Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(13)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35723257

RESUMEN

Precise vascular patterning is crucial for normal growth and development. The ERG transcription factor drives Delta-like ligand 4 (DLL4)/Notch signalling and is thought to act as a pivotal regulator of endothelial cell (EC) dynamics and developmental angiogenesis. However, molecular regulation of ERG activity remains obscure. Using a series of EC-specific focal adhesion kinase (FAK)-knockout (KO) and point-mutant FAK-knock-in mice, we show that loss of ECFAK, its kinase activity or phosphorylation at FAK-Y397, but not FAK-Y861, reduces ERG and DLL4 expression levels together with concomitant aberrations in vascular patterning. Rapid immunoprecipitation mass spectrometry of endogenous proteins identified that endothelial nuclear-FAK interacts with the deubiquitinase USP9x and the ubiquitin ligase TRIM25. Further in silico analysis confirms that ERG interacts with USP9x and TRIM25. Moreover, ERG levels are reduced in FAKKO ECs via a ubiquitin-mediated post-translational modification programme involving USP9x and TRIM25. Re-expression of ERG in vivo and in vitro rescues the aberrant vessel-sprouting defects observed in the absence of ECFAK. Our findings identify ECFAK as a regulator of retinal vascular patterning by controlling ERG protein degradation via TRIM25/USP9x.


Asunto(s)
Células Endoteliales , Factores de Transcripción , Animales , Células Endoteliales/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Ratones , Neovascularización Fisiológica/genética , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitinas/metabolismo
2.
Genes Dev ; 28(14): 1592-603, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25030698

RESUMEN

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.


Asunto(s)
Angiopoyetina 2/metabolismo , Células Endoteliales/metabolismo , Uniones Intercelulares/fisiología , Angiopoyetina 2/antagonistas & inhibidores , Angiopoyetina 2/genética , Animales , Cadherinas/metabolismo , Embrión de Mamíferos , Células Endoteliales/citología , Eliminación de Gen , Linfangiogénesis/fisiología , Tejido Linfoide/embriología , Tejido Linfoide/patología , Ratones , Ratones Endogámicos C57BL , Fosforilación
3.
J Pathol ; 249(4): 523-535, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31424556

RESUMEN

Coronary microvascular dysfunction combined with maladaptive cardiomyocyte morphology and energetics is a major contributor to heart failure advancement. Thus, dually enhancing cardiac angiogenesis and targeting cardiomyocyte function to slow, or reverse, the development of heart failure is a logical step towards improved therapy. We present evidence for the potential to repurpose a former anti-cancer Arg-Gly-Asp (RGD)-mimetic pentapeptide, cilengitide, here used at low doses. Cilengitide targets αvß3 integrin and this protein is upregulated in human dilated and ischaemic cardiomyopathies. Treatment of mice after abdominal aortic constriction (AAC) surgery with low-dose cilengitide (ldCil) enhances coronary angiogenesis and directly affects cardiomyocyte hypertrophy with an associated reduction in disease severity. At a molecular level, ldCil treatment has a direct effect on cardiac endothelial cell transcriptomic profiles, with a significant enhancement of pro-angiogenic signalling pathways, corroborating the enhanced angiogenic phenotype after ldCil treatment. Moreover, ldCil treatment of Angiotensin II-stimulated AngII-stimulated cardiomyocytes significantly restores transcriptomic profiles similar to those found in normal human heart. The significance of this finding is enhanced by transcriptional similarities between AngII-treated cardiomyocytes and failing human hearts. Taken together, our data provide evidence supporting a possible new strategy for improved heart failure treatment using low-dose RGD-mimetics with relevance to human disease. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Fármacos Cardiovasculares/farmacología , Reposicionamiento de Medicamentos , Insuficiencia Cardíaca/tratamiento farmacológico , Integrina alfaVbeta3/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Venenos de Serpiente/farmacología , Angiotensina II/farmacología , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Integrina alfaVbeta3/metabolismo , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Neovascularización Fisiológica/efectos de los fármacos , Recuperación de la Función , Transducción de Señal , Transcriptoma
4.
Eur Heart J ; 40(30): 2507-2520, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31162546

RESUMEN

AIMS: A better understanding of the pathways that regulate regeneration of the coronary vasculature is of fundamental importance for the advancement of strategies to treat patients with heart disease. Here, we aimed to investigate the origin and clonal dynamics of endothelial cells (ECs) associated with neovascularization in the adult mouse heart following myocardial infarction (MI). Furthermore, we sought to define murine cardiac endothelial heterogeneity and to characterize the transcriptional profiles of pro-angiogenic resident ECs in the adult mouse heart, at single-cell resolution. METHODS AND RESULTS: An EC-specific multispectral lineage-tracing mouse (Pdgfb-iCreERT2-R26R-Brainbow2.1) was used to demonstrate that structural integrity of adult cardiac endothelium following MI was maintained through clonal proliferation by resident ECs in the infarct border region, without significant contributions from bone marrow cells or endothelial-to-mesenchymal transition. Ten transcriptionally discrete heterogeneous EC states, as well as the pathways through which each endothelial state is likely to enhance neovasculogenesis and tissue regeneration following ischaemic injury were defined. Plasmalemma vesicle-associated protein (Plvap) was selected for further study, which showed an endothelial-specific and increased expression in both the ischaemic mouse and human heart, and played a direct role in regulating human endothelial proliferation in vitro. CONCLUSION: We present a single-cell gene expression atlas of cardiac specific resident ECs, and the transcriptional hierarchy underpinning endogenous vascular repair following MI. These data provide a rich resource that could assist in the development of new therapeutic interventions to augment endogenous myocardial perfusion and enhance regeneration in the injured heart.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Infarto del Miocardio/metabolismo , Neovascularización Fisiológica/genética , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Animales , Proliferación Celular/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Transgénicos , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología
5.
J Cell Sci ; 130(9): 1583-1595, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28289267

RESUMEN

The α6ß1-integrin is a major laminin receptor, and formation of a laminin-rich basement membrane is a key feature in tumour blood vessel stabilisation and pericyte recruitment, processes that are important in the growth and maturation of tumour blood vessels. However, the role of pericyte α6ß1-integrin in angiogenesis is largely unknown. We developed mice where the α6-integrin subunit is deleted in pericytes and examined tumour angiogenesis and growth. These mice had: (1) reduced pericyte coverage of tumour blood vessels; (2) reduced tumour blood vessel stability; (3) increased blood vessel diameter; (4) enhanced blood vessel leakiness, and (5) abnormal blood vessel basement membrane architecture. Surprisingly, tumour growth, blood vessel density and metastasis were not altered. Analysis of retinas revealed that deletion of pericyte α6-integrin did not affect physiological angiogenesis. At the molecular level, we provide evidence that pericyte α6-integrin controls PDGFRß expression and AKT-mTOR signalling. Taken together, we show that pericyte α6ß1-integrin regulates tumour blood vessels by both controlling PDGFRß and basement membrane architecture. These data establish a novel dual role for pericyte α6-integrin as modulating the blood vessel phenotype during pathological angiogenesis.


Asunto(s)
Vasos Sanguíneos/metabolismo , Integrina alfa6beta1/metabolismo , Neoplasias/irrigación sanguínea , Pericitos/metabolismo , Animales , Membrana Basal/efectos de los fármacos , Membrana Basal/metabolismo , Becaplermina , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Integrasas/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Pericitos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
6.
Circ Res ; 120(9): 1414-1425, 2017 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-28298294

RESUMEN

RATIONALE: Vascular endothelial growth factor (VEGF) is the main driver of angiogenesis and vascular permeability via VEGF receptor 2 (VEGFR2), whereas lymphangiogenesis signals are transduced by VEGFC/D via VEGFR3. VEGFR3 also regulates sprouting angiogenesis and blood vessel growth, but to what extent VEGFR3 signaling controls blood vessel permeability remains unknown. OBJECTIVE: To investigate the role of VEGFR3 in the regulation of VEGF-induced vascular permeability. METHODS AND RESULTS: Long-term global Vegfr3 gene deletion in adult mice resulted in increased fibrinogen deposition in lungs and kidneys, indicating enhanced vascular leakage at the steady state. Short-term deletion of Vegfr3 in blood vascular endothelial cells increased baseline leakage in various tissues, as well as in tumors, and exacerbated vascular permeability in response to VEGF, administered via intradermal adenoviral delivery or through systemic injection of recombinant protein. VEGFR3 gene silencing upregulated VEGFR2 protein levels and phosphorylation in cultured endothelial cells. Consistent with elevated VEGFR2 activity, vascular endothelial cadherin showed reduced localization at endothelial cell-cell junctions in postnatal retinas after Vegfr3 deletion, or after VEGFR3 silencing in cultured endothelial cells. Furthermore, concurrent deletion of Vegfr2 prevented VEGF-induced excessive vascular leakage in mice lacking Vegfr3. CONCLUSIONS: VEGFR3 limits VEGFR2 expression and VEGF/VEGFR2 pathway activity in quiescent and angiogenic blood vascular endothelial cells, thereby preventing excessive vascular permeability.


Asunto(s)
Permeabilidad Capilar , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Células Endoteliales/metabolismo , Pulmón/irrigación sanguínea , Vasos Retinianos/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Uniones Adherentes/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Carcinoma Pulmonar de Lewis/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Femenino , Genotipo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica , Neovascularización Fisiológica , Fenotipo , Vasos Retinianos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Uniones Estrechas/metabolismo , Transfección , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/deficiencia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/deficiencia , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
7.
J Pathol ; 242(3): 358-370, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28444899

RESUMEN

Focal adhesion kinase (FAK) inhibitors have been developed as potential anticancer agents and are undergoing clinical trials. In vitro activation of the FAK kinase domain triggers autophosphorylation of Y397, Src activation, and subsequent phosphorylation of other FAK tyrosine residues. However, how FAK Y397 mutations affect FAK kinase-dead (KD) phenotypes in tumour angiogenesis in vivo is unknown. We developed three Pdgfb-iCreert -driven endothelial cell (EC)-specific, tamoxifen-inducible homozygous mutant mouse lines: FAK wild-type (WT), FAK KD, and FAK double mutant (DM), i.e. KD with a putatively phosphomimetic Y397E mutation. These ECCre+;FAKWT/WT , ECCre+;FAKKD/KD and ECCre+;FAKDM/DM mice were injected subcutaneously with syngeneic B16F0 melanoma cells. Tumour growth and tumour blood vessel functions were unchanged between ECCre+;FAKWT/WT and ECCre-;FAKWT/WT control mice. In contrast, tumour growth and vessel density were decreased in ECCre+;FAKKD/KD and ECCre+;FAKDM/DM mice, as compared with Cre - littermates. Despite no change in the percentage of perfused vessels or pericyte coverage in either genotype, tumour hypoxia was elevated in ECCre+;FAKKD/KD and ECCre+;FAKDM/DM mice. Furthermore, although ECCre+;FAKKD/KD mice showed reduced blood vessel leakage, ECCre+;FAKDM/DM and ECCre-;FAKDM/DM mice showed no difference in leakage. Mechanistically, fibronectin-stimulated Y397 autophosphorylation was reduced in Cre+;FAKKD/KD ECs as compared with Cre+;FAKWT/WT cells, with no change in phosphorylation of the known Src targets FAK-Y577, FAK-Y861, FAK-Y925, paxillin-Y118, p130Cas-Y410. Cre+;FAKDM/DM ECs showed decreased Src target phosphorylation levels, suggesting that the Y397E substitution actually disrupted Src activation. Reduced VE-cadherin-pY658 levels in Cre+;FAKKD/KD ECs were rescued in Cre+FAKDM/DM ECs, corresponding with the rescue in vessel leakage in the ECCre+;FAKDM/DM mice. We show that EC-specific FAK kinase activity is required for tumour growth, angiogenesis, and vascular permeability. The ECCre+;FAKDM/DM mice restored the KD-dependent tumour vascular leakage observed in ECCre+;FAKKD/KD mice in vivo. This study opens new fields in in vivo FAK signalling. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Permeabilidad Capilar/genética , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Melanoma/enzimología , Animales , Antineoplásicos Hormonales/farmacología , Permeabilidad Capilar/efectos de los fármacos , División Celular/genética , Hipoxia de la Célula/genética , Línea Celular Tumoral , Endotelio Vascular/enzimología , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/deficiencia , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Homocigoto , Melanoma/irrigación sanguínea , Melanoma/genética , Ratones , Mutación/genética , Trasplante de Neoplasias , Neovascularización Patológica/enzimología , Neovascularización Patológica/genética , Tamoxifeno/farmacología
8.
Genes Dev ; 24(9): 875-80, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20439428

RESUMEN

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.


Asunto(s)
Vasos Linfáticos/embriología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Transducción de Señal/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Vasos Linfáticos/patología , Ratones , Miocitos del Músculo Liso/patología , Fosforilación , Piel/citología
9.
Nature ; 465(7299): 813-7, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20535211

RESUMEN

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.


Asunto(s)
Carcinoma Pulmonar de Lewis/irrigación sanguínea , Modelos Animales de Enfermedad , Síndrome de Down/genética , Dosificación de Gen/genética , Melanoma Experimental/irrigación sanguínea , Neovascularización Patológica/genética , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS1 , Animales , Carcinoma Pulmonar de Lewis/complicaciones , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Cromosomas de los Mamíferos/genética , Síndrome de Down/complicaciones , Síndrome de Down/fisiopatología , Femenino , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Masculino , Melanoma Experimental/complicaciones , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Trasplante de Neoplasias , Neovascularización Patológica/patología , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteína Proto-Oncogénica c-ets-2/genética , Proteína Proto-Oncogénica c-ets-2/metabolismo , Factores de Transcripción , Regulador Transcripcional ERG , Trisomía/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
Development ; 136(23): 4043-53, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19906871

RESUMEN

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.


Asunto(s)
Vasos Sanguíneos/metabolismo , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Vasos Linfáticos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Separación Celular/métodos , Células Cultivadas , Embrión de Mamíferos , Endotelio Vascular/citología , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Técnica del Anticuerpo Fluorescente Directa , Colorantes Fluorescentes/metabolismo , Galactósidos/metabolismo , Eliminación de Gen , Inmunohistoquímica , Indoles/metabolismo , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , ARN Interferente Pequeño/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Transfección , beta-Galactosidasa/metabolismo , Proteína de Unión al GTP rac1/análisis , Proteína de Unión al GTP rac1/genética
11.
Methods Mol Biol ; 2441: 3-18, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35099724

RESUMEN

Blood vessel growth is a fundamental process for organ development and wound healing but is also associated with ischemic diseases and cancer. The growth of new blood vessels from preexisting vasculature, termed sprouting angiogenesis, is the predominant mode of blood vessel growth in central nervous system vascularization and pathological vessel growth. Accordingly, studying the molecular and cellular mechanisms of angiogenesis holds the promise to find novel therapeutic targets to stimulate new vessel formation in ischemic tissues or inhibit pathological vessel growth in disease. The embryonic mouse hindbrain provides an excellent model to study sprouting angiogenesis in vivo by histochemical or fluorescent wholemount immunolabeling, thus allowing high-resolution image capture of nascent vasculature and subsequent quantification of relevant angiogenic parameters. This chapter describes how to use the mouse embryonic hindbrain as a model to study physiological angiogenesis, including detailed protocols for hindbrain dissection, wholemount staining, and angiogenic parameters analysis.


Asunto(s)
Neovascularización Fisiológica , Rombencéfalo , Animales , Modelos Animales de Enfermedad , Ratones , Neovascularización Patológica , Rombencéfalo/irrigación sanguínea , Coloración y Etiquetado
12.
Arterioscler Thromb Vasc Biol ; 30(2): 207-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19910638

RESUMEN

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 Tie1(neo) locus and found that the homozygous Tie1(neo/neo) hypomorphic embryos survived until E15.5 with lymphatic malformations resembling those seen in the Tie1(-/-) mutants. CONCLUSIONS: 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.


Asunto(s)
Células Endoteliales/enzimología , Linfangiogénesis , Vasos Linfáticos/enzimología , Receptor TIE-1/metabolismo , Animales , Edema/enzimología , Edema/fisiopatología , Pérdida del Embrión , Células Endoteliales/patología , Edad Gestacional , Hemorragia/enzimología , Hemorragia/fisiopatología , Homocigoto , Inmunohistoquímica , Linfangiogénesis/genética , Vasos Linfáticos/embriología , Vasos Linfáticos/fisiopatología , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Fenotipo , Receptor TIE-1/deficiencia , Receptor TIE-1/genética
13.
J Pathol ; 220(3): 370-81, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19967723

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Integrina alfa6/genética , Proteínas de Neoplasias/genética , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/genética , Animales , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/irrigación sanguínea , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Genotipo , Humanos , Integrina alfa6/metabolismo , Integrina alfa6/fisiología , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Trasplante de Neoplasias , Neoplasias Experimentales/metabolismo , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Factor A de Crecimiento Endotelial Vascular/toxicidad , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
EMBO Mol Med ; 12(2): e11663, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31858727

RESUMEN

Pathological angiogenesis contributes to tumour progression as well as to chronic inflammatory diseases. In this issue of EMBO Molecular Medicine, Esteban and co-workers identify endothelial cell MT1-MMP as a key regulator of intussusceptive angiogenesis (IA) in inflammatory colitis. Thrombospondin 1 (TSP1) cleavage by MT1-MMP results in the binding of the c-terminal fragment of TSP1 to αvß3 integrin, which induces nitric oxide (NO) production, vasodilation and further initiation of IA. This novel control mechanism of inflammatory IA points towards promising new therapeutic targets for inflammatory bowel disease.


Asunto(s)
Metaloproteinasa 14 de la Matriz , Metaloendopeptidasas , Células Endoteliales , Humanos , Metaloproteinasas de la Matriz Asociadas a la Membrana , Neovascularización Patológica
15.
Nat Commun ; 11(1): 2810, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32499572

RESUMEN

The overexpression of the protein tyrosine kinase, Focal adhesion kinase (FAK), in endothelial cells has implicated its requirement in angiogenesis and tumour growth, but how pericyte FAK regulates tumour angiogenesis is unknown. We show that pericyte FAK regulates tumour growth and angiogenesis in multiple mouse models of melanoma, lung carcinoma and pancreatic B-cell insulinoma and provide evidence that loss of pericyte FAK enhances Gas6-stimulated phosphorylation of the receptor tyrosine kinase, Axl with an upregulation of Cyr61, driving enhanced tumour growth. We further show that pericyte derived Cyr61 instructs tumour cells to elevate expression of the proangiogenic/protumourigenic transmembrane receptor Tissue Factor. Finally, in human melanoma we show that when 50% or more tumour blood vessels are pericyte-FAK negative, melanoma patients are stratified into those with increased tumour size, enhanced blood vessel density and metastasis. Overall our data uncover a previously unknown mechanism of tumour growth by pericytes that is controlled by pericyte FAK.


Asunto(s)
Proteína 61 Rica en Cisteína/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neovascularización Patológica , Pericitos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Aorta Torácica/patología , Carcinoma Pulmonar de Lewis/metabolismo , Adhesión Celular , Proliferación Celular , Femenino , Quinasa 1 de Adhesión Focal/genética , Humanos , Linfocinas/metabolismo , Masculino , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/patología , Factor de Crecimiento Placentario/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Transducción de Señal , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Tirosina Quinasa del Receptor Axl
16.
Cancer Res ; 80(16): 3345-3358, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32586981

RESUMEN

Platelet-derived growth factor B (PDGFB) plays a crucial role in recruitment of PDGF receptor ß-positive pericytes to blood vessels. The endothelium is an essential source of PDGFB in this process. Platelets constitute a major reservoir of PDGFB and are continuously activated in the tumor microenvironment, exposing tumors to the plethora of growth factors contained in platelet granules. Here, we show that tumor vascular function, as well as pericyte coverage is significantly impaired in mice with conditional knockout of PDGFB in platelets. A lack of PDGFB in platelets led to enhanced hypoxia and epithelial-to-mesenchymal transition in the primary tumors, elevated levels of circulating tumor cells, and increased spontaneous metastasis to the liver or lungs in two mouse models. These findings establish a previously unknown role for platelet-derived PDGFB, whereby it promotes and maintains vascular integrity in the tumor microenvironment by contributing to the recruitment of pericytes. SIGNIFICANCE: Conditional knockout of PDGFB in platelets demonstrates its previously unknown role in the maintenance of tumor vascular integrity and host protection against metastasis.


Asunto(s)
Movimiento Celular , Endotelio Vascular/metabolismo , Pericitos/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Animales , Vasos Sanguíneos , Neoplasias del Colon/irrigación sanguínea , Transición Epitelial-Mesenquimal , Matriz Extracelular , Técnicas de Inactivación de Genes , Hibridación Genética , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Melanoma/irrigación sanguínea , Melanoma/secundario , Ratones , Células Neoplásicas Circulantes , Neoplasias Pancreáticas , Pericitos/metabolismo , Activación Plaquetaria/fisiología , Proteínas Proto-Oncogénicas c-sis/deficiencia , Proteínas Proto-Oncogénicas c-sis/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Trombocitopenia , Hipoxia Tumoral , Microambiente Tumoral
17.
Arterioscler Thromb Vasc Biol ; 28(10): 1703-13, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18658045

RESUMEN

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.


Asunto(s)
Endotelio Vascular/metabolismo , Integrinas/metabolismo , Neovascularización Fisiológica , Transducción de Señal , Inhibidores de la Angiogénesis/farmacología , Animales , Endotelio Vascular/efectos de los fármacos , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Pericitos/metabolismo , Receptores de Colágeno/metabolismo , Receptores de Fibronectina/metabolismo , Receptores de Laminina/metabolismo , Receptores de Vitronectina/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Blood ; 116(4): 512-3, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20671134
20.
J Clin Invest ; 126(9): 3495-510, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27548530

RESUMEN

The angiopoietin/Tie (ANG/Tie) receptor system controls developmental and tumor angiogenesis, inflammatory vascular remodeling, and vessel leakage. ANG1 is a Tie2 agonist that promotes vascular stabilization in inflammation and sepsis, whereas ANG2 is a context-dependent Tie2 agonist or antagonist. A limited understanding of ANG signaling mechanisms and the orphan receptor Tie1 has hindered development of ANG/Tie-targeted therapeutics. Here, we determined that both ANG1 and ANG2 binding to Tie2 increases Tie1-Tie2 interactions in a ß1 integrin-dependent manner and that Tie1 regulates ANG-induced Tie2 trafficking in endothelial cells. Endothelial Tie1 was essential for the agonist activity of ANG1 and autocrine ANG2. Deletion of endothelial Tie1 in mice reduced Tie2 phosphorylation and downstream Akt activation, increased FOXO1 nuclear localization and transcriptional activation, and prevented ANG1- and ANG2-induced capillary-to-venous remodeling. However, in acute endotoxemia, the Tie1 ectodomain that is responsible for interaction with Tie2 was rapidly cleaved, ANG1 agonist activity was decreased, and autocrine ANG2 agonist activity was lost, which led to suppression of Tie2 signaling. Tie1 cleavage also occurred in patients with hantavirus infection. These results support a model in which Tie1 directly interacts with Tie2 to promote ANG-induced vascular responses under noninflammatory conditions, whereas in inflammation, Tie1 cleavage contributes to loss of ANG2 agonist activity and vascular stability.


Asunto(s)
Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Inflamación , Receptor TIE-1/metabolismo , Receptor TIE-2/metabolismo , Remodelación Vascular , Adulto , Anciano , Animales , Estudios de Casos y Controles , Estudios de Cohortes , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Endotoxemia/metabolismo , Femenino , Eliminación de Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina beta1/metabolismo , Lipopolisacáridos/química , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Fosforilación , Sepsis , Transducción de Señal , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA