Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Gabriela D'Amico is active.

Publication


Featured researches published by Gabriela D'Amico.


Nature Protocols | 2012

Use of the mouse aortic ring assay to study angiogenesis

Marianne Baker; Stephen Robinson; Tanguy Lechertier; Paul R. Barber; Bernardo Tavora; Gabriela D'Amico; Dylan T. Jones; Boris Vojnovic; Kairbaan Hodivala-Dilke

Here we provide a protocol for quantitative three-dimensional ex vivo mouse aortic ring angiogenesis assays, in which developing microvessels undergo many key features of angiogenesis over a timescale similar to that observed in vivo. The aortic ring assay allows analysis of cellular proliferation, migration, tube formation, microvessel branching, perivascular recruitment and remodeling—all without the need for cellular dissociation—thus providing a more complete picture of angiogenic processes compared with traditional cell-based assays. Our protocol can be applied to aortic rings from embryonic stage E18 through to adulthood and can incorporate genetic manipulation, treatment with growth factors, drugs or siRNA. This robust assay allows assessment of the salient steps in angiogenesis and quantification of the developing microvessels, and it can be used to identify new modulators of angiogenesis. The assay takes 6–14 d to complete, depending on the age of the mice, treatments applied and whether immunostaining is performed.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2008

Integrins The Keys to Unlocking Angiogenesis

Rita Silva; Gabriela D'Amico; Kairbaan Hodivala-Dilke; Louise E. Reynolds

Angiogenesis, the formation of new blood vessels from preexisting vasculature, contributes to the pathogenesis of many disorders, including ischemic diseases and cancer. Integrins are cell adhesion molecules that are expressed on the surface of endothelial cells and pericytes, making them potential targets for antiangiogenic therapy. Here we review the contribution of endothelial and mural cell integrins to angiogenesis and highlight their potential as antiangiogenesis targets.


Nature | 2010

Tumour angiogenesis is reduced in the Tc1 mouse model of Down’s syndrome

Louise E. Reynolds; Alan Watson; Marianne Baker; Tania A. Jones; Gabriela D'Amico; Stephen Robinson; Carine Joffre; Sarah Garrido-Urbani; Juan Carlos Rodríguez-Manzaneque; Estefanía Martino-Echarri; Michel Aurrand-Lions; Denise Sheer; Franca Dagna-Bricarelli; Dean Nizetic; Christopher J. McCabe; Andrew Turnell; Stéphanie Kermorgant; Beat A. Imhof; Ralf H. Adams; Elizabeth M. C. Fisher; Victor L. J. Tybulewicz; Ian R. Hart; Kairbaan Hodivala-Dilke

Down’s syndrome (DS) is a genetic disorder caused by full or partial trisomy of human chromosome 21 and presents with many clinical phenotypes including a reduced incidence of solid tumours. Recent work with the Ts65Dn model of DS, which has orthologues of about 50% of the genes on chromosome 21 (Hsa21), has indicated that three copies of the ETS2 (ref. 3) or DS candidate region 1 (DSCR1) genes (a previously known suppressor of angiogenesis) is sufficient to inhibit tumour growth. Here we use the Tc1 transchromosomic mouse model of DS to dissect the contribution of extra copies of genes on Hsa21 to tumour angiogenesis. This mouse expresses roughly 81% of Hsa21 genes but not the human DSCR1 region. We transplanted B16F0 and Lewis lung carcinoma tumour cells into Tc1 mice and showed that growth of these tumours was substantially reduced compared with wild-type littermate controls. Furthermore, tumour angiogenesis was significantly repressed in Tc1 mice. In particular, in vitro and in vivo angiogenic responses to vascular endothelial growth factor (VEGF) were inhibited. Examination of the genes on the segment of Hsa21 in Tc1 mice identified putative anti-angiogenic genes (ADAMTS1and ERG) and novel endothelial cell-specific genes, never previously shown to be involved in angiogenesis (JAM-B and PTTG1IP), that, when overexpressed, are responsible for inhibiting angiogenic responses to VEGF. Three copies of these genes within the stromal compartment reduced tumour angiogenesis, explaining the reduced tumour growth in DS. Furthermore, we expect that, in addition to the candidate genes that we show to be involved in the repression of angiogenesis, the Tc1 mouse model of DS will permit the identification of other endothelium-specific anti-angiogenic targets relevant to a broad spectrum of cancer patients.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

Loss of Endothelial Tie1 Receptor Impairs Lymphatic Vessel Development-Brief Report

Gabriela D'Amico; Emilia A. Korhonen; Marika Waltari; Pipsa Saharinen; Pirjo Laakkonen; Kari Alitalo

