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










Publication year range
1.
Nat Commun ; 15(1): 4575, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834586

ABSTRACT

Bone regeneration requires a well-orchestrated cellular and molecular response including robust vascularization and recruitment of mesenchymal and osteogenic cells. In femoral fractures, angiogenesis and osteogenesis are closely coupled during the complex healing process. Here, we show with advanced longitudinal intravital multiphoton microscopy that early vascular sprouting is not directly coupled to osteoprogenitor invasion during calvarial bone regeneration. Early osteoprogenitors emerging from the periosteum give rise to bone-forming osteoblasts at the injured calvarial bone edge. Microvessels growing inside the lesions are not associated with osteoprogenitors. Subsequently, osteogenic cells collectively invade the vascularized and perfused lesion as a multicellular layer, thereby advancing regenerative ossification. Vascular sprouting and remodeling result in dynamic blood flow alterations to accommodate the growing bone. Single cell profiling of injured calvarial bones demonstrates mesenchymal stromal cell heterogeneity comparable to femoral fractures with increase in cell types promoting bone regeneration. Expression of angiogenesis and hypoxia-related genes are slightly elevated reflecting ossification of a vascularized lesion site. Endothelial Notch and VEGF signaling alter vascular growth in calvarial bone repair without affecting the ossification progress. Our findings may have clinical implications for bone regeneration and bioengineering approaches.


Subject(s)
Bone Regeneration , Mesenchymal Stem Cells , Neovascularization, Physiologic , Osteogenesis , Skull , Animals , Bone Regeneration/physiology , Mice , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Male , Receptors, Notch/metabolism , Receptors, Notch/genetics , Mice, Inbred C57BL , Signal Transduction , Female , Angiogenesis
2.
Nat Commun ; 15(1): 2539, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38570531

ABSTRACT

Cell segregation allows the compartmentalization of cells with similar fates during morphogenesis, which can be enhanced by cell fate plasticity in response to local molecular and biomechanical cues. Endothelial tip cells in the growing retina, which lead vessel sprouts, give rise to arterial endothelial cells and thereby mediate arterial growth. Here, we have combined cell type-specific and inducible mouse genetics, flow experiments in vitro, single-cell RNA sequencing and biochemistry to show that the balance between ephrin-B2 and its receptor EphB4 is critical for arterial specification, cell sorting and arteriovenous patterning. At the molecular level, elevated ephrin-B2 function after loss of EphB4 enhances signaling responses by the Notch pathway, VEGF and the transcription factor Dach1, which is influenced by endothelial shear stress. Our findings reveal how Eph-ephrin interactions integrate cell segregation and arteriovenous specification in the vasculature, which has potential relevance for human vascular malformations caused by EPHB4 mutations.


Subject(s)
Endothelial Cells , Ephrins , Mice , Humans , Animals , Endothelial Cells/metabolism , Ephrin-B2/genetics , Ephrin-B2/metabolism , Arteries/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Cell Separation , Receptor, EphB4/genetics , Receptor, EphB4/metabolism
3.
Elife ; 112022 02 04.
Article in English | MEDLINE | ID: mdl-35119364

ABSTRACT

Declining bone mass is associated with aging and osteoporosis, a disease characterized by progressive weakening of the skeleton and increased fracture incidence. Growth and lifelong homeostasis of bone rely on interactions between different cell types including vascular cells and mesenchymal stromal cells (MSCs). As these interactions involve Notch signaling, we have explored whether treatment with secreted Notch ligand proteins can enhance osteogenesis in adult mice. We show that a bone-targeting, high affinity version of the ligand Delta-like 4, termed Dll4(E12), induces bone formation in male mice without causing adverse effects in other organs, which are known to rely on intact Notch signaling. Due to lower bone surface and thereby reduced retention of Dll4(E12), the same approach failed to promote osteogenesis in female and ovariectomized mice but strongly enhanced trabecular bone formation in combination with parathyroid hormone. Single cell analysis of stromal cells indicates that Dll4(E12) primarily acts on MSCs and has comparably minor effects on osteoblasts, endothelial cells, or chondrocytes. We propose that activation of Notch signaling by bone-targeted fusion proteins might be therapeutically useful and can avoid detrimental effects in Notch-dependent processes in other organs.


Subject(s)
Osteogenesis , Osteoporosis/metabolism , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone and Bones/metabolism , Calcium-Binding Proteins/metabolism , Chondrocytes/metabolism , Endothelial Cells/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Signal Transduction
4.
Cell Rep ; 36(2): 109352, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260921

ABSTRACT

Bone stroma contributes to the regulation of osteogenesis and hematopoiesis but also to fracture healing and disease processes. Mesenchymal stromal cells from bone (BMSCs) represent a heterogenous mixture of different subpopulations with distinct molecular and functional properties. The lineage relationship between BMSC subsets and their regulation by intrinsic and extrinsic factors are not well understood. Here, we show with mouse genetics, ex vivo cell differentiation assays, and transcriptional profiling that BMSCs from metaphysis (mpMSCs) and diaphysis (dpMSCs) are fundamentally distinct. Fate-tracking experiments and single-cell RNA sequencing indicate that bone-forming osteoblast lineage cells and dpMSCs, including leptin receptor-positive (LepR+) reticular cells in bone marrow, emerge from mpMSCs in the postnatal metaphysis. Finally, we show that BMSC fate is controlled by platelet-derived growth factor receptor ß (PDGFRß) signaling and the transcription factor Jun-B. The sum of our findings improves our understanding of BMSC development, lineage relationships, and differentiation.


Subject(s)
Bone Development , Bone and Bones/cytology , Cell Lineage , Animals , Animals, Newborn , Bone and Bones/ultrastructure , Cell Differentiation , Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/ultrastructure , Mice, Inbred C57BL , Organ Specificity , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Single-Cell Analysis , Stromal Cells/cytology , Stromal Cells/ultrastructure , Transcription, Genetic
5.
Elife ; 92020 01 20.
Article in English | MEDLINE | ID: mdl-31958058

ABSTRACT

Blood vessels are integrated into different organ environments with distinct properties and physiology (Augustin and Koh, 2017). A striking example of organ-specific specialization is the bone vasculature where certain molecular signals yield the opposite effect as in other tissues (Glomski et al., 2011; Kusumbe et al., 2014; Ramasamy et al., 2014). Here, we show that the transcriptional coregulators Yap1 and Taz, components of the Hippo pathway, suppress vascular growth in the hypoxic microenvironment of bone, in contrast to their pro-angiogenic role in other organs. Likewise, the kinase Lats2, which limits Yap1/Taz activity, is essential for bone angiogenesis but dispensable in organs with lower levels of hypoxia. With mouse genetics, RNA sequencing, biochemistry, and cell culture experiments, we show that Yap1/Taz constrain hypoxia-inducible factor 1α (HIF1α) target gene expression in vivo and in vitro. We propose that crosstalk between Yap1/Taz and HIF1α controls angiogenesis depending on the level of tissue hypoxia, resulting in organ-specific biological responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Physiologic/genetics , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Hypoxia/genetics , Hippo Signaling Pathway , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , Mice, Inbred C57BL , Osteogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Trans-Activators/genetics , YAP-Signaling Proteins
6.
Nat Commun ; 10(1): 2817, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31249304

ABSTRACT

Sufficient vascular supply is indispensable for brain development and function, whereas dysfunctional blood vessels are associated with human diseases such as vascular malformations, stroke or neurodegeneration. Pericytes are capillary-associated mesenchymal cells that limit vascular permeability and protect the brain by preserving blood-brain barrier integrity. Loss of pericytes has been linked to neurodegenerative changes in genetically modified mice. Here, we report that postnatal inactivation of the Rbpj gene, encoding the transcription factor RBPJ, leads to alteration of cell identity markers in brain pericytes, increases local TGFß signalling, and triggers profound changes in endothelial behaviour. These changes, which are not mimicked by pericyte ablation, imperil vascular stability and induce the acquisition of pathological landmarks associated with cerebral cavernous malformations. In adult mice, loss of Rbpj results in bigger stroke lesions upon ischemic insult. We propose that brain pericytes can acquire deleterious properties that actively enhance vascular lesion formation and promote pathogenic processes.


