Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 15(2): e1007551, 2019 02.
Article in English | MEDLINE | ID: mdl-30730983

ABSTRACT

By engulfing potentially harmful microbes, professional phagocytes are continually at risk from intracellular pathogens. To avoid becoming infected, the host must kill pathogens in the phagosome before they can escape or establish a survival niche. Here, we analyse the role of the phosphoinositide (PI) 5-kinase PIKfyve in phagosome maturation and killing, using the amoeba and model phagocyte Dictyostelium discoideum. PIKfyve plays important but poorly understood roles in vesicular trafficking by catalysing formation of the lipids phosphatidylinositol (3,5)-bisphosphate (PI(3,5)2) and phosphatidylinositol-5-phosphate (PI(5)P). Here we show that its activity is essential during early phagosome maturation in Dictyostelium. Disruption of PIKfyve inhibited delivery of both the vacuolar V-ATPase and proteases, dramatically reducing the ability of cells to acidify newly formed phagosomes and digest their contents. Consequently, PIKfyve- cells were unable to generate an effective antimicrobial environment and efficiently kill captured bacteria. Moreover, we demonstrate that cells lacking PIKfyve are more susceptible to infection by the intracellular pathogen Legionella pneumophila. We conclude that PIKfyve-catalysed phosphoinositide production plays a crucial and general role in ensuring early phagosomal maturation, protecting host cells from diverse pathogenic microbes.


Subject(s)
Dictyostelium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Adenosine Triphosphatases , Animals , Cell Line , Dictyostelium/pathogenicity , Humans , Hydrolases/metabolism , Legionella pneumophila/pathogenicity , Legionellosis/metabolism , Macrophages , Phagocytosis , Phagosomes , Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositols , Protein Transport , Protozoan Infections/metabolism
2.
Haematologica ; 104(9): 1892-1905, 2019 09.
Article in English | MEDLINE | ID: mdl-30573509

ABSTRACT

Ca2+ entry via Orai1 store-operated Ca2+ channels in the plasma membrane is critical to cell function, and Orai1 loss causes severe immunodeficiency and developmental defects. The tetraspanins are a superfamily of transmembrane proteins that interact with specific 'partner proteins' and regulate their trafficking and clustering. The aim of this study was to functionally characterize tetraspanin Tspan18. We show that Tspan18 is expressed by endothelial cells at several-fold higher levels than most other cell types analyzed. Tspan18-knockdown primary human umbilical vein endothelial cells have 55-70% decreased Ca2+ mobilization upon stimulation with the inflammatory mediators thrombin or histamine, similar to Orai1-knockdown. Tspan18 interacts with Orai1, and Orai1 cell surface localization is reduced by 70% in Tspan18-knockdown endothelial cells. Tspan18 overexpression in lymphocyte model cell lines induces 20-fold activation of Ca2+ -responsive nuclear factor of activated T cell (NFAT) signaling, in an Orai1-dependent manner. Tspan18-knockout mice are viable. They lose on average 6-fold more blood in a tail-bleed assay. This is due to Tspan18 deficiency in non-hematopoietic cells, as assessed using chimeric mice. Tspan18-knockout mice have 60% reduced thrombus size in a deep vein thrombosis model, and 50% reduced platelet deposition in the microcirculation following myocardial ischemia-reperfusion injury. Histamine- or thrombin-induced von Willebrand factor release from endothelial cells is reduced by 90% following Tspan18-knockdown, and histamine-induced increase of plasma von Willebrand factor is reduced by 45% in Tspan18-knockout mice. These findings identify Tspan18 as a novel regulator of endothelial cell Orai1/Ca2+ signaling and von Willebrand factor release in response to inflammatory stimuli.


