Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Front Immunol ; 13: 752215, 2022.
Article in English | MEDLINE | ID: mdl-35222366

ABSTRACT

The four isoforms of the RNA-binding protein hnRNPD/AUF1 have been proposed to limit the use of inflammatory mRNAs in innate immune cells. Mice engineered to lack AUF1s in all tissues are sensitive to acute inflammatory assaults; however, they also manifest complex degenerations obscuring assessment of AUF1s' roles in innate immune cells. Here, we restricted a debilitating AUF1 mutation to the mouse myeloid lineage and performed disease-oriented phenotypic analyses to assess the requirement of AUF1s in variable contexts of innate immune reactivity. Contrary to the whole-body mutants, the myeloid mutants of AUF1s did not show differences in their susceptibility to cytokine storms occurring during endotoxemia; neither in type-I cell-mediated reactions driving intestinal inflammation by chemical irritants. Instead, they were resistant to allergic airway inflammation and displayed reductions in inflammatory infiltrates and an altered T-helper balance. The ex-vivo analysis of macrophages revealed that the loss of AUF1s had a minimal effect on their proinflammatory gene expression. Moreover, AUF1s were dispensable for the classical polarization of cultured macrophages by LPS & IFNγ correlating with the unchanged response of mutant mice to systemic and intestinal inflammation. Notably, AUF1s were also dispensable for the alternative polarization of macrophages by IL4, TGFß and IL10, known to be engaged in allergic reactions. In contrast, they were required to switch proinflammatory macrophages towards a pro-angiogenic phenotype induced by adenosine receptor signals. Congruent to this, the myeloid mutants of AUF1 displayed lower levels of vascular remodeling factors in exudates from allergen exposed lungs; were unable to support the growth and inflammatory infiltration of transplanted melanoma tumors; and failed to vascularize inert grafts unless supplemented with angiogenic factors. Mechanistically, adenosine receptor signals enhanced the association of AUF1s with the Vegfa, Il12b, and Tnf mRNAs to differentially regulate and facilitate the pro-angiogenic switch. Our data collectively demonstrates that AUF1s do not act as general anti-inflammatory factors in innate immune cells but have more specialized roles in regulons allowing specific innate immune cell transitions to support tissue infiltration and remodeling processes.


Subject(s)
Hypersensitivity , Neoplasms , Adenosine/metabolism , Animals , Hypersensitivity/metabolism , Inflammation , Lung/metabolism , Macrophages , Mice , Myeloid Cells/metabolism , Neoplasms/metabolism , RNA, Messenger/genetics
3.
Int J Dev Biol ; 66(1-2-3): 103-113, 2022.
Article in English | MEDLINE | ID: mdl-34881799

ABSTRACT

The development and homeostasis of vertebrate organisms depend on the "tree of life", in other words, the intricate network of vascular tubes composed of endothelial cells attached to the basement membrane and surrounded by perivascular cells. Although many studies have revealed the fundamental role of cytokines, growth factors and Notch signalling in vascular morphogenesis, we still lack sufficient understanding of the molecular mechanisms controlling the various steps of the angiogenic processes. Emerging data highlight that cell adhesions are key players in vascular morphogenesis. In this review, we focus on endothelial cells and we present the current state of knowledge regarding the role of cell-matrix adhesions in developmental and tumour angiogenesis, attained mainly from genetic studies and animal models.


Subject(s)
Endothelial Cells , Neoplasms , Animals , Cell-Matrix Junctions , Morphogenesis/genetics , Neoplasms/etiology , Neoplasms/metabolism , Neovascularization, Physiologic
4.
Cancers (Basel) ; 13(3)2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33573141

ABSTRACT

Beyond the conventional perception of solid tumours as mere masses of cancer cells, advanced cancer research focuses on the complex contributions of tumour-associated host cells that are known as "tumour microenvironment" (TME). It has been long appreciated that the tumour stroma, composed mainly of blood vessels, cancer-associated fibroblasts and immune cells, together with the extracellular matrix (ECM), define the tumour architecture and influence cancer cell properties. Besides soluble cues, that mediate the crosstalk between tumour and stroma cells, cell adhesion to ECM arises as a crucial determinant in cancer progression. In this review, we discuss how adhesome, the intracellular protein network formed at cell adhesions, regulate the TME and control malignancy. The role of adhesome extends beyond the physical attachment of cells to ECM and the regulation of cytoskeletal remodelling and acts as a signalling and mechanosensing hub, orchestrating cellular responses that shape the tumour milieu.

