Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 15(11): e18144, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37791581

ABSTRACT

Glioblastoma (GBM) remains the most malignant primary brain tumor, with a median survival rarely exceeding 2 years. Tumor heterogeneity and an immunosuppressive microenvironment are key factors contributing to the poor response rates of current therapeutic approaches. GBM-associated macrophages (GAMs) often exhibit immunosuppressive features that promote tumor progression. However, their dynamic interactions with GBM tumor cells remain poorly understood. Here, we used patient-derived GBM stem cell cultures and combined single-cell RNA sequencing of GAM-GBM co-cultures and real-time in vivo monitoring of GAM-GBM interactions in orthotopic zebrafish xenograft models to provide insight into the cellular, molecular, and spatial heterogeneity. Our analyses revealed substantial heterogeneity across GBM patients in GBM-induced GAM polarization and the ability to attract and activate GAMs-features that correlated with patient survival. Differential gene expression analysis, immunohistochemistry on original tumor samples, and knock-out experiments in zebrafish subsequently identified LGALS1 as a primary regulator of immunosuppression. Overall, our work highlights that GAM-GBM interactions can be studied in a clinically relevant way using co-cultures and avatar models, while offering new opportunities to identify promising immune-modulating targets.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Humans , Glioblastoma/pathology , Zebrafish , Galectin 1/genetics , Galectin 1/metabolism , Galectin 1/therapeutic use , Cell Line, Tumor , Macrophages/metabolism , Brain Neoplasms/pathology , Tumor Microenvironment/genetics
2.
Acta Neuropathol ; 144(3): 465-488, 2022 09.
Article in English | MEDLINE | ID: mdl-35895140

ABSTRACT

A 'GGGGCC' repeat expansion in the first intron of the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The exact mechanism resulting in these neurodegenerative diseases remains elusive, but C9 repeat RNA toxicity has been implicated as a gain-of-function mechanism. Our aim was to use a zebrafish model for C9orf72 RNA toxicity to identify modifiers of the ALS-linked phenotype. We discovered that the RNA-binding protein heterogeneous nuclear ribonucleoprotein K (HNRNPK) reverses the toxicity of both sense and antisense repeat RNA, which is dependent on its subcellular localization and RNA recognition, and not on C9orf72 repeat RNA binding. We observed HNRNPK cytoplasmic mislocalization in C9orf72 ALS patient fibroblasts, induced pluripotent stem cell (iPSC)-derived motor neurons and post-mortem motor cortex and spinal cord, in line with a disrupted HNRNPK function in C9orf72 ALS. In C9orf72 ALS/FTD patient tissue, we discovered an increased nuclear translocation, but reduced expression of ribonucleotide reductase regulatory subunit M2 (RRM2), a downstream target of HNRNPK involved in the DNA damage response. Last but not least, we showed that increasing the expression of HNRNPK or RRM2 was sufficient to mitigate DNA damage in our C9orf72 RNA toxicity zebrafish model. Overall, our study strengthens the relevance of RNA toxicity as a pathogenic mechanism in C9orf72 ALS and demonstrates its link with an aberrant DNA damage response, opening novel therapeutic avenues for C9orf72 ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Pick Disease of the Brain , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , DNA Damage , DNA Repeat Expansion/genetics , Frontotemporal Dementia/pathology , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Pick Disease of the Brain/genetics , RNA/metabolism , RNA, Antisense , Zebrafish/genetics , Zebrafish/metabolism
3.
Development ; 148(4)2021 02 19.
Article in English | MEDLINE | ID: mdl-33547133

ABSTRACT

Previous studies have shown that Vasohibin 1 (Vash1) is stimulated by VEGFs in endothelial cells and that its overexpression interferes with angiogenesis in vivo Recently, Vash1 was found to mediate tubulin detyrosination, a post-translational modification that is implicated in many cell functions, such as cell division. Here, we used the zebrafish embryo to investigate the cellular and subcellular mechanisms of Vash1 on endothelial microtubules during formation of the trunk vasculature. We show that microtubules within venous-derived secondary sprouts are strongly and selectively detyrosinated in comparison with other endothelial cells, and that this difference is lost upon vash1 knockdown. Vash1 depletion in zebrafish specifically affected secondary sprouting from the posterior cardinal vein, increasing endothelial cell divisions and cell number in the sprouts. We show that altering secondary sprout numbers and structure upon Vash1 depletion leads to defective lymphatic vessel formation and ectopic lymphatic progenitor specification in the zebrafish trunk.


Subject(s)
Cell Cycle Proteins/genetics , Embryonic Development/genetics , Lymphangiogenesis/genetics , Zebrafish/embryology , Zebrafish/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Conserved Sequence , Evolution, Molecular , Gene Expression Regulation, Developmental , Immunohistochemistry , Microtubules/metabolism , Models, Biological
4.
Development ; 146(16)2019 08 27.
Article in English | MEDLINE | ID: mdl-31375478

ABSTRACT

How developing vascular networks acquire the right balance of arteries, veins and lymphatic vessels to efficiently supply and drain tissues is poorly understood. In zebrafish embryos, the robust and regular 50:50 global balance of intersegmental veins and arteries that form along the trunk prompts the intriguing question of how does the organism keep 'count'? Previous studies have suggested that the ultimate fate of an intersegmental vessel (ISV) is determined by the identity of the approaching secondary sprout emerging from the posterior cardinal vein. Here, we show that the formation of a balanced trunk vasculature involves an early heterogeneity in endothelial cell behaviour and Notch signalling activity in the seemingly identical primary ISVs that is independent of secondary sprouting and flow. We show that Notch signalling mediates the local patterning of ISVs, and an adaptive flow-mediated mechanism subsequently fine-tunes the global balance of arteries and veins along the trunk. We propose that this dual mechanism provides the adaptability required to establish a balanced network of arteries, veins and lymphatic vessels.


Subject(s)
Body Patterning , Receptors, Notch/metabolism , Zebrafish/embryology , Animals , Arteries/embryology , Cell Polarity , Endothelial Cells/physiology , Genetic Heterogeneity , Lymphatic Vessels/embryology , Regional Blood Flow , Signal Transduction , Veins/embryology , Zebrafish/blood
5.
Elife ; 52016 04 13.
Article in English | MEDLINE | ID: mdl-27074663

ABSTRACT

Formation of a regularly branched blood vessel network is crucial in development and physiology. Here we show that the expression of the Notch ligand Dll4 fluctuates in individual endothelial cells within sprouting vessels in the mouse retina in vivo and in correlation with dynamic cell movement in mouse embryonic stem cell-derived sprouting assays. We also find that sprout elongation and branching associates with a highly differential phase pattern of Dll4 between endothelial cells. Stimulation with pathologically high levels of Vegf, or overexpression of Dll4, leads to Notch dependent synchronization of Dll4 fluctuations within clusters, both in vitro and in vivo. Our results demonstrate that the Vegf-Dll4/Notch feedback system normally operates to generate heterogeneity between endothelial cells driving branching, whilst synchronization drives vessel expansion. We propose that this sensitive phase transition in the behaviour of the Vegf-Dll4/Notch feedback loop underlies the morphogen function of Vegfa in vascular patterning.