Subject(s)
Calcium/metabolism , Myocardial Reperfusion Injury/genetics , ORAI1 Protein/genetics , Tetraspanins/genetics , Venous Thrombosis/genetics , von Willebrand Factor/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Chickens , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Histamine/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ion Transport/drug effects , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , ORAI1 Protein/metabolism , Signal Transduction , Tetraspanins/metabolism , Thrombin/pharmacology , Venous Thrombosis/metabolism , Venous Thrombosis/pathology , von Willebrand Factor/metabolism
3.
Angiogenesis ; 20(3): 341-358, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28271280

ABSTRACT

Regulator of calcineurin 1 (RCAN1) is an endogenous inhibitor of the calcineurin pathway in cells. It is expressed as two isoforms in vertebrates: RCAN1.1 is constitutively expressed in most tissues, whereas transcription of RCAN1.4 is induced by several stimuli that activate the calcineurin-NFAT pathway. RCAN1.4 is highly upregulated in response to VEGF in human endothelial cells in contrast to RCAN1.1 and is essential for efficient endothelial cell migration and tubular morphogenesis. Here, we show that RCAN1.4 has a role in the regulation of agonist-stimulated VEGFR-2 internalisation and establishment of endothelial cell polarity. siRNA-mediated gene silencing revealed that RCAN1 plays a vital role in regulating VEGF-mediated cytoskeletal reorganisation and directed cell migration and sprouting angiogenesis. Adenoviral-mediated overexpression of RCAN1.4 resulted in increased endothelial cell migration. Antisense-mediated morpholino silencing of the zebrafish RCAN1.4 orthologue revealed a disrupted vascular development further confirming a role for the RCAN1.4 isoform in regulating vascular endothelial cell physiology. Our data suggest that RCAN1.4 plays a novel role in regulating endothelial cell migration by establishing endothelial cell polarity in response to VEGF.


Subject(s)
Cell Movement , Cell Polarity , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microvessels/cytology , Muscle Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cytoskeleton/metabolism , DNA-Binding Proteins , Embryo, Nonmammalian/metabolism , Humans , Ligands , Models, Biological , Neovascularization, Physiologic , Protein Binding , Protein Isoforms/metabolism , Zebrafish/embryology
4.
Cancer Res ; 77(4): 1008-1020, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28011623

ABSTRACT

Antiangiogenic therapies have failed to confer survival benefits in patients with metastatic breast cancer (mBC). However, to date, there has not been an inquiry into the roles for acquired versus innate drug resistance in this setting. In this study, we report roles for these distinct phenotypes in determining therapeutic response in a murine model of mBC resistance to the antiangiogenic tyrosine kinase inhibitor sunitinib. Using tumor measurement and vascular patterning approaches, we differentiated tumors displaying innate versus acquired resistance. Bioluminescent imaging of tumor metastases to the liver, lungs, and spleen revealed that sunitinib administration enhances metastasis, but only in tumors displaying innate resistance to therapy. Transcriptomic analysis of tumors displaying acquired versus innate resistance allowed the identification of specific biomarkers, many of which have a role in angiogenesis. In particular, aquaporin-1 upregulation occurred in acquired resistance, mTOR in innate resistance, and pleiotrophin in both settings, suggesting their utility as candidate diagnostics to predict drug response or to design tactics to circumvent resistance. Our results unravel specific features of antiangiogenic resistance, with potential therapeutic implications. Cancer Res; 77(4); 1008-20. ©2016 AACR.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Indoles/therapeutic use , Pyrroles/therapeutic use , Animals , Aquaporin 1/physiology , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Carrier Proteins/physiology , Cell Movement , Cytokines/physiology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Luminescent Measurements , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Real-Time Polymerase Chain Reaction , Sunitinib
5.
Methods Mol Biol ; 1430: 149-57, 2016.
Article in English | MEDLINE | ID: mdl-27172951

ABSTRACT

Angiogenesis involves the generation of new blood vessels from the existing vasculature and is dependent on many growth factors and signaling events. In vivo angiogenesis is dynamic and complex, meaning assays are commonly utilized to explore specific targets for research into this area. Tube-forming assays offer an excellent overview of the molecular processes in angiogenesis. The Matrigel tube forming assay is a simple-to-implement but powerful tool for identifying biomolecules involved in angiogenesis. A detailed experimental protocol on the implementation of the assay is described in conjunction with an in-depth review of methods that can be applied to the analysis of the tube formation. In addition, an ImageJ plug-in is presented which allows automatic quantification of tube images reducing analysis times while removing user bias and subjectivity.


Subject(s)
Endothelial Cells/ultrastructure , Neovascularization, Physiologic , Cell Culture Techniques , Cell Movement , Cell Proliferation , Endothelial Cells/cytology , High-Throughput Screening Assays , Human Umbilical Vein Endothelial Cells , Humans
6.
Oncotarget ; 7(15): 20440-54, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26943033

ABSTRACT

Current vascular-targeted therapies in colorectal cancer (CRC) have shown limited benefit. The lack of novel, specific treatment in CRC has been hampered by a dearth of specific endothelial markers. Microarray comparison of endothelial gene expression in patient-matched CRC and normal colon identified a panel of putative colorectal tumour endothelial markers. Of these the glutamate dependent NMDA receptor GRIN2D emerged as the most interesting target. GRIN2D expression was shown to be specific to colorectal cancer vessels by RTqPCR and IHC analysis. Its expression was additionally shown be predictive of improved survival in CRC. Targeted knockdown studies in vitro demonstrated a role for GRIN2D in endothelial function and angiogenesis. This effect was also shown in vivo as vaccination against the extracellular region of GRIN2D resulted in reduced vascularisation in the subcutaneous sponge angiogenesis assay. The utility of immunologically targeting GRIN2D in CRC was demonstrated by the vaccination approach inhibiting murine CRC tumour growth and vascularisation. GRIN2D represents a promising target for the future treatment of CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Endothelium, Vascular/pathology , Neovascularization, Pathologic/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Apoptosis , Cell Movement , Cell Proliferation , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/pathology , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Inbred BALB C , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Cancer Res ; 76(8): 2314-26, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26921326

ABSTRACT

The structure and molecular signature of tumor-associated vasculature are distinct from those of the host tissue, offering an opportunity to selectively target the tumor blood vessels. To identify tumor-specific endothelial markers, we performed a microarray on tumor-associated and nonmalignant endothelium collected from patients with renal cell carcinoma (RCC), colorectal carcinoma, or colorectal liver metastasis. We identified a panel of genes consistently upregulated by tumor blood vessels, of which melanoma cell adhesion molecule (MCAM) and its extracellular matrix interaction partner laminin alpha 4 (LAMA4) emerged as the most consistently expressed genes. This result was subsequently confirmed by immunohistochemical analysis of MCAM and LAMA4 expression in RCC and colorectal carcinoma blood vessels. Strong MCAM and LAMA4 expression was also shown to predict poor survival in RCC, but not in colorectal carcinoma. Notably, MCAM and LAMA4 were enhanced in locally advanced tumors as well as both the primary tumor and secondary metastases. Expression analysis in 18 different cancers and matched healthy tissues revealed vascular MCAM as highly specific in RCC, where it was induced strongly by VEGF, which is highly abundant in this disease. Lastly, MCAM monoclonal antibodies specifically localized to vessels in a murine model of RCC, offering an opportunity for endothelial-specific targeting of anticancer agents. Overall, our findings highlight MCAM and LAMA4 as prime candidates for RCC prognosis and therapeutic targeting. Cancer Res; 76(8); 2314-26. ©2016 AACR.


Subject(s)
Carcinoma, Renal Cell/blood supply , Kidney Neoplasms/blood supply , Laminin/metabolism , Animals , CD146 Antigen/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/therapy , Human Umbilical Vein Endothelial Cells , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/therapy , Mice , Neoplasm Metastasis , Treatment Outcome , Vascular Endothelial Growth Factor A/pharmacology
8.
Angiogenesis ; 18(1): 83-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348086

ABSTRACT

Tumor endothelial specific expression of Robo4 in adults identifies this plasma membrane protein as an anti-cancer target for immunotherapeutic approaches, such as vaccination. In this report, we describe how vaccination against Robo4 inhibits angiogenesis and tumor growth. To break tolerance to the auto-antigen Robo4, mice were immunised with the extracellular domain of mouse Robo4, fused to the Fc domain of human immunoglobulin within an adjuvant. Vaccinated mice show a strong antibody response to Robo4, with no objectively detectable adverse effects on health. Robo4 vaccinated mice showed impaired fibrovascular invasion and angiogenesis in a rodent sponge implantation assay, as well as a reduced growth of implanted syngeneic Lewis lung carcinoma. The anti-tumor effect of Robo4 vaccination was present in CD8 deficient mice but absent in B cell or IgG1 knockout mice, suggesting antibody dependent cell mediated cytotoxicity as the anti-vascular/anti-tumor mechanism. Finally, we show that an adjuvant free soluble Robo4-carrier conjugate can retard tumor growth in carrier primed mice. These results point to appropriate Robo4 conjugates as potential anti-angiogenic vaccines for cancer patients.