5.
Dev Cell ; 50(6): 767-779.e7, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31447263

ABSTRACT

Eukaryotic organisms accomplish the removal of introns to produce mature mRNAs through splicing. Nuclear and organelle splicing mechanisms are distinctively executed by spliceosome and group II intron complex, respectively. Here, we show that LEFKOTHEA, a nuclear encoded RNA-binding protein, participates in chloroplast group II intron and nuclear pre-mRNA splicing. Transiently optimized LEFKOTHEA nuclear activity is fundamental for plant growth, whereas the loss of function abruptly arrests embryogenesis. Nucleocytoplasmic partitioning and chloroplast allocation are efficiently balanced via functional motifs in LEFKOTHEA polypeptide. In the context of nuclear-chloroplast coevolution, our results provide a strong paradigm of the convergence of RNA maturation mechanisms in the nucleus and chloroplasts to coordinately regulate gene expression and effectively control plant growth.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Cell Nucleus/genetics , Chloroplasts/genetics , RNA Splicing/genetics , RNA-Binding Proteins/metabolism , Arabidopsis/embryology , Arabidopsis/ultrastructure , Arabidopsis Proteins/genetics , Cell Nucleus/ultrastructure , Chloroplasts/ultrastructure , Gene Expression Regulation, Plant , Genes, Plant , Introns/genetics , Meristem/metabolism , Models, Biological , Mutation/genetics , Phenotype , Protein Binding/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Seeds/metabolism , Seeds/ultrastructure , Spliceosomes/metabolism
6.
Cancer Res ; 79(17): 4371-4386, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31189647

ABSTRACT

Expression of focal adhesion kinase (FAK) in endothelial cells (EC) is essential for angiogenesis, but how FAK phosphorylation at tyrosine-(Y)397 and Y861 regulate tumor angiogenesis in vivo is unknown. Here, we show that tumor growth and angiogenesis are constitutively reduced in inducible, ECCre+;FAKY397F/Y397F -mutant mice. Conversely, ECCre+;FAKY861F/Y861F mice exhibit normal tumor growth with an initial reduction in angiogenesis that recovered in end-stage tumors. Mechanistically, FAK-Y397F ECs exhibit increased Tie2 expression, reduced Vegfr2 expression, decreased ß1 integrin activation, and disrupted downstream FAK/Src/PI3K(p55)/Akt signaling. In contrast, FAK-Y861F ECs showed decreased Vegfr2 and Tie2 expression with an enhancement in ß1 integrin activation. This corresponds with a decrease in Vegfa-stimulated response, but an increase in Vegfa+Ang2- or conditioned medium from tumor cell-stimulated cellular/angiogenic responses, mimicking responses in end-stage tumors with elevated Ang2 levels. Mechanistically, FAK-Y861F, but not FAK-Y397F ECs showed enhanced p190RhoGEF/P130Cas-dependent signaling that is required for the elevated responses to Vegfa+Ang2. This study establishes the differential requirements of EC-FAK-Y397 and EC-FAK-Y861 phosphorylation in the regulation of EC signaling and tumor angiogenesis in vivo. SIGNIFICANCE: Distinct motifs of the focal adhesion kinase differentially regulate tumor blood vessel formation and remodeling.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Neovascularization, Pathologic/metabolism , Angiotensin II/pharmacology , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Focal Adhesion Kinase 1/genetics , Integrin beta1/metabolism , Mice, Knockout , Mice, Mutant Strains , Neovascularization, Pathologic/drug therapy , Phosphorylation , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/metabolism
8.
Front Physiol ; 6: 170, 2015.
Article in English | MEDLINE | ID: mdl-26106332

ABSTRACT

Tissue regeneration and wound healing are severely impaired in diabetes and are associated with poor circulation and dysfunctional blood vessels. Angiotensin II inhibitors are anti-hypertensive drugs used in clinical practice to regulate blood pressure and could affect tissue remodeling. We hypothesize that blocking angiotensin II, using Losartan, could facilitate tissue regeneration in diabetic mice. To this end, we established an experimental model of wound healing in streptozotocin-induced diabetic mice. Our data demonstrated that Losartan accelerates wound repair and normalizes wound stromal responses, having a beneficial role in wounds of diabetic individuals. Our findings highlight a potential therapeutic use of Losartan in improving wound repair in diabetic conditions.