Subject(s)
Brain/metabolism , Hemangioma, Cavernous, Central Nervous System/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Pericytes/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/cytology , Disease Progression , Female , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/pathology , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Male , Mice, Knockout
7.
Sci Rep ; 8(1): 9596, 2018 06 25.
Article in English | MEDLINE | ID: mdl-29941944

ABSTRACT

Endothelial cells and pericytes are integral cellular components of the vasculature with distinct interactive functionalities. To study dynamic interactions between these two cells we created two transgenic animal lines. A truncated eNOS (endothelial nitric oxide synthase) construct was used as a GFP tag for endothelial cell evaluation and an inducible Cre-lox recombination, under control of the Pdgfrb (platelet derived growth factor receptor beta) promoter, was created for pericyte assessment. Also, eNOStag-GFP animals were crossed with the already established Cspg4-DsRed mice expressing DsRed fluorescent protein in pericytes. For intravital imaging we used tumors implanted in the dorsal skinfold of these transgenic animals. This setup allowed us to study time and space dependent complexities, such as distribution, morphology, motility, and association between both vascular cell types in all angiogenetic stages, without the need for additional labeling. Moreover, as fluorescence was still clearly detectable after fixation, it is possible to perform comparative histology following intravital evaluation. These transgenic mouse lines form an excellent model to capture collective and individual cellular and subcellular endothelial cell - pericyte dynamics and will help answer key questions on the cellular and molecular relationship between these two cells.


Subject(s)
Endothelial Cells/pathology , Imaging, Three-Dimensional , Intravital Microscopy , Pericytes/pathology , Animals , Cell Communication , Cell Line, Tumor , Mice , Spatio-Temporal Analysis
8.
Nat Commun ; 9(1): 2448, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29934496

ABSTRACT

Blood vessels are essential for blood circulation but also control organ growth, homeostasis, and regeneration, which has been attributed to the release of paracrine signals by endothelial cells. Endothelial tubules are associated with specialised mesenchymal cells, termed pericytes, which help to maintain vessel wall integrity. Here we identify pericytes as regulators of epithelial and endothelial morphogenesis in postnatal lung. Mice lacking expression of the Hippo pathway components YAP and TAZ in pericytes show defective alveologenesis. Mutant pericytes are present in normal numbers but display strongly reduced expression of hepatocyte growth factor leading to impaired activation of the c-Met receptor, which is expressed by alveolar epithelial cells. YAP and TAZ are also required for expression of angiopoietin-1 by pulmonary pericytes, which also controls hepatocyte growth factor expression and thereby alveologenesis in an autocrine fashion. These findings establish that pericytes have important, organ-specific signalling properties and coordinate the behavior of epithelial and vascular cells during lung morphogenesis.


Subject(s)
Morphogenesis/physiology , Neovascularization, Physiologic/physiology , Pericytes/metabolism , Pulmonary Alveoli/growth & development , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Angiopoietin-1/metabolism , Animals , Cell Cycle Proteins , Cells, Cultured , Endothelial Cells/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoproteins/genetics , Phosphoproteins/metabolism , Primary Cell Culture , Pulmonary Alveoli/cytology , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
9.
Nat Commun ; 8(1): 1574, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29146905

ABSTRACT

Pericytes adhere to the abluminal surface of endothelial tubules and are required for the formation of stable vascular networks. Defective endothelial cell-pericyte interactions are frequently observed in diseases characterized by compromised vascular integrity such as diabetic retinopathy. Many functional properties of pericytes and their exact role in the regulation of angiogenic blood vessel growth remain elusive. Here we show that pericytes promote endothelial sprouting in the postnatal retinal vasculature. Using genetic and pharmacological approaches, we show that the expression of vascular endothelial growth factor receptor 1 (VEGFR1) by pericytes spatially restricts VEGF signalling. Angiogenic defects caused by pericyte depletion are phenocopied by intraocular injection of VEGF-A or pericyte-specific inactivation of the murine gene encoding VEGFR1. Our findings establish that pericytes promote endothelial sprouting, which results in the loss of side branches and the enlargement of vessels when pericyte function is impaired or lost.


Subject(s)
Endothelial Cells/metabolism , Eye/blood supply , Neovascularization, Physiologic/physiology , Pericytes/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Capillaries/cytology , Capillaries/growth & development , Cell Line , Diphtheria Toxin/toxicity , Endothelial Cells/cytology , Heparin-binding EGF-like Growth Factor/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Pericytes/cytology , Receptor, Platelet-Derived Growth Factor beta/genetics , Retina/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/genetics
10.
Nat Commun ; 8(1): 726, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28959057

ABSTRACT

Mediating the expansion of vascular beds in many physiological and pathological settings, angiogenesis requires dynamic changes in endothelial cell behavior. However, the molecular mechanisms governing endothelial cell activity during different phases of vascular growth, remodeling, maturation, and quiescence remain elusive. Here, we characterize dynamic gene expression changes during postnatal development and identify critical angiogenic factors in mouse retinal endothelial cells. Using actively translating transcriptome analysis and in silico computational analyses, we determine candidate regulators controlling endothelial cell behavior at different developmental stages. We further show that one of the identified candidates, the transcription factor MafB, controls endothelial sprouting in vitro and in vivo, and perform an integrative analysis of RNA-Seq and ChIP-Seq data to define putative direct MafB targets, which are activated or repressed by the transcriptional regulator. Together, our results identify novel cell-autonomous regulatory mechanisms controlling sprouting angiogenesis.Angiogenesis is a complex process that requires coordinated changes in endothelial cell behavior. Here the authors use Ribo-tag and RNA-Seq to determine temporal profiles of transcriptional activity during postnatal retinal angiogenesis, identifying transcriptional regulators of the process.


Subject(s)
Endothelial Cells , Gene Expression Regulation, Developmental , Gene Regulatory Networks , MafB Transcription Factor/genetics , Neovascularization, Physiologic/genetics , Retinal Vessels/growth & development , Animals , Gene Expression Regulation , In Vitro Techniques , Mice , Mice, Knockout , Retina/growth & development
11.
Angiogenesis ; 20(4): 581-598, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28795242

ABSTRACT

The roles of angiogenesis in development, health, and disease have been studied extensively; however, the studies related to lymphatic system are limited due to the difficulty in observing colorless lymphatic vessels. But recently, with the improved technique, the relative importance of the lymphatic system is just being revealed. We bred transgenic mice in which lymphatic endothelial cells express GFP (Prox1-GFP) with mice in which vascular endothelial cells express DsRed (Flt1-DsRed) to generate Prox1-GFP/Flt1-DsRed (PGFD) mice. The inherent fluorescence of blood and lymphatic vessels allows for direct visualization of blood and lymphatic vessels in various organs via confocal and two-photon microscopy and the formation, branching, and regression of both vessel types in the same live mouse cornea throughout an experimental time course. PGFD mice were bred with CDh5CreERT2 and VEGFR2lox knockout mice to examine specific knockouts. These studies showed a novel role for vascular endothelial cell VEGFR2 in regulating VEGFC-induced corneal lymphangiogenesis. Conditional deletion of vascular endothelial VEGFR2 abolished VEGFA- and VEGFC-induced corneal lymphangiogenesis. These results demonstrate the potential use of the PGFD mouse as a powerful animal model for studying angiogenesis and lymphangiogenesis.


Subject(s)
Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Imaging, Three-Dimensional , Luminescent Proteins/metabolism , Lymphangiogenesis , Neovascularization, Physiologic , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Endothelial Cells/metabolism , Female , Fluorescence , Male , Mice, Transgenic , Microscopy, Confocal , Models, Animal , Organ Specificity , Reproducibility of Results
12.
Nat Commun ; 7: 12422, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27516371

ABSTRACT

Mural cells of the vessel wall, namely pericytes and vascular smooth muscle cells, are essential for vascular integrity. The developmental sources of these cells and molecular mechanisms controlling their progenitors in the heart are only partially understood. Here we show that endocardial endothelial cells are progenitors of pericytes and vascular smooth muscle cells in the murine embryonic heart. Endocardial cells undergo endothelial-mesenchymal transition and convert into primitive mesenchymal progenitors expressing the platelet-derived growth factor receptors, PDGFRα and PDGFRß. These progenitors migrate into the myocardium, differentiate and assemble the wall of coronary vessels, which requires canonical Wnt signalling involving Frizzled4, ß-catenin and endothelial cell-derived Wnt ligands. Our findings identify a novel and unexpected population of progenitors for coronary mural cells with potential relevance for heart function and disease conditions.