Subject(s)
Immunoglobulin Fc Fragments/immunology , Immunotherapy/methods , Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Nerve Tissue Proteins/immunology , Receptors, Immunologic/immunology , Vaccines, Synthetic/pharmacology , Adult , Amino Acid Sequence , Animals , Chromatography, Affinity , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Genetic Vectors/genetics , HEK293 Cells , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Papain , Polymerase Chain Reaction , Receptors, Cell Surface , Tumor Cells, Cultured , Vaccines, Synthetic/immunology
9.
J Cell Sci ; 127(Pt 14): 3039-51, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24928894

ABSTRACT

RhoJ is a Rho GTPase expressed in endothelial cells and tumour cells, which regulates cell motility, invasion, endothelial tube formation and focal adhesion numbers. This study aimed to further delineate the molecular function of RhoJ. Using timelapse microscopy RhoJ was found to regulate focal adhesion disassembly; small interfering RNA (siRNA)-mediated knockdown of RhoJ increased focal adhesion disassembly time, whereas expression of an active mutant (daRhoJ) decreased it. Furthermore, daRhoJ co-precipitated with the GIT-PIX complex, a regulator of focal adhesion disassembly. An interaction between daRhoJ and GIT1 was confirmed using yeast two-hybrid experiments, and this depended on the Spa homology domain of GIT1. GIT1, GIT2, ß-PIX (also known as ARHGEF7) and RhoJ all colocalised in focal adhesions and depended on each other for their recruitment to focal adhesions. Functionally, the GIT-PIX complex regulated endothelial tube formation, with knockdown of both GIT1 and GIT2, or ß-PIX phenocopying RhoJ knockdown. RhoJ-knockout mice showed reduced tumour growth and diminished tumour vessel density, identifying a role for RhoJ in mediating tumour angiogenesis. These studies give new insight into the molecular function of RhoJ in regulating cell motility and tumour vessel formation.


Subject(s)
Cell Cycle Proteins/metabolism , Focal Adhesions/metabolism , GTP Phosphohydrolases/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , rho GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Movement/physiology , GTPase-Activating Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/metabolism , Signal Transduction
10.
Biochem Soc Trans ; 39(6): 1571-5, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22103489

ABSTRACT

We have in recent years described several endothelial-specific genes that mediate cell migration. These include Robo4 (roundabout 4), CLEC14A (C-type lectin 14A) and ECSCR (endothelial cell-specific chemotaxis regulator) [formerly known as ECSM2 (endothelial cell-specific molecule 2)]. Loss of laminar shear stress induces Robo4 and CLEC14A expression and an endothelial 'tip cell' phenotype. Low shear stress is found not only at sites of vascular occlusion such as thrombosis and embolism, but also in the poorly structured vessels that populate solid tumours. The latter probably accounts for strong expression of Robo4 and CLEC14A on tumour vessels. The function of Robo4 has, in the past, aroused controversy. However, the recent identification of Unc5B as a Robo4 ligand has increased our understanding and we hypothesize that Robo4 function is context-dependent. ECSCR is another endothelial-specific protein that promotes filopodia formation and migration, but, in this case, expression is independent of shear stress. We discuss recent papers describing ECSCR, including intracellular signalling pathways, and briefly contrast these with signalling by Robo4.


Subject(s)
Cell Movement , Endothelial Cells/cytology , Endothelial Cells/metabolism , Stress, Mechanical , Animals , Humans , Neovascularization, Physiologic/genetics , Signal Transduction/genetics
11.
Biochem Soc Trans ; 39(6): 1606-11, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22103495

ABSTRACT

RhoJ is an endothelially expressed member of the Cdc42 (cell division cycle 42) subfamily of small Rho GTPases. It is expressed in both the developing mammalian vasculature and the vascular beds of a number of adult tissues, with its expression regulated by the endothelial transcription factor ERG (ETS-related gene). RhoJ has been shown to regulate endothelial motility, tubulogenesis and lumen formation in vitro, and modulates the vascularization of Matrigel plugs in vivo. Both vascular endothelial growth factor and semaphorin 3E have been found to affect its activation. RhoJ has been shown to be a focal-adhesion-localized Rho GTPase which can modulate focal adhesion number, actomyosin contractility and activity of Cdc42 and Rac1. The present review discusses the biology of RhoJ with a focus on recent reports of its role in endothelial cells and angiogenesis.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/enzymology , Neovascularization, Physiologic , rho GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Animals , Focal Adhesions/metabolism , Humans
12.
Biochem Soc Trans ; 39(6): 1667-73, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22103505

