(s)
Gastrula/metabolism , Heparan Sulfate Proteoglycans/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Body Patterning/genetics , Cell Polarity/genetics , Embryo, Nonmammalian/chemistry , Embryo, Nonmammalian/metabolism , Gastrula/chemistry , Glypicans/metabolism , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/genetics , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/genetics , Transfection , Wnt Proteins/metabolism , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/analysis , Xenopus Proteins/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/analysis , Zebrafish Proteins/genetics , beta Catenin/metabolism
6.
Dev Cell ; 7(4): 525-34, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15469841

ABSTRACT

We have carried out a small pool expression screen for modulators of the Wnt/beta-catenin pathway and identified Xenopus R-spondin2 (Rspo2) as a secreted activator of this cascade. Rspo2 is coexpressed with and positively regulated by Wnt signals and synergizes with Wnts to activate beta-catenin. Analyses of functional interaction with components of the Wnt/beta-catenin pathway suggest that Rspo2 functions extracellularly at the level of receptor ligand interaction. In addition to activating the Wnt/beta-catenin pathway, Rspo2 overexpression blocks Activin, Nodal, and BMP4 signaling in Xenopus, raising the possibility that it may negatively regulate the TGF-beta pathway. Antisense Morpholino experiments in Xenopus embryos and RNAi experiments in HeLa cells reveal that Rspo2 is required for Wnt/beta-catenin signaling. In Xenopus embryos depleted of Rspo2, the muscle markers myoD and myf5 fail to be activated and later muscle development is impaired. Thus, Rspo2 functions in a positive feedback loop to stimulate the Wnt/beta-catenin cascade.


Subject(s)
Muscle Development , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Xenopus Proteins/metabolism , Xenopus/embryology , Animals , Blotting, Western , Cell Line , Culture Techniques/methods , Cytoskeletal Proteins , Extracellular Matrix Proteins , Genes, Reporter , HeLa Cells , Humans , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Luciferases/metabolism , Microinjections , Molecular Sequence Data , Neoplasm Proteins/chemistry , RNA/biosynthesis , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism , Trans-Activators , Wnt Proteins , Xenopus/genetics , Xenopus Proteins/chemistry , beta Catenin
7.
Gene ; 285(1-2): 279-86, 2002 Feb 20.
Article in English | MEDLINE | ID: mdl-12039055

ABSTRACT

Investigation of molecular mechanisms underlying early patterning of the nervous system is an important task of modern developmental biology. Previously, we identified a novel homeobox gene, Anf, that is expressed in the most anterior zone at the beginning of neuroectoderm specification. The expression pattern of Anf corresponds to primordia of the telencephalon and the rostral part of the diencephalon. In the present work, we investigated cis-regulation of expression of the Xenopus laevis Anf, Xanf-1. Two elements, highly conserved in Xenopus, chick and human, were identified within the Xanf-1 promoter region. The first element, located near position -500, is necessary for overall enhancement of the Xanf-1 expression. The second element, near position -200, is crucial for maintenance of the Xanf-1 expression at moderate levels and also for specific localization of the expression in the anterior neuroectoderm. Thus, the distal part of this element is responsible for suppression of Xanf-1 posterior to the normal expression domain of this gene. The data obtained corroborate with the Nieuwkoop two-signal model of neural induction. This model states that at the first step of induction, all neuroectoderm acquires potencies to develop toward forebrain structures, but later these potencies are suppressed in posterior regions.


Subject(s)
Homeodomain Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Xenopus Proteins , Animals , Base Sequence , Binding Sites/genetics , Brain/embryology , Brain/metabolism , Chickens , Conserved Sequence/genetics , Embryo, Nonmammalian/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Luciferases/genetics , Luciferases/metabolism , Microinjections , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...