Subject(s)
Cell Differentiation/physiology , Endothelial Cells/physiology , Epithelial-Mesenchymal Transition/physiology , Myocytes, Smooth Muscle/physiology , Pericytes/physiology , Animals , Cells, Cultured , Endocardium/cytology , Endocardium/physiology , Female , Heart/physiology , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Myocardium/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism
13.
Circ Res ; 115(6): 581-90, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25057127

ABSTRACT

RATIONALE: Endothelial cell-specific molecule 1 (Esm1) is a secreted protein thought to play a role in angiogenesis and inflammation. However, there is currently no direct in vivo evidence supporting a function of Esm1 in either of these processes. OBJECTIVE: To determine the role of Esm1 in vivo and the underlying molecular mechanisms. METHODS AND RESULTS: We generated and analyzed Esm1 knockout (Esm1(KO)) mice to study its role in angiogenesis and inflammation. Esm1 expression is induced by the vascular endothelial growth factor A (VEGF-A) in endothelial tip cells of the mouse retina. Esm1(KO) mice showed delayed vascular outgrowth and reduced filopodia extension, which are both VEGF-A-dependent processes. Impairment of Esm1 function led to a decrease in phosphorylated Erk1/2 (extracellular-signal regulated kinases 1/2) in sprouting vessels. We also found that Esm1(KO) mice displayed a 40% decrease in leukocyte transmigration. Moreover, VEGF-induced vascular permeability was decreased by 30% in Esm1(KO) mice and specifically on stimulation with VEGF-A165 but not VEGF-A121. Accordingly, cerebral edema attributable to ischemic stroke-induced vascular permeability was reduced by 50% in the absence of Esm1. Mechanistically, we show that Esm1 binds directly to fibronectin and thereby displaces fibronectin-bound VEGF-A165 leading to increased bioavailability of VEGF-A165 and subsequently enhanced levels of VEGF-A signaling. CONCLUSIONS: Esm1 is simultaneously a target and modulator of VEGF signaling in endothelial cells, playing a role in angiogenesis, inflammation, and vascular permeability, which might be of potential interest for therapeutic applications.


Subject(s)
Cell Membrane Permeability/physiology , Cell Membrane/physiology , Endothelial Cells/physiology , Proteoglycans/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Biological Availability , Fibronectins/metabolism , Inflammation/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Models, Animal , Neovascularization, Physiologic/physiology , Proteoglycans/deficiency , Proteoglycans/genetics , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism
14.
Development ; 140(14): 3051-61, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23785053

ABSTRACT

Blood vessels form a hierarchically organized network of arteries, capillaries and veins, which develops through a series of growth, pruning and maturation processes. In contrast to the rapidly increasing insight into the processes controlling vascular growth and, in particular, endothelial sprouting and proliferation, the conversion of immature vessels into a fully functional, quiescent vasculature remains little understood. Here we used inducible, cell type-specific genetic approaches to show that endothelial Notch signaling is crucial for the remodeling of veins and the perivenous capillary plexus, which occurs after the completion of the initial angiogenic growth phase in the retina of adolescent mice. Mutant vessels showed ectopic proliferation and sprouting, defective recruitment of supporting mural cells, and failed to downregulate the expression of VEGF receptors. Surprisingly, by contrast Notch was dispensable in the endothelium of remodeling postnatal arteries. Taken together, our results identify key processes contributing to vessel remodeling, maturation and the acquisition of a quiescent phenotype in the final stage of developmental angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic , Receptors, Notch/metabolism , Retinal Vessels/cytology , Signal Transduction , Animals , Cell Proliferation , Down-Regulation , Gene Targeting , Mice , Receptors, Vascular Endothelial Growth Factor/genetics , Retinal Artery/cytology , Retinal Vessels/growth & development , Retinal Vessels/metabolism
15.
FEBS Lett ; 586(22): 4046-51, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23068611

ABSTRACT

Metastasis is a major clinical issue and results in poor prognosis for most cancers. The Junctional Adhesion Molecule-C (JAM-C) expressed by B16 melanoma and endothelial cells has been involved in metastasis of tumor cells through homophilic JAM-C/JAM-C trans-interactions. Here, we show that JAM-B expressed by endothelial cells contributes to murine B16 melanoma cells metastasis through its interaction with JAM-C on tumor cells. We further show that this adhesion molecular pair mediates melanoma cell adhesion to primary Lung Microvascular Endothelial Cells and that it is functional in vivo as demonstrated by the reduced metastasis of B16 cells in Jam-b deficient mice.


Subject(s)
Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Immunoglobulins/metabolism , Melanoma, Experimental/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Female , Flow Cytometry , Immunoglobulins/genetics , Immunohistochemistry , Lung/blood supply , Lung/metabolism , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Neoplasm Metastasis , Protein Binding , RNA Interference
16.
Blood ; 118(17): 4609-19, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21868569

ABSTRACT

In adult mammals, hematopoietic stem cells (HSCs) reside in the bone marrow (BM) and are maintained in a quiescent and undifferentiated state through adhesive interactions with specialized microenvironmental niches. Although junctional adhesion molecule-C (JAM-C) is expressed by HSCs, its function in adult hematopoiesis remains elusive. Here, we show that HSCs adhere to JAM-B expressed by BM stromal cells in a JAM-C dependent manner. The interaction regulates the interplay between HSCs and BM stromal cells as illustrated by the decreased pool of quiescent HSCs observed in jam-b deficient mice. We further show that this is probably because of alterations of BM stromal compartments and changes in SDF-1α BM content in jam-b(-/-) mice, suggesting that JAM-B is an active player in the maintenance of the BM stromal microenvironment.


Subject(s)
Bone Marrow Cells/physiology , Cell Adhesion Molecules/physiology , Hematopoietic Stem Cells/physiology , Immunoglobulins/physiology , Animals , Bone Marrow/metabolism , Bone Marrow/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Immunoglobulins/genetics , Immunoglobulins/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
Nature ; 465(7297): 483-6, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20445537

ABSTRACT

In development, tissue regeneration or certain diseases, angiogenic growth leads to the expansion of blood vessels and the lymphatic vasculature. This involves endothelial cell proliferation as well as angiogenic sprouting, in which a subset of cells, termed tip cells, acquires motile, invasive behaviour and extends filopodial protrusions. Although it is already appreciated that angiogenesis is triggered by tissue-derived signals, such as vascular endothelial growth factor (VEGF) family growth factors, the resulting signalling processes in endothelial cells are only partly understood. Here we show with genetic experiments in mouse and zebrafish that ephrin-B2, a transmembrane ligand for Eph receptor tyrosine kinases, promotes sprouting behaviour and motility in the angiogenic endothelium. We link this pro-angiogenic function to a crucial role of ephrin-B2 in the VEGF signalling pathway, which we have studied in detail for VEGFR3, the receptor for VEGF-C. In the absence of ephrin-B2, the internalization of VEGFR3 in cultured cells and mutant mice is defective, which compromises downstream signal transduction by the small GTPase Rac1, Akt and the mitogen-activated protein kinase Erk. Our results show that full VEGFR3 signalling is coupled to receptor internalization. Ephrin-B2 is a key regulator of this process and thereby controls angiogenic and lymphangiogenic growth.


Subject(s)
Ephrin-B2/metabolism , Lymphangiogenesis , Neovascularization, Physiologic , Vascular Endothelial Growth Factor C/metabolism , Animals , Cells, Cultured , Embryo Loss , Embryo, Mammalian/blood supply , Embryo, Mammalian/metabolism , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ephrin-B2/deficiency , Ephrin-B2/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Lymphangiogenesis/genetics , Lymphatic Vessels , Mice , Mice, Transgenic , Neovascularization, Physiologic/genetics , Neuropeptides/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphB4/deficiency , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
18.
Cell ; 137(6): 1124-35, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524514

ABSTRACT

The Notch pathway is a highly conserved signaling system that controls a diversity of growth, differentiation, and patterning processes. In growing blood vessels, sprouting of endothelial tip cells is inhibited by Notch signaling, which is activated by binding of the Notch receptor to its ligand Delta-like 4 (Dll4). Here, we show that the Notch ligand Jagged1 is a potent proangiogenic regulator in mice that antagonizes Dll4-Notch signaling in cells expressing Fringe family glycosyltransferases. Upon glycosylation of Notch, Dll4-Notch signaling is enhanced, whereas Jagged1 has weak signaling capacity and competes with Dll4. Our findings establish that the equilibrium between two Notch ligands with distinct spatial expression patterns and opposing functional roles regulates angiogenesis, a mechanism that might also apply to other Notch-controlled biological processes.


Subject(s)
Blood Vessels/embryology , Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Adaptor Proteins, Signal Transducing , Animals , Blood Vessels/cytology , Calcium-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Female , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Mutation , Receptors, Notch/metabolism , Retina/embryology , Serrate-Jagged Proteins
19.
Cell ; 124(1): 161-73, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16413489

ABSTRACT

New blood vessels are initially formed through the assembly or sprouting of endothelial cells, but the recruitment of supporting pericytes and vascular smooth muscle cells (mural cells) ensures the formation of a mature and stable vascular network. Defective mural-cell coverage is associated with the poorly organized and leaky vasculature seen in tumors or other human diseases. Here we report that mural cells require ephrin-B2, a ligand for Eph receptor tyrosine kinases, for normal association with small-diameter blood vessels (microvessels). Tissue-specific mutant mice display perinatal lethality; vascular defects in skin, lung, gastrointestinal tract, and kidney glomeruli; and abnormal migration of smooth muscle cells to lymphatic capillaries. Cultured ephrin-B2-deficient smooth muscle cells are defective in spreading, focal-adhesion formation, and polarized migration and show increased motility. Our results indicate that the role of ephrin-B2 and EphB receptors in these processes involves Crk-p130(CAS) signaling and suggest that ephrin-B2 has some cell-cell-contact-independent functions.


Subject(s)
Blood Vessels/growth & development , Cell Movement/drug effects , Ephrin-B2/physiology , Animals , Blood Vessels/cytology , Blood Vessels/drug effects , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/physiology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Ephrin-B2/genetics , Ephrin-B2/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Neovascularization, Physiologic/physiology , Phenotype , Signal Transduction/physiology
20.
Nat Genet ; 36(2): 172-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14730302

ABSTRACT

Cell adhesion to extracellular matrix (ECM) proteins is crucial for the structural integrity of tissues and epithelial-mesenchymal interactions mediating organ morphogenesis. Here we describe how the loss of a cytoplasmic multi-PDZ scaffolding protein, glutamate receptor interacting protein 1 (GRIP1), leads to the formation of subepidermal hemorrhagic blisters, renal agenesis, syndactyly or polydactyly and permanent fusion of eyelids (cryptophthalmos). Similar malformations are characteristic of individuals with Fraser syndrome and animal models of this human genetic disorder, such as mice carrying the blebbed mutation (bl) in the gene encoding the Fras1 ECM protein. GRIP1 can physically interact with Fras1 and is required for the localization of Fras1 to the basal side of cells. In one animal model of Fraser syndrome, the eye-blebs (eb) mouse, Grip1 is disrupted by a deletion of two coding exons. Our data indicate that GRIP1 is required for normal cell-matrix interactions during early embryonic development and that inactivation of Grip1 causes Fraser syndrome-like defects in mice.


Subject(s)
Carrier Proteins/genetics , Extracellular Matrix Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, AMPA/genetics , Adaptor Proteins, Signal Transducing , Animals , Antigens/biosynthesis , Antigens/genetics , Carrier Proteins/physiology , Denys-Drash Syndrome/genetics , Denys-Drash Syndrome/metabolism , Disease Models, Animal , Embryo, Mammalian/abnormalities , Extracellular Matrix Proteins/physiology , Fluorescent Antibody Technique , Kidney/abnormalities , Mice , Nerve Tissue Proteins/physiology , Protein Structure, Tertiary , Proteoglycans/biosynthesis , Proteoglycans/genetics , Receptors, AMPA/physiology , Skin/embryology , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...