ABSTRACT

Tetraspanins function as organizers of the cell surface by recruiting specific partner proteins into tetraspanin-enriched microdomains, which regulate processes such as cell adhesion, signalling and intracellular trafficking. Endothelial cells appear to express at least 23 of the 33 human tetraspanins, and a number of recent studies have demonstrated their importance in endothelial cell biology. Tetraspanin CD151 is essential for pathological angiogenesis, which may in part be due to regulation of its main partner proteins, the laminin-binding integrins α3ß1, α6ß1 and α6ß4. CD9 and CD151 are essential for leucocyte recruitment during an inflammatory response, through the formation of pre-assembled nano-platforms containing the adhesion molecules ICAM-1 (intercellular adhesion molecule 1) and VCAM-1 (vascular cell adhesion molecule 1), which ultimately coalesce to form docking structures around captured leucocytes. Tetraspanin CD63 also facilitates leucocyte capture by promoting clustering of the adhesion molecule P-selectin. Finally, Tspan12 is required for blood vessel development in the eye, through regulation of Norrin-induced Frizzled-4 signalling, such that Tspan12 mutations can lead to human disease. Future studies on these and other endothelial tetraspanins are likely to provide further novel insights into angiogenesis and inflammation.


Subject(s)
Endothelial Cells/metabolism , Membrane Microdomains/metabolism , Tetraspanins/metabolism , Animals , Blood Vessels/growth & development , Blood Vessels/metabolism , Cell Adhesion , Endothelial Cells/cytology , Humans , Neovascularization, Physiologic , Tetraspanins/chemistry
13.
Arterioscler Thromb Vasc Biol ; 31(3): 657-64, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21148427

ABSTRACT

OBJECTIVE: RhoJ/TCL was identified by our group as an endothelial-expressed Rho GTPase. The aim of this study was to determine its tissue distribution, subcellular localization, and function in endothelial migration and tube formation. METHODS AND RESULTS: Using in situ hybridization, RhoJ was localized to endothelial cells in a set of normal and cancerous tissues and in the vasculature of mouse embryos; endogenous RhoJ was localized to focal adhesions by immunofluorescence. The proangiogenic factor vascular endothelial growth factor activated RhoJ in endothelial cells. Using either small interfering (si)RNA-mediated knockdown of RhoJ expression or overexpression of constitutively active RhoJ (daRhoJ), RhoJ was found to positively regulate endothelial motility and tubule formation. Downregulating RhoJ expression increased focal adhesions and stress fibers in migrating cells, whereas daRhoJ overexpression resulted in the converse. RhoJ downregulation resulted in increased contraction of a collagen gel and increased phospho-myosin light chain, indicative of increased actomyosin contractility. Pharmacological inhibition of Rho-kinase (which phosphorylates myosin light chain) or nonmuscle myosin II reversed the defective tube formation and migration of RhoJ knockdown cells. CONCLUSIONS: RhoJ is endothelial-expressed in vivo, activated by vascular endothelial growth factor, localizes to focal adhesions, regulates endothelial cell migration and tube formation, and modulates actomyosin contractility and focal adhesion numbers.


Subject(s)
Actomyosin/metabolism , Cell Movement , Endothelial Cells/enzymology , Focal Adhesions/metabolism , GTP Phosphohydrolases/metabolism , Neovascularization, Physiologic , rho GTP-Binding Proteins/metabolism , Animals , Cell Movement/drug effects , Cell Shape , Cells, Cultured , Endothelial Cells/drug effects , Fluorescent Antibody Technique , GTP Phosphohydrolases/genetics , Humans , In Situ Hybridization , Mice , Myosin Light Chains/metabolism , Neovascularization, Physiologic/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Stress Fibers/metabolism , Transfection , Vascular Endothelial Growth Factor A/metabolism , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
14.
Biochem Soc Trans ; 37(Pt 6): 1214-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19909249