9.
Front Genet ; 5: 236, 2014.
Article in English | MEDLINE | ID: mdl-25101116

ABSTRACT

Alternative reading frame (ARF) is a tumor suppressor protein that senses oncogenic and other stressogenic signals. It can trigger p53-dependent and -independent responses with cell cycle arrest and apoptosis induction being the most prominent ones. Other ARF activities, particularly p53-independent ones, that could help in understanding cancer development and provide potential therapeutic exploitation are underrated. Although ARF is generally not expressed in normal tissues, it is essential for ocular and male germ cells development. The underlying mechanism(s) in these processes, while not clearly defined, point toward a functional link between ARF, DNA damage and angiogenesis. Based on a recent study from our group demonstrating a functional interplay between ataxia-telangiectasia mutated (ATM) and ARF during carcinogenesis, we discuss the role of ARF at the crossroads of cancer and developmental processes.

10.
Biochim Biophys Acta ; 1840(8): 2403-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24576673

ABSTRACT

BACKGROUND: The extracellular matrix (ECM) is constituted by diverse composite structures, which determine the specific to each organ, histological architecture and provides cells with biological information, mechanical support and a scaffold for adhesion and migration. The pleiotropic effects of the ECM stem from the dynamic changes in its molecular composition and the ability to remodel in order to effectively regulate biological outcomes. Besides collagens, fibronectin and laminin are two major fiber-forming constituents of various ECM structures. SCOPE OF REVIEW: This review will focus on the properties and the biological functions of non-collagenous extracellular matrix especially on laminin and fibronectin that are currently emerging as important regulators of blood vessel formation and function in health and disease. MAJOR CONCLUSIONS: The ECM is a fundamental component of the microenvironment of blood vessels, with activities extending beyond providing a vascular scaffold; extremely versatile it directly or indirectly modulates all essential cellular functions crucial for angiogenesis, including cell adhesion, migration, proliferation, differentiation and lumen formation. Specifically, fibronectin and laminins play decisive roles in blood vessel morphogenesis both during embryonic development and in pathological conditions, such as cancer. GENERAL SIGNIFICANCE: Emerging evidence demonstrates the importance of ECM function during embryonic development, organ formation and tissue homeostasis. A wealth of data also illustrates the crucial role of the ECM in several human pathophysiological processes, including fibrosis, skeletal diseases, vascular pathologies and cancer. Notably, several ECM components have been identified as potential therapeutic targets for various diseases, including cancer. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.


Subject(s)
Blood Vessels/growth & development , Blood Vessels/metabolism , Extracellular Matrix Proteins/metabolism , Morphogenesis , Neoplasms/metabolism , Neovascularization, Physiologic , Animals , Collagen/metabolism , Extracellular Matrix Proteins/chemistry , Humans , Neoplasms/blood supply , Neoplasms/pathology
11.
Nat Commun ; 4: 2020, 2013.
Article in English | MEDLINE | ID: mdl-23799510

ABSTRACT

Genetic ablation of endothelial focal adhesion kinase (FAK) can inhibit pathological angiogenesis, suggesting that loss of endothelial FAK is sufficient to reduce neovascularization. Here we show that reduced stromal FAK expression in FAK-heterozygous mice unexpectedly enhances both B16F0 and CMT19T tumour growth and angiogenesis. We further demonstrate that cell proliferation and microvessel sprouting, but not migration, are increased in serum-stimulated FAK-heterozygous endothelial cells. FAK-heterozygous endothelial cells display an imbalance in FAK phosphorylation at pY397 and pY861 without changes in Pyk2 or Erk1/2 activity. By contrast, serum-stimulated phosphorylation of Akt is enhanced in FAK-heterozygous endothelial cells and these cells are more sensitive to Akt inhibition. Additionally, low doses of a pharmacological FAK inhibitor, although too low to affect FAK autophosphorylation in vitro, can enhance angiogenesis ex vivo and tumour growth in vivo. Our results highlight a potential novel role for FAK as a nonlinear, dose-dependent regulator of angiogenesis where heterozygous levels of FAK enhance angiogenesis.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Neoplasms/blood supply , Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Animals , Cell Proliferation , Cell Separation , Cell Survival , Endothelial Cells/pathology , Heterozygote , Immunohistochemistry , In Vitro Techniques , Mice , Mutant Proteins/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Paxillin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Subcutaneous Tissue/pathology , Talin/metabolism , Tumor Burden , Vinculin/metabolism
12.
J Proteomics ; 75(17): 5356-69, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22800643

ABSTRACT

Mutation of RAS genes is one of the most common oncogenic alterations in cancer and acquisition of activating RAS mutations has been demonstrated to cause progression of colorectal adenoma to cancer. The aim of this study was to identify changes in the proteome of the intermediate-stage colorectal cancer cell line Caco2, induced by ectopic expression of two distinct RAS proteins, KRAS(V12) and HRAS(V12), in their mutated, constitutively active form. Using 2D-gel electrophoresis, followed by LC-MS/MS we identified almost 200 differentially expressed proteins in pair-wise comparisons of Caco2 vs Caco2-KRAS(V12) and Caco2 vs Caco2-HRAS(V12). Although many of the affected proteins were unique for each pair, there were also substantial similarities. Interestingly, transformation by the mutant KRAS(V12) gene resulted in elevated expression levels and activity of endogenous H-ras protein. Silencing the latter with a specific RNAi reversed several proteomic changes observed in KRAS(V12)-transformed cells, suggesting that oncogenic K-ras partly exerts its effects through endogenous H-ras activation. Alterations in the expression of cytoskeletal and cell adhesion proteins, caused by HRAS siRNA treatment, correlated with a reduction in the invasive properties of Caco2-KRAS(V12) cells. Our data suggest a novel interplay between K-ras and H-ras, with possible implications for colorectal carcinogenesis.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Genes, ras/physiology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Caco-2 Cells , Cell Line, Tumor , Cluster Analysis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Electrophoresis, Gel, Two-Dimensional , HCT116 Cells , Humans , Metabolic Networks and Pathways/genetics , Models, Biological , Oncogenes/physiology , Proteomics/methods , Signal Transduction/genetics , Signal Transduction/physiology , Tandem Mass Spectrometry
13.
Dev Biol ; 349(2): 494-502, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21081121

ABSTRACT

Using Tln1(fl/fl);CreER mice, we show that tamoxifen-induced inactivation of the talin1 gene throughout the embryo produces an angiogenesis phenotype that is restricted to newly forming blood vessels. The phenotype has a rapid onset in early embryos, resulting in vessel defects by 48 h and death of the embryo within 72 h. Very similar vascular defects were obtained using a Tie2-Cre endothelial cell-specific Tln1 knockout, a phenotype that was rescued by expression of a Tln1 mini-gene in endothelial cells. We show that endothelial cells, unlike most other cell types, do not express talin2, which can compensate for loss of talin1, and demonstrate for the first time that endothelial cells in vivo lacking talin1 are unable to undergo the cell spreading and flattening required to form vessels.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , Talin/metabolism , Animals , Blotting, Western , DNA Primers/genetics , Embryo, Mammalian , Endothelial Cells/physiology , Gene Silencing/drug effects , Histological Techniques , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Electron , Talin/genetics , Tamoxifen
14.
EMBO Mol Med ; 2(12): 516-28, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21154724

ABSTRACT

Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that plays a fundamental role in integrin and growth factor mediated signalling and is an important player in cell migration and proliferation, processes vital for angiogenesis. However, the role of FAK in adult pathological angiogenesis is unknown. We have generated endothelial-specific tamoxifen-inducible FAK knockout mice by crossing FAK-floxed (FAKfl/fl) mice with the platelet derived growth factor b (Pdgfb)-iCreER mice. Tamoxifen-treatment of Pdgfb-iCreER;FAKfl/fl mice results in FAK deletion in adult endothelial cells (ECs) without any adverse effects. Importantly however, endothelial FAK-deletion in adult mice inhibited tumour growth and reduced tumour angiogenesis. Furthermore, in in vivo angiogenic assays FAK deletion impairs vascular endothelial growth factor (VEGF)-induced neovascularization. In addition, in vitro deletion of FAK in ECs resulted in reduced VEGF-stimulated Akt phosphorylation and correlating reduced cellular proliferation as well as increased cell death. Our data suggest that FAK is required for adult pathological angiogenesis and validates FAK as a possible target for anti-angiogenic therapies.


Subject(s)
Endothelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Animals , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Deletion , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/physiopathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Phosphorylation , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Am J Pathol ; 177(3): 1534-48, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20639457

ABSTRACT

Integrin alpha3beta1 is a major receptor for laminin. The expression levels of laminins-8 and -10 in the basement membrane surrounding blood vessels are known to change during tumor angiogenesis. Although some studies have suggested that certain ligands of alpha3beta1 can affect angiogenesis either positively or negatively, either a direct in vivo role for alpha3beta1 in this process or its mechanism of action in endothelial cells during angiogenesis is still unknown. Because the global genetic ablation of alpha3-integrin results in an early lethal phenotype, we have generated conditional-knockout mice where alpha3 is deleted specifically in endothelial cells (ec-alpha3-/-). Here we show that ec-alpha3-/- mice are viable, fertile, and display enhanced tumor growth, elevated tumor angiogenesis, augmented hypoxia-induced retinal angiogenesis, and increased vascular endothelial growth factor (VEGF)-mediated neovascularization ex vivo and in vivo. Furthermore, our data provide a novel method by which an integrin may regulate angiogenesis. We show that alpha3beta1 is a positive regulator of endothelial-VEGF and that, surprisingly, the VEGF produced by endothelial cells can actually repress VEGF-receptor 2 (Flk-1) expression. These data, therefore, identify directly that endothelial alpha3beta1 negatively regulates pathological angiogenesis and implicate an unexpected role for low levels of endothelial-VEGF as an activator of neovascularization.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Hypoxia/metabolism , Integrin alpha3beta1/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Female , Flow Cytometry , Hypoxia/genetics , Hypoxia/pathology , Immunohistochemistry , Integrin alpha3beta1/genetics , Male , Mice , Mice, Knockout , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Mol Cancer Res ; 8(4): 554-68, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20353996

ABSTRACT

The TAF4b subunit of the transcription factor IID, which has a central role in transcription by polymerase II, is involved in promoter recognition by selective recruitment of activators. The activating protein-1 (AP-1) family members participate in oncogenic transformation via gene regulation. Utilizing immunoprecipitation of endogenous protein complexes, we documented specific interactions between Jun family members and TATA box binding protein-associated factors (TAF) in colon HT29 adenocarcinoma cells. Particularly, TAF4b and c-Jun were found to colocalize and interact in the nucleus of advanced carcinoma cells and in cells with epithelial-to-mesenchymal transition (EMT) characteristics. TAF4b was found to specifically regulate the AP-1 target gene involved in EMT integrin alpha6, thus altering related cellular properties such as migration potential. Using a chromatin immunoprecipitation approach in colon adenocarcinoma cell lines, we further identified a synergistic role for TAF4b and c-Jun and other AP-1 family members on the promoter of integrin alpha6, underlining the existence of a specific mechanism related to gene expression control. We show evidence for the first time of an interdependence of TAF4b and AP-1 family members in cell type-specific promoter recognition and initiation of transcription in the context of cancer progression and EMT.


Subject(s)
Integrin alpha6/genetics , Neoplasm Invasiveness/genetics , Neoplasms/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor AP-1/genetics , Transcription Factor TFIID/genetics , Transcriptional Activation/genetics , Caco-2 Cells , Cell Dedifferentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Integrin alpha6/metabolism , Neoplasms/metabolism , Promoter Regions, Genetic/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor AP-1/metabolism , Transcription Factor TFIID/metabolism
17.
J Pathol ; 220(3): 370-81, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19967723

ABSTRACT

Laminins are expressed highly in blood vessel basement membranes and have been implicated in angiogenesis. alpha6beta1- and alpha6beta4-integrins are major receptors for laminins in endothelial cells, but the precise role of endothelial alpha6-integrin in tumour angiogenesis is not clear. We show that blood vessels in human invasive ductal carcinoma of the breast have decreased expression of the alpha6-integrin-subunit when compared with normal breast tissue. These data suggest that a decrease in alpha6-integrin-subunit expression in endothelial cells is associated with tumour angiogenesis. To test whether the loss of the endothelial alpha6-integrin subunit affects tumour growth and angiogenesis, we generated alpha6fl/fl-Tie1Cre+ mice and showed that endothelial deletion of alpha6-integrin is sufficient to enhance tumour size and tumour angiogenesis in both murine B16F0 melanoma and Lewis cell lung carcinoma. Mechanistically, endothelial alpha6-integrin deficiency elevated significantly VEGF-mediated angiogenesis both in vivo and ex vivo. In particular, alpha6-integrin-deficient endothelial cells displayed increased levels of VEGF-receptor 2 (VEGFR2) and VEGF-mediated downstream ERK1/2 activation. By developing the first endothelial-specific alpha6-knockout mice, we show that the expression of the alpha6-integrin subunit in endothelial cells acts as a negative regulator of angiogenesis both in vivo and ex vivo.


Subject(s)
Breast Neoplasms/blood supply , Integrin alpha6/genetics , Neoplasm Proteins/genetics , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/genetics , Animals , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/blood supply , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genotype , Humans , Integrin alpha6/metabolism , Integrin alpha6/physiology , Melanoma/blood supply , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/metabolism , Polymerase Chain Reaction/methods , Vascular Endothelial Growth Factor A/toxicity , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Development ; 136(23): 4043-53, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19906871

ABSTRACT

Sprouting angiogenesis and lymphatic-blood vessel segregation both involve the migration of endothelial cells, but the precise migratory molecules that govern the decision of blood vascular endothelial cells to segregate into lymphatic vasculature are unknown. Here, we deleted endothelial Rac1 in mice (Tie1-Cre(+);Rac1(fl/fl)) and revealed, unexpectedly, that whereas blood vessel morphology appeared normal, lymphatic-blood vessel separation was impaired, with corresponding edema, haemorrhage and embryonic lethality. Importantly, normal levels of Rac1 were essential for directed endothelial cell migratory responses to lymphatic-inductive signals. Our studies identify Rac1 as a crucial part of the migratory machinery required for endothelial cells to separate and form lymphatic vasculature.


Subject(s)
Blood Vessels/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Lymphatic Vessels/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Separation/methods , Cells, Cultured , Embryo, Mammalian , Endothelium, Vascular/cytology , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique, Direct , Fluorescent Dyes/metabolism , Galactosides/metabolism , Gene Deletion , Immunohistochemistry , Indoles/metabolism , Mice , Mice, Transgenic , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , RNA, Small Interfering/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Transfection , beta-Galactosidase/metabolism , rac1 GTP-Binding Protein/analysis , rac1 GTP-Binding Protein/genetics
19.
J Biol Chem ; 284(49): 33966-81, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19837659

ABSTRACT

Both vascular endothelial growth factor receptors (VEGFR) and integrins are major regulators of VEGF-induced angiogenesis. Previous work has shown that beta3 integrin can regulate negatively VEGFR2 expression. Here we show that beta3 integrin can regulate negatively VEGF-mediated angiogenesis by limiting the interaction of the co-receptor NRP1 (neuropilin-1) with VEGFR2. In the presence of alphav beta3 integrin, NRP1 contributed minimally to VEGF-induced angiogenic processes in vivo, ex vivo, and in vitro. Conversely, when beta3 integrin expression is absent or low or its function is blocked with RGD-mimetic inhibitors, VEGF-mediated responses became NRP1-dependent. Indeed, combined inhibition of beta3 integrin and NRP1 decreased VEGF-mediated angiogenic responses further than individual inhibition of these receptors. We also show that alphav beta3 integrin can associate with NRP1 in a VEGF-dependent fashion. Our data suggest that beta3 integrin may, in part, negatively regulate VEGF signaling by sequestering NRP1 and preventing it from interacting with VEGFR2.


Subject(s)
Integrin alphaVbeta3/metabolism , Neovascularization, Pathologic , Neuropilin-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Aorta/cytology , Base Sequence , Endothelial Cells/cytology , Humans , Mice , Microcirculation , Molecular Sequence Data , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing
20.
Nat Med ; 15(4): 392-400, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19305413

ABSTRACT

Inhibitors of alpha(v)beta(3) and alpha(v)beta(5) integrin have entered clinical trials as antiangiogenic agents for cancer treatment but generally have been unsuccessful. Here we present in vivo evidence that low (nanomolar) concentrations of RGD-mimetic alpha(v)beta(3) and alpha(v)beta(5) inhibitors can paradoxically stimulate tumor growth and tumor angiogenesis. We show that low concentrations of these inhibitors promote VEGF-mediated angiogenesis by altering alpha(v)beta(3) integrin and vascular endothelial growth factor receptor-2 trafficking, thereby promoting endothelial cell migration to VEGF. The proangiogenic effects of low concentrations of RGD-mimetic integrin inhibitors could compromise their efficacy as anticancer agents and have major implications for the use of RGD-mimetic compounds in humans.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Integrin alphaVbeta3/therapeutic use , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Receptors, Vitronectin/therapeutic use , Animals , Disease Models, Animal , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...