Subject(s)
Brain Neoplasms/genetics , Endothelial Cells/metabolism , Glioblastoma/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Pathologic/genetics , Receptors, Notch/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adaptor Proteins, Signal Transducing , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Calcium-Binding Proteins , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Feedback, Physiological , Gene Expression Regulation , Genes, Reporter , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Notch/genetics , Retina/cytology , Retina/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology
6.
Nat Cell Biol ; 18(4): 443-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26928868

ABSTRACT

How vascular tubes build, maintain and adapt continuously perfused lumens to meet local metabolic needs remains poorly understood. Recent studies showed that blood flow itself plays a critical role in the remodelling of vascular networks, and suggested it is also required for the lumenization of new vascular connections. However, it is still unknown how haemodynamic forces contribute to the formation of new vascular lumens during blood vessel morphogenesis. Here we report that blood flow drives lumen expansion during sprouting angiogenesis in vivo by inducing spherical deformations of the apical membrane of endothelial cells, in a process that we have termed inverse blebbing. We show that endothelial cells react to these membrane intrusions by local and transient recruitment and contraction of actomyosin, and that this mechanism is required for single, unidirectional lumen expansion in angiogenic sprouts. Our work identifies inverse membrane blebbing as a cellular response to high external pressure. We show that in the case of blood vessels such membrane dynamics can drive local cell shape changes required for global tissue morphogenesis, shedding light on a pressure-driven mechanism of lumen formation in vertebrates.


Subject(s)
Blood Vessels/embryology , Morphogenesis , Neovascularization, Physiologic , Actomyosin/metabolism , Animals , Animals, Genetically Modified , Blood Vessels/cytology , Blood Vessels/metabolism , Endothelial Cells/metabolism , Female , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Regional Blood Flow , Time-Lapse Imaging , Zebrafish
7.
Nat Commun ; 6: 7264, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26081042

ABSTRACT

Sprouting angiogenesis drives blood vessel growth in healthy and diseased tissues. Vegf and Dll4/Notch signalling cooperate in a negative feedback loop that specifies endothelial tip and stalk cells to ensure adequate vessel branching and function. Current concepts posit that endothelial cells default to the tip-cell phenotype when Notch is inactive. Here we identify instead that the stalk-cell phenotype needs to be actively repressed to allow tip-cell formation. We show this is a key endothelial function of neuropilin-1 (Nrp1), which suppresses the stalk-cell phenotype by limiting Smad2/3 activation through Alk1 and Alk5. Notch downregulates Nrp1, thus relieving the inhibition of Alk1 and Alk5, thereby driving stalk-cell behaviour. Conceptually, our work shows that the heterogeneity between neighbouring endothelial cells established by the lateral feedback loop of Dll4/Notch utilizes Nrp1 levels as the pivot, which in turn establishes differential responsiveness to TGF-ß/BMP signalling.


Subject(s)
Activin Receptors, Type I/metabolism , Endothelium, Vascular/growth & development , Neuropilin-1/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Notch/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Activin Receptors, Type II , Animals , Growth Differentiation Factor 2/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Phenotype , Receptor, Transforming Growth Factor-beta Type I , Smad2 Protein/metabolism , Smad3 Protein/metabolism
9.
PLoS Biol ; 13(4): e1002125, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25884288

ABSTRACT

Patterning of functional blood vessel networks is achieved by pruning of superfluous connections. The cellular and molecular principles of vessel regression are poorly understood. Here we show that regression is mediated by dynamic and polarized migration of endothelial cells, representing anastomosis in reverse. Establishing and analyzing the first axial polarity map of all endothelial cells in a remodeling vascular network, we propose that balanced movement of cells maintains the primitive plexus under low shear conditions in a metastable dynamic state. We predict that flow-induced polarized migration of endothelial cells breaks symmetry and leads to stabilization of high flow/shear segments and regression of adjacent low flow/shear segments.


Subject(s)
Blood Vessels/cytology , Endothelium, Vascular/cytology , Animals , Cell Polarity , Models, Biological
10.
Biol Open ; 2(9): 882-90, 2013.
Article in English | MEDLINE | ID: mdl-24143274

ABSTRACT

The importance of the blood- and lymph vessels in the transport of essential fluids, gases, macromolecules and cells in vertebrates warrants optimal insight into the regulatory mechanisms underlying their development. Mouse and zebrafish models of lymphatic development are instrumental for gene discovery and gene characterization but are challenging for certain aspects, e.g. no direct accessibility of embryonic stages, or non-straightforward visualization of early lymphatic sprouting, respectively. We previously demonstrated that the Xenopus tadpole is a valuable model to study the processes of lymphatic development. However, a fluorescent Xenopus reporter directly visualizing the lymph vessels was lacking. Here, we created transgenic Tg(Flk1:eGFP) Xenopus laevis reporter lines expressing green fluorescent protein (GFP) in blood- and lymph vessels driven by the Flk1 (VEGFR-2) promoter. We also established a high-resolution fluorescent dye labeling technique selectively and persistently visualizing lymphatic endothelial cells, even in conditions of impaired lymph vessel formation or drainage function upon silencing of lymphangiogenic factors. Next, we applied the model to dynamically document blood and lymphatic sprouting and patterning of the initially avascular tadpole fin. Furthermore, quantifiable models of spontaneous or induced lymphatic sprouting into the tadpole fin were developed for dynamic analysis of loss-of-function and gain-of-function phenotypes using pharmacologic or genetic manipulation. Together with angiography and lymphangiography to assess functionality, Tg(Flk1:eGFP) reporter tadpoles readily allowed detailed lymphatic phenotyping of live tadpoles by fluorescence microscopy. The Tg(Flk1:eGFP) tadpoles represent a versatile model for functional lymph/angiogenomics and drug screening.

11.
Cell ; 154(3): 651-63, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911327

ABSTRACT

Vessel sprouting by migrating tip and proliferating stalk endothelial cells (ECs) is controlled by genetic signals (such as Notch), but it is unknown whether metabolism also regulates this process. Here, we show that ECs relied on glycolysis rather than on oxidative phosphorylation for ATP production and that loss of the glycolytic activator PFKFB3 in ECs impaired vessel formation. Mechanistically, PFKFB3 not only regulated EC proliferation but also controlled the formation of filopodia/lamellipodia and directional migration, in part by compartmentalizing with F-actin in motile protrusions. Mosaic in vitro and in vivo sprouting assays further revealed that PFKFB3 overexpression overruled the pro-stalk activity of Notch, whereas PFKFB3 deficiency impaired tip cell formation upon Notch blockade, implying that glycolysis regulates vessel branching.


Subject(s)
Endothelial Cells/metabolism , Glycolysis , Neovascularization, Physiologic , Phosphofructokinase-2/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/cytology , Female , Gene Deletion , Gene Silencing , Humans , Male , Mice , Mice, Inbred C57BL , Phosphofructokinase-2/genetics , Pseudopodia/metabolism , Zebrafish
12.
EMBO J ; 32(18): 2491-503, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-23955003

ABSTRACT

To supply tissues with nutrients and oxygen, the cardiovascular system forms a seamless, hierarchically branched, network of lumenized tubes. Here, we show that maintenance of patent vessel lumens requires the Bα regulatory subunit of protein phosphatase 2A (PP2A). Deficiency of Bα in zebrafish precludes vascular lumen stabilization resulting in perfusion defects. Similarly, inactivation of PP2A-Bα in cultured ECs induces tubulogenesis failure due to alteration of cytoskeleton dynamics, actomyosin contractility and maturation of cell-extracellular matrix (ECM) contacts. Mechanistically, we show that PP2A-Bα controls the activity of HDAC7, an essential transcriptional regulator of vascular stability. In the absence of PP2A-Bα, transcriptional repression by HDAC7 is abrogated leading to enhanced expression of the cytoskeleton adaptor protein ArgBP2. ArgBP2 hyperactivates RhoA causing inadequate rearrangements of the EC actomyosin cytoskeleton. This study unravels the first specific role for a PP2A holoenzyme in development: the PP2A-Bα/HDAC7/ArgBP2 axis maintains vascular lumens by balancing endothelial cytoskeletal dynamics and cell-matrix adhesion.


Subject(s)
Endothelium, Vascular/physiology , Gene Expression Regulation/physiology , Histone Deacetylases/metabolism , Neovascularization, Physiologic/physiology , Protein Phosphatase 2/metabolism , Vascular Patency/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Adhesion/physiology , Collagen , Drug Combinations , Fluorescent Antibody Technique , Gene Expression Regulation/genetics , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Image Processing, Computer-Assisted , Laminin , Microscopy, Confocal , Proteoglycans , RNA, Small Interfering/genetics , RNA-Binding Proteins , Vascular Patency/genetics , Zebrafish
13.
Development ; 138(21): 4569-83, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21965610

ABSTRACT

The correct development of blood vessels is crucial for all aspects of tissue growth and physiology in vertebrates. The formation of an elaborate hierarchically branched network of endothelial tubes, through either angiogenesis or vasculogenesis, relies on a series of coordinated morphogenic events, but how individual endothelial cells adopt specific phenotypes and how they coordinate their behaviour during vascular patterning is unclear. Recent progress in our understanding of blood vessel formation has been driven by advanced imaging techniques and detailed analyses that have used a combination of powerful in vitro, in vivo and in silico model systems. Here, we summarise these models and discuss their advantages and disadvantages. We then review the different stages of blood vessel development, highlighting the cellular mechanisms and molecular players involved at each step and focusing on cell specification and coordination within the network.


Subject(s)
Cell Differentiation/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Models, Biological , Neovascularization, Physiologic/physiology , Animals , Humans , Morphogenesis/physiology
14.
J Neurosci ; 30(45): 15052-66, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21068311

ABSTRACT

Vascular endothelial growth factor (VEGF) regulates angiogenesis, but also has important, yet poorly characterized roles in neuronal wiring. Using several genetic and in vitro approaches, we discovered a novel role for VEGF in the control of cerebellar granule cell (GC) migration from the external granule cell layer (EGL) toward the Purkinje cell layer (PCL). GCs express the VEGF receptor Flk1, and are chemoattracted by VEGF, whose levels are higher in the PCL than EGL. Lowering VEGF levels in mice in vivo or ectopic VEGF expression in the EGL ex vivo perturbs GC migration. Using GC-specific Flk1 knock-out mice, we provide for the first time in vivo evidence for a direct chemoattractive effect of VEGF on neurons via Flk1 signaling. Finally, using knock-in mice expressing single VEGF isoforms, we show that pericellular deposition of matrix-bound VEGF isoforms around PC dendrites is necessary for proper GC migration in vivo. These findings identify a previously unknown role for VEGF in neuronal migration.


Subject(s)
Cell Movement/physiology , Cerebellum/physiology , Neurons/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Cells, Cultured , Cerebellum/cytology , Enzyme-Linked Immunosorbent Assay , Growth Cones/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Neurons/cytology , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
15.
Blood ; 116(17): 3356-66, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20631376

ABSTRACT

The molecular basis of lymphangiogenesis remains incompletely characterized. Here, we document a novel role for the PDZ domain-containing scaffold protein synectin in lymphangiogenesis using genetic studies in zebrafish and tadpoles. In zebrafish, the thoracic duct arises from parachordal lymphangioblast cells, which in turn derive from secondary lymphangiogenic sprouts from the posterior cardinal vein. Morpholino knockdown of synectin in zebrafish impaired formation of the thoracic duct, due to selective defects in lymphangiogenic but not angiogenic sprouting. Synectin genetically interacted with Vegfr3 and neuropilin-2a in regulating lymphangiogenesis. Silencing of synectin in tadpoles caused lymphatic defects due to an underdevelopment and impaired migration of Prox-1(+) lymphatic endothelial cells. Molecular analysis further revealed that synectin regulated Sox18-induced expression of Prox-1 and vascular endothelial growth factor C-induced migration of lymphatic endothelial cells in vitro. These findings reveal a novel role for synectin in lymphatic development.


Subject(s)
Carrier Proteins/metabolism , Lymphangiogenesis , Lymphatic Vessels/physiology , Xenopus Proteins/metabolism , Xenopus laevis/physiology , Zebrafish/physiology , Animals , Carrier Proteins/genetics , Cell Line , Gene Expression Regulation, Developmental , Gene Silencing , Humans , Larva/genetics , Larva/physiology , Neovascularization, Physiologic , Neuropilin-2/genetics , Thoracic Duct/embryology , Thoracic Duct/growth & development , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
16.
Arterioscler Thromb Vasc Biol ; 30(9): 1695-702, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20466977

ABSTRACT

OBJECTIVE: To study whether Notch signaling, which regulates cell fate decisions and vessel morphogenesis, controls lymphatic development. METHODS AND RESULTS: In zebrafish embryos, sprouts from the axial vein have lymphangiogenic potential because they give rise to the first lymphatics. Knockdown of delta-like-4 (Dll4) or its receptors Notch-1b or Notch-6 in zebrafish impaired lymphangiogenesis. Dll4/Notch silencing reduced the number of sprouts producing the string of parchordal lymphangioblasts; instead, sprouts connecting to the intersomitic vessels were formed. At a later phase, Notch silencing impaired navigation of lymphatic intersomitic vessels along their arterial templates. CONCLUSIONS: These studies imply critical roles for Notch signaling in the formation and wiring of the lymphatic network.


Subject(s)
Lymphangiogenesis , Lymphatic System/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Biomarkers/metabolism , COS Cells , Cell Movement , Cell Proliferation , Chlorocebus aethiops , Coculture Techniques , Embryo, Nonmammalian/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Gene Silencing , Humans , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Lymphangiogenesis/genetics , Lymphatic System/embryology , Membrane Proteins/genetics , RNA, Messenger/metabolism , Receptors, Notch/genetics , Thoracic Duct/embryology , Thoracic Duct/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
17.
Blood ; 112(5): 1740-9, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18474726

ABSTRACT

The importance of the lymphangiogenic factor VEGF-D and its receptor VEGFR-3 in early lymphatic development remains largely unresolved. We therefore investigated their role in Xenopus laevis tadpoles, a small animal model allowing chemicogenetic dissection of developmental lymphangiogenesis. Single morpholino antisense oligo knockdown of xVEGF-D did not affect lymphatic commitment, but transiently impaired lymphatic endothelial cell (LEC) migration. Notably, combined knockdown of xVEGF-D with xVEGF-C at suboptimal morpholino concentrations resulted in more severe migration defects and lymphedema formation than the corresponding single knockdowns. Knockdown of VEGFR-3 or treatment with the VEGFR-3 inhibitor MAZ51 similarly impaired lymph vessel formation and function and caused pronounced edema. VEGFR-3 silencing by morpholino knockdown, MAZ51 treatment, or xVEGF-C/D double knockdown also resulted in dilation and dysfunction of the lymph heart. These findings document a critical role of VEGFR-3 in embryonic lymphatic development and function, and reveal a previously unrecognized modifier role of VEGF-D in the regulation of embryonic lymphangiogenesis in frog embryos.


Subject(s)
Lymphangiogenesis/physiology , Vascular Endothelial Growth Factor D/physiology , Vascular Endothelial Growth Factor Receptor-3/physiology , Xenopus Proteins/physiology , Xenopus laevis/growth & development , Xenopus laevis/physiology , Animals , Gene Silencing , Larva/growth & development , Lymphangiogenesis/genetics , Lymphatic Vessels/abnormalities , Lymphatic Vessels/embryology , Oligodeoxyribonucleotides, Antisense/genetics , Vascular Endothelial Growth Factor D/antagonists & inhibitors , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...