Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 11(1): 5400, 2020 10 26.
Article in English | MEDLINE | ID: mdl-33106502

ABSTRACT

Slug (SNAI2), a member of the well-conserved Snail family of transcription factors, has multiple developmental roles, including in epithelial-to-mesenchymal transition (EMT). Here, we show that Slug is critical for the pathological angiogenesis needed to sustain tumor growth, and transiently necessary for normal developmental angiogenesis. We find that Slug upregulation in angiogenic endothelial cells (EC) regulates an EMT-like suite of target genes, and suppresses Dll4-Notch signaling thereby promoting VEGFR2 expression. Both EC-specific Slug re-expression and reduced Notch signaling, either by γ-secretase inhibition or loss of Dll4, rescue retinal angiogenesis in SlugKO mice. Conversely, inhibition of VEGF signaling prevents excessive angiogenic sprouting of Slug overexpressing EC. Finally, endothelial Slug (but not Snail) is activated by the pro-angiogenic factor SDF1α via its canonical receptor CXCR4 and the MAP kinase ERK5. Altogether, our data support a critical role for Slug in determining the angiogenic response during development and disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Snail Family Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Calcium-Binding Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Snail Family Transcription Factors/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
2.
Arterioscler Thromb Vasc Biol ; 35(2): 303-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25425619

ABSTRACT

The contribution of epithelial-to-mesenchymal transitions (EMT) in both developmental and pathological conditions has been widely recognized and studied. In a parallel process, governed by a similar set of signaling and transcription factors, endothelial-to-mesenchymal transitions (EndoMT) contribute to heart valve formation and the generation of cancer-associated fibroblasts. During angiogenic sprouting, endothelial cells express many of the same genes and break down basement membrane; however, they retain intercellular junctions and migrate as a connected train of cells rather than as individual cells. This has been termed a partial endothelial-to-mesenchymal transition. A key regulatory check-point determines whether cells undergo a full or a partial epithelial-to-mesenchymal transitions/endothelial-to-mesenchymal transition; however, very little is known about how this switch is controlled. Here we discuss these developmental/pathological pathways, with a particular focus on their role in vascular biology.


Subject(s)
Endothelial Cells/pathology , Epithelial-Mesenchymal Transition , Neovascularization, Pathologic , Neovascularization, Physiologic , Angiogenic Proteins/metabolism , Animals , Endothelial Cells/metabolism , Humans , Signal Transduction , Transcription Factors/metabolism
3.
Integr Biol (Camb) ; 6(6): 603-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24763498

ABSTRACT

Metastasis is the cause of over 90% of all human cancer deaths. Early steps in the metastatic process include: the formation of new blood vessels, the initiation of epithelial-mesenchymal transition (EMT), and the mobilization of tumor cells into the circulation. There are ongoing efforts to replicate the physiological landscape of human tumor tissue using three-dimensional in vitro culture models; however, few systems are able to capture the full range of authentic, complex in vivo events such as neovascularization and intravasation. Here we introduce the Prevascularized Tumor (PVT) model to investigate early events of solid tumor progression. PVT spheroids are composed of endothelial and tumor cells, and are embedded in a fibrin matrix containing fibroblasts. The PVT model facilitates two mechanisms of vessel formation: robust sprouting angiogenesis into the matrix, and contiguous vascularization within the spheroid. Furthermore, the PVT model enables the intravasation of tumor cells that is enhanced under low oxygen conditions and is also dependent on the key EMT transcription factor Slug. The PVT model provides a significant advance in the mimicry of human tumors in vitro, and may improve investigation and targeting of events in the metastatic process.


Subject(s)
Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Fibrin/metabolism , Hypoxia/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Blotting, Western , Cell Line, Tumor , Humans , In Vitro Techniques , Microscopy, Fluorescence , RNA, Small Interfering/pharmacology , Snail Family Transcription Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
4.
Arterioscler Thromb Vasc Biol ; 34(5): 1011-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24603679