ABSTRACT

We have applied search algorithms to expression databases to identify genes whose expression is restricted to the endothelial cell. Such genes frequently play a critical role in endothelial biology and angiogenesis. Two such genes are the roundabout receptor Robo4 and the ECSCR (endothelial-cell-specific chemotaxis regulator). Endothelial cells express both Robo1 and Robo4, which we have knocked down using siRNA (small interfering RNA) and then studied the effect in a variety of in vitro assays. Both Robo4 and Robo1 knockdown inhibited in vitro tube formation on Matrigel. Transfection of Robo4 into endothelial cells increased the number of filopodial extensions from the cell, but failed to do so in Robo1-knockdown cells. Separate immunoprecipitation studies showed that Robo1 and Robo4 heterodimerize. We conclude from this and other work that a heteroduplex of Robo1 and Robo4 signals through WASP (Wiskott-Aldrich syndrome protein) and other actin nucleation-promoting factors to increase the number of filopodia and cell migration. Knockdown of the transmembrane ECSCR protein in endothelial cells also reduced chemotaxis and impaired tube formation on Matrigel. Yeast two-hybrid analysis and immunoprecipitation studies showed that, in contrast with the roundabouts, ECSCR binds to the actin-modulatory filamin A. We conclude that all three of these genes are critical for effective endothelial cell migration and, in turn, angiogenesis.


Subject(s)
Endothelial Cells/physiology , Membrane Proteins , Receptors, Cell Surface , Animals , Apoptosis Regulatory Proteins , Cell Movement/physiology , Databases, Genetic , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neovascularization, Physiologic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction/physiology , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism , Roundabout Proteins
15.
Nat Rev Clin Oncol ; 6(7): 395-404, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19424102

ABSTRACT

The growth and metastasis of solid tumors critically depends on their ability to develop their own blood supply, a process known as tumor angiogenesis. Over the past decade much work has been performed to understand this process, and modifying this process provides a key point of therapeutic intervention in the fight against cancer. This Review explores the development of anti-VEGF-based antiangiogenic therapies, of which there are currently three licensed for clinical use worldwide. Although originally anticipated to inhibit the growth of tumor vessels, the induction of vascular normalization caused by these approved agents has provided a novel means of effective delivery of known chemotherapeutic agents. The development of small molecules that target VEGF receptors has resulted in the generation of inhibitors with not only vascular activity but antitumor activity in certain cancers. This Review will address the current status of vascular-disrupting strategies, such as therapies designed to induce tumor collapse by selectively destroying existing tumor vessels. These therapies can be broadly divided into small-molecular-weight vascular-disrupting agents and ligand-directed approaches. We discuss the current status of development, drug mechanisms of actions, combination with conventional chemotherapy and radiotherapy, and potential future targets for therapeutic intervention.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Phytogenic/therapeutic use , Bevacizumab , Clinical Trials as Topic , Humans , Regional Blood Flow/drug effects , Stilbenes/therapeutic use , Vascular Endothelial Growth Factor A/therapeutic use
16.
FASEB J ; 23(2): 513-22, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18948384

ABSTRACT

This study aimed to further elucidate the function of Roundabout proteins in endothelium. We show that both Robo1 and Robo4 are present in human umbilical vein endothelial cells (HUVECs) and have knocked expression down using small interfering RNA (siRNA) technology. Roundabout knockout endothelial cells were then studied in a variety of in vitro assays. We also performed a yeast 2-hybrid analysis using the intracellular domain of Robo4 as bait to identify interacting proteins and downstream signaling. Both Robo1 and Robo4 siRNA knockdown and transfection of Robo4-green fluorescent protein inhibited endothelial cell movement and disrupted tube formation on Matrigel. Consistent with a role in regulating cell movement, yeast 2-hybrid and glutathione-S-transferase pulldown analyses show Robo4 binding to a Wiskott-Aldrich syndrome protein (WASP), neural Wiskott-Aldrich syndrome protein, and WASP-interacting protein actin-nucleating complex. We have further shown that Robo1 forms a heterodimeric complex with Robo4, and that transfection of Robo4GFP into HUVECs induces filopodia formation. We finally show using Robo1 knockdown cells that Robo1 is essential for Robo4-mediated filopodia induction. Our results favor a model whereby Slit2 binding to a Robo1/Robo4 heterodimer activates actin nucleation-promoting factors to promote endothelial cell migration.


Subject(s)
Actins/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Nerve Tissue Proteins/metabolism , Pseudopodia/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Animals , Cell Movement , Cells, Cultured , Chlorocebus aethiops , Humans , Nerve Tissue Proteins/genetics , Protein Multimerization , RNA, Small Interfering/genetics , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics , Signal Transduction , Wiskott-Aldrich Syndrome Protein/genetics , Roundabout Proteins
17.
Biochem J ; 417(1): 391-400, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18795891

ABSTRACT

Platelets are essential for wound healing and inflammatory processes, but can also play a deleterious role by causing heart attack and stroke. Normal platelet activation is dependent on tetraspanins, a superfamily of glycoproteins that function as 'organisers' of cell membranes by recruiting other receptors and signalling proteins into tetraspanin-enriched microdomains. However, our understanding of how tetraspanin microdomains regulate platelets is hindered by the fact that only four of the 33 mammalian tetraspanins have been identified in platelets. This is because of a lack of antibodies to most tetraspanins and difficulties in measuring mRNA, due to low levels in this anucleate cell. To identify potentially platelet-expressed tetraspanins, mRNA was measured in their nucleated progenitor cell, the megakaryocyte, using serial analysis of gene expression and DNA microarrays. Amongst 19 tetraspanins identified in megakaryocytes, Tspan9, a previously uncharacterized tetraspanin, was relatively specific to these cells. Through generating the first Tspan9 antibodies, Tspan9 expression was found to be tightly regulated in platelets. The relative levels of CD9, CD151, Tspan9 and CD63 were 100, 14, 6 and 2 respectively. Since CD9 was expressed at 49000 cell surface copies per platelet, this suggested a copy number of 2800 Tspan9 molecules. Finally, Tspan9 was shown to be a component of tetraspanin microdomains that included the collagen receptor GPVI (glycoprotein VI) and integrin alpha6beta1, but not the von Willebrand receptor GPIbalpha or the integrins alphaIIbbeta3 or alpha2beta1. These findings suggest a role for Tspan9 in regulating platelet function in concert with other platelet tetraspanins and their associated proteins.


Subject(s)
Blood Platelets/metabolism , Megakaryocytes/metabolism , Membrane Proteins/metabolism , Platelet Membrane Glycoproteins/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cells, Cultured , Humans , Megakaryocytes/cytology , Membrane Microdomains/metabolism , Membrane Proteins/genetics , Mice , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Platelet Membrane Glycoproteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tetraspanins
18.
Arterioscler Thromb Vasc Biol ; 28(9): 1640-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18556573

ABSTRACT

OBJECTIVE: We aimed to characterize the expression and function of a novel transcript that bioinformatics analysis predicted to be endothelial specific, called endothelial-specific molecule-2 (ECSM2). METHODS AND RESULTS: A full-length cDNA was isolated and predicted ECSM2 to be a putative 205-amino acid transmembrane protein that bears no homology to any known protein. Quantitative polymerase chain reaction analysis in vitro and in situ hybridization analysis in vivo confirmed ECSM2 expression to be exclusively endothelial, and localization to the plasma membrane was shown. Knockdown of ECSM2 expression in human umbilical vein endothelial cells using siRNA resulted in both reduced chemotaxis and impaired tube formation on matrigel, a solubilized basement membrane, both processes involved in angiogenesis. A yeast 2 hybrid analysis using the ECSM2 intracellular domain identified filamin A as an interacting protein. This interaction was confirmed by precipitation of filamin-A from endothelial cell lysates by a GST-tagged intracellular domain of ECSM2. CONCLUSIONS: This study is the first to characterize a novel cell surface protein ECSM2 that regulates endothelial chemotaxis and tube formation, and interacts with filamin A. These studies implicate a role for ECSM2 in angiogenesis via modulation of the actin cytoskeleton.