Objective—Studies of Tie1 gene-targeted embryos have demonstrated loss of blood vessel integrity, but the relevance of Tie1 in lymphatic vasculature development is unknown. We tested the hypothesis that the swelling observed in Tie1 mutant embryos is associated with lymphatic vascular defects. Methods and Results—We could extend the survival of the Tie1-deficient embryos in the ICR background, which allowed us to study their lymphatic vessel development. At embryonic day (E) 14.5, the Tie1−/− embryos had edema and hemorrhages and began to die. Immunohistochemical analysis revealed that they have abnormal lymph sacs. Tie1−/− mutants were swollen already at E12.5 without signs of hemorrhage. Their lymph sacs were abnormally patterned, suggesting that lymphatic malformations precede the blood vascular defects. We generated mice with a conditional Cre/loxP Tie1neo locus and found that the homozygous Tie1neo/neo hypomorphic embryos survived until E15.5 with lymphatic malformations resembling those seen in the Tie1−/− mutants. Conclusion—Our data show that loss of Tie1 results in lymphatic vascular abnormalities that precede the blood vessel phenotype. These findings indicate that Tie1 is involved in lymphangiogenesis and suggest differential requirements for Tie1 signaling in the two vascular compartments.


Genes & Development | 2014

Angiopoietin 2 regulates the transformation and integrity of lymphatic endothelial cell junctions

Wei Zheng; Harri Nurmi; Sila Appak; Amélie Sabine; Esther Bovay; Emilia A. Korhonen; Fabrizio Orsenigo; Marja Lohela; Gabriela D'Amico; Tanja Holopainen; Ching Ching Leow; Elisabetta Dejana; Tatiana V. Petrova; Hellmut G. Augustin; Kari Alitalo

Primitive lymphatic vessels are remodeled into functionally specialized initial and collecting lymphatics during development. Lymphatic endothelial cell (LEC) junctions in initial lymphatics transform from a zipper-like to a button-like pattern during collecting vessel development, but what regulates this process is largely unknown. Angiopoietin 2 (Ang2) deficiency leads to abnormal lymphatic vessels. Here we found that an ANG2-blocking antibody inhibited embryonic lymphangiogenesis, whereas endothelium-specific ANG2 overexpression induced lymphatic hyperplasia. ANG2 inhibition blocked VE-cadherin phosphorylation at tyrosine residue 685 and the concomitant formation of button-like junctions in initial lymphatics. The defective junctions were associated with impaired lymph uptake. In collecting lymphatics, adherens junctions were disrupted, and the vessels leaked upon ANG2 blockade or gene deletion. ANG2 inhibition also suppressed the onset of lymphatic valve formation and subsequent valve maturation. These data identify ANG2 as the first essential regulator of the functionally important interendothelial cell-cell junctions that form during lymphatic development.


Development | 2009

Regulation of lymphatic-blood vessel separation by endothelial Rac1

Gabriela D'Amico; Dylan T. Jones; Emma Nye; Karen Sapienza; Antoine R. Ramjuan; Louise E. Reynolds; Stephen Robinson; Vassiliki Kostourou; Dolores Martinez; Deborah Aubyn; Richard Grose; Gareth J. Thomas; Bradley Spencer-Dene; Daniel Zicha; Derek Davies; Victor L. J. Tybulewicz; Kairbaan Hodivala-Dilke

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+;Rac1fl/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.


The Journal of Pathology | 2009

Genetic ablation of the alpha 6-integrin subunit in Tie1Cre mice enhances tumour angiogenesis†

Mitchel Germain; Adèle De Arcangelis; Stephen Robinson; Marianne Baker; Bernardo Tavora; Gabriela D'Amico; Rita Silva; Vassiliki Kostourou; Louise E. Reynolds; Alan Watson; J. Louise Jones; Elisabeth Georges-Labouesse; Kairbaan Hodivala-Dilke

Laminins are expressed highly in blood vessel basement membranes and have been implicated in angiogenesis. α6β1‐ and α6β4‐integrins are major receptors for laminins in endothelial cells, but the precise role of endothelial α6‐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 α6‐integrin‐subunit when compared with normal breast tissue. These data suggest that a decrease in α6‐integrin‐subunit expression in endothelial cells is associated with tumour angiogenesis. To test whether the loss of the endothelial α6‐integrin subunit affects tumour growth and angiogenesis, we generated α6fl/fl‐Tie1Cre+ mice and showed that endothelial deletion of α6‐integrin is sufficient to enhance tumour size and tumour angiogenesis in both murine B16F0 melanoma and Lewis cell lung carcinoma. Mechanistically, endothelial α6‐integrin deficiency elevated significantly VEGF‐mediated angiogenesis both in vivo and ex vivo. In particular, α6‐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 α6‐knockout mice, we show that the expression of the α6‐integrin subunit in endothelial cells acts as a negative regulator of angiogenesis both in vivo and ex vivo. Copyright


Genes & Development | 2010

Claudin-like protein 24 interacts with the VEGFR-2 and VEGFR-3 pathways and regulates lymphatic vessel development

Pipsa Saharinen; Hanna Heloterä; Juho Miettinen; Camilla Norrmén; Gabriela D'Amico; Michael Jeltsch; Tobias Langenberg; Wouter Vandevelde; Annelii Ny; Mieke Dewerchin; Peter Carmeliet; Kari Alitalo

The Claudin-like protein of 24 kDa (CLP24) is a hypoxia-regulated transmembrane protein of unknown function. We show here that clp24 knockdown in Danio rerio and Xenopus laevis results in defective lymphatic development. Targeted disruption of Clp24 in mice led to enlarged lymphatic vessels having an abnormal smooth muscle cell coating. We also show that the Clp24(-/-) phenotype was further aggravated in the Vegfr2(+/LacZ) or Vegfr3(+/LacZ) backgrounds and that CLP24 interacts with vascular endothelial growth factor receptor-2 (VEGFR-2) and VEGFR-3 and attenuates the transcription factor CREB phosphorylation via these receptors. Our results indicate that CLP24 is a novel regulator of VEGFR-2 and VEGFR-3 signaling pathways and of normal lymphatic vessel structure.


PLOS ONE | 2010

Endothelial-Rac1 Is Not Required for Tumor Angiogenesis unless αvβ3-Integrin Is Absent

Gabriela D'Amico; Stephen Robinson; Mitchel Germain; Louise E. Reynolds; Gareth J. Thomas; George Elia; Garry Saunders; Marcus Fruttiger; Victor A Tybulewicz; Georgia Mavria; Kairbaan Hodivala-Dilke

Endothelial cell migration is an essential aspect of tumor angiogenesis. Rac1 activity is needed for cell migration in vitro implying a requirement for this molecule in angiogenesis in vivo. However, a precise role for Rac1 in tumor angiogenesis has never been addressed. Here we show that depletion of endothelial Rac1 expression in adult mice, unexpectedly, has no effect on tumor growth or tumor angiogenesis. In addition, repression of Rac1 expression does not inhibit VEGF-mediated angiogenesis in vivo or ex vivo, nor does it affect chemotactic migratory responses to VEGF in 3-dimensions. In contrast, the requirement for Rac1 in tumor growth and angiogenesis becomes important when endothelial β3-integrin levels are reduced or absent: the enhanced tumor growth, tumor angiogenesis and VEGF-mediated responses in β3-null mice are all Rac1-dependent. These data indicate that in the presence of αvβ3-integrin Rac1 is not required for tumor angiogenesis.


Cancer Research | 2012

Deletion of the endothelial Bmx tyrosine kinase decreases tumor angiogenesis and growth

Tanja Holopainen; Vanessa López Alpuche; Wei Zheng; Ritva Heljasvaara; Dennis Jones; Yun He; Denis Tvorogov; Gabriela D'Amico; Zoltán Wiener; Leif C. Andersson; Taina Pihlajaniemi; Wang Min; Kari Alitalo

Bmx, [corrected] also known as Etk, is a member of the Tec family of nonreceptor tyrosine kinases. Bmx is expressed mainly in arterial endothelia and in myeloid hematopoietic cells. Bmx regulates ischemia-mediated arteriogenesis and lymphangiogenesis, but its role in tumor angiogenesis is not known. In this study, we characterized the function of Bmx in tumor growth using both Bmx knockout and transgenic mice. Isogenic colon, lung, and melanoma tumor xenotransplants showed reductions in growth and tumor angiogenesis in Bmx gene-deleted ((-/-)) mice, whereas developmental angiogenesis was not affected. In addition, growth of transgenic pancreatic islet carcinomas and intestinal adenomas was also slower in Bmx(-/-) mice. Knockout mice showed high levels of Bmx expression in endothelial cells of tumor-associated and peritumoral arteries. Moreover, endothelial cells lacking Bmx showed impaired phosphorylation of extracellular signal-regulated kinase (Erk) upon VEGF stimulation, indicating that Bmx contributes to the transduction of vascular endothelial growth factor signals. In transgenic mice overexpressing Bmx in epidermal keratinocytes, tumors induced by a two-stage chemical skin carcinogenesis treatment showed increased growth and angiogenesis. Our findings therefore indicate that Bmx activity contributes to tumor angiogenesis and growth.

Collaboration


Dive into the Gabriela D'Amico's collaboration.

Top Co-Authors

Avatar

Kairbaan Hodivala-Dilke

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Louise E. Reynolds

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Bernardo Tavora

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tanguy Lechertier

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marianne Baker

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Annika N Alexopoulou

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Delphine M. Lees

Queen Mary University of London

View shared research outputs
Researchain Logo
Decentralizing Knowledge