ABSTRACT

OBJECTIVE: It is well established that angiogenesis is a complex and coordinated multistep process. However, there remains a lack of information about the genes that regulate individual stages of vessel formation. Here, we aimed to define the role of human interferon-induced transmembrane protein 1 (IFITM1) during blood vessel formation. APPROACH AND RESULTS: We identified IFITM1 in a microarray screen for genes differentially regulated by endothelial cells (ECs) during an in vitro angiogenesis assay and found that IFITM1 expression was strongly induced as ECs sprouted and formed lumens. We showed by immunohistochemistry that human IFITM1 was expressed by stable blood vessels in multiple organs. siRNA-mediated knockdown of IFITM1 expression spared EC sprouting but completely disrupted lumen formation, in both in vitro and in an in vivo xeno-transplant model. ECs lacking IFITM1 underwent early stages of lumenogenesis (ie, intracellular vacuole formation) but failed to mature or expand lumens. Coimmunoprecipitation studies confirmed occludin as an IFITM1 binding partner in ECs, and immunocytochemistry showed a lack of occludin at endothelial tight junctions in the absence of IFITM1. Finally, time-lapse video microscopy revealed that IFITM1 is required for the formation of stable cell-cell contacts during endothelial lumen formation. CONCLUSIONS: IFITM1 is essential for the formation of functional blood vessels and stabilizes EC-EC interactions during endothelial lumen formation by regulating tight junction assembly.


Subject(s)
Antigens, Differentiation/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Animals , Antigens, Differentiation/genetics , Cells, Cultured , Gene Expression Profiling/methods , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Immunoprecipitation , Mice , Mice, Inbred ICR , Mice, SCID , Microscopy, Video , Occludin/metabolism , Oligonucleotide Array Sequence Analysis , Protein Binding , RNA Interference , Signal Transduction , Tight Junctions/metabolism , Time Factors , Time-Lapse Imaging , Transfection
5.
J Cell Sci ; 127(Pt 9): 2017-28, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24554431

ABSTRACT

The Snail family of zinc-finger transcription factors are evolutionarily conserved proteins that control processes requiring cell movement. Specifically, they regulate epithelial-to-mesenchymal transitions (EMT) where an epithelial cell severs intercellular junctions, degrades basement membrane and becomes a migratory, mesenchymal-like cell. Interestingly, Slug expression has been observed in angiogenic endothelial cells (EC) in vivo, suggesting that angiogenic sprouting may share common attributes with EMT. Here, we demonstrate that sprouting EC in vitro express both Slug and Snail, and that siRNA-mediated knockdown of either inhibits sprouting and migration in multiple in vitro angiogenesis assays. We find that expression of MT1-MMP, but not of VE-Cadherin, is regulated by Slug and that loss of sprouting as a consequence of reduced Slug expression can be reversed by lentiviral-mediated re-expression of MT1-MMP. Activity of MMP2 and MMP9 are also affected by Slug expression, likely through MT1-MMP. Importantly, we find enhanced expression of Slug in EC in human colorectal cancer samples compared with normal colon tissue, suggesting a role for Slug in pathological angiogenesis. In summary, these data implicate Slug as an important regulator of sprouting angiogenesis, particularly in pathological settings.


Subject(s)
Transcription Factors/metabolism , Cells, Cultured , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Methylcellulose/metabolism , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors
6.
Arterioscler Thromb Vasc Biol ; 33(3): 513-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23288153

ABSTRACT

OBJECTIVE: Angiogenesis requires tightly coordinated crosstalk between endothelial cells (ECs) and stromal cells, such as fibroblasts and smooth muscle cells. The specific molecular mechanisms moderating this process are still poorly understood. METHODS AND RESULTS: Stromal cell-derived factors are essential for EC sprouting and lumen formation. We therefore compared the abilities of 2 primary fibroblast isolates and a primary smooth muscle cell isolate to promote in vitro angiogenesis, and analyzed their secretomes using a combination of nano liquid chromatography-mass spectrometry/mass spectrometry, quantitative PCR, and ELISA. Each isolate exhibited a different level of angiogenic ability. Using quantitative MS, we then compared the secretomes of a fibroblast isolate exhibiting low angiogenic activity, a fibroblast isolate exhibiting high angiogenic activity, and human umbilical vein ECs. High angiogenic fibroblast supernatants exhibited an overabundance of proteins associated with extracellular matrix constituents compared with low angiogenic fibroblasts or ECs. Finally, small interfering RNA technology and purified protein were used to confirm a role for stromal cell-derived hepatocyte growth factor and fibronectin in inducing EC sprouting. CONCLUSIONS: Differences in stromal cell ability to induce angiogenesis are a result of differences in the secreted proteomes of both extracellular matrix proteins and proangiogenic growth factors.


Subject(s)
Fibronectins/metabolism , Hepatocyte Growth Factor/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Paracrine Communication , Stromal Cells/metabolism , Cells, Cultured , Chromatography, Liquid , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Fibronectins/genetics , Hepatocyte Growth Factor/genetics , Humans , Myocytes, Smooth Muscle/metabolism , Nanotechnology , Proteomics/methods , RNA Interference , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...