Subject(s)
Cell Membrane/metabolism , Chemotaxis , Contractile Proteins/metabolism , Endothelial Cells/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Neovascularization, Physiologic , Amino Acid Sequence , Apoptosis Regulatory Proteins , Cells, Cultured , Cloning, Molecular , Filamins , Humans , In Situ Hybridization , Membrane Proteins/genetics , Molecular Sequence Data , Polymerase Chain Reaction , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , Two-Hybrid System Techniques
19.
BMC Genomics ; 9: 153, 2008 Apr 07.
Article in English | MEDLINE | ID: mdl-18394197

ABSTRACT

BACKGROUND: In this study, differential gene expression analysis using complementary DNA (cDNA) libraries has been improved. Firstly by the introduction of an accurate method of assigning Expressed Sequence Tags (ESTs) to genes and secondly, by using a novel likelihood ratio statistical scoring of differential gene expression between two pools of cDNA libraries. These methods were applied to the latest available cell line and bulk tissue cDNA libraries in a two-step screen to predict novel tumour endothelial markers. Initially, endothelial cell lines were in silico subtracted from non-endothelial cell lines to identify endothelial genes. Subsequently, a second bulk tumour versus normal tissue subtraction was employed to predict tumour endothelial markers. RESULTS: From an endothelial cDNA library analysis, 431 genes were significantly up regulated in endothelial cells with a False Discovery Rate adjusted q-value of 0.01 or less and 104 of these were expressed only in endothelial cells. Combining the cDNA library data with the latest Serial Analysis of Gene Expression (SAGE) library data derived a complete list of 459 genes preferentially expressed in endothelium. 27 genes were predicted tumour endothelial markers in multiple tissues based on the second bulk tissue screen. CONCLUSION: This approach represents a significant advance on earlier work in its ability to accurately assign an EST to a gene, statistically measure differential expression between two pools of cDNA libraries and predict putative tumour endothelial markers before entering the laboratory. These methods are of value and available http://www.compbio.ox.ac.uk/data/diffex.html to researchers that are interested in the analysis of transcriptomic data.


Subject(s)
Algorithms , Computational Biology/methods , Endothelial Cells/metabolism , Expressed Sequence Tags , Gene Expression Profiling/methods , Genetic Markers/genetics , Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Library , Humans , Likelihood Functions , Neoplasms/metabolism
20.
Mol Cell Proteomics ; 6(3): 548-64, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17186946

ABSTRACT

The platelet surface is poorly characterized due to the low abundance of many membrane proteins and the lack of specialist tools for their investigation. In this study we identified novel human platelet and mouse megakaryocyte membrane proteins using specialist proteomics and genomics approaches. Three separate methods were used to enrich platelet surface proteins prior to identification by liquid chromatography and tandem mass spectrometry: lectin affinity chromatography, biotin/NeutrAvidin affinity chromatography, and free flow electrophoresis. Many known, abundant platelet surface transmembrane proteins and several novel proteins were identified using each receptor enrichment strategy. In total, two or more unique peptides were identified for 46, 68, and 22 surface membrane, intracellular membrane, and membrane proteins of unknown subcellular localization, respectively. The majority of these were single transmembrane proteins. To complement the proteomics studies, we analyzed the transcriptome of a highly purified preparation of mature primary mouse megakaryocytes using serial analysis of gene expression in view of the increasing importance of mutant mouse models in establishing protein function in platelets. This approach identified all of the major classes of platelet transmembrane receptors, including multitransmembrane proteins. Strikingly 17 of the 25 most megakaryocyte-specific genes (relative to 30 other serial analysis of gene expression libraries) were transmembrane proteins, illustrating the unique nature of the megakaryocyte/platelet surface. The list of novel plasma membrane proteins identified using proteomics includes the immunoglobulin superfamily member G6b, which undergoes extensive alternate splicing. Specific antibodies were used to demonstrate expression of the G6b-B isoform, which contains an immunoreceptor tyrosine-based inhibition motif. G6b-B undergoes tyrosine phosphorylation and association with the SH2 domain-containing phosphatase, SHP-1, in stimulated platelets suggesting that it may play a novel role in limiting platelet activation.


Subject(s)
Blood Platelets/chemistry , Megakaryocytes/chemistry , Membrane Proteins/analysis , Animals , Cell Membrane/chemistry , Cells, Cultured , Chromatography, Affinity , Gene Expression , Genomics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Platelet Activation , Proteomics , RNA/metabolism , Receptors, Immunologic/analysis , Receptors, Immunologic/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL