Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 18(7): e1010733, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35849637

RESUMEN

Emerging SARS-CoV-2 variants are creating major challenges in the ongoing COVID-19 pandemic. Being able to predict mutations that could arise in SARS-CoV-2 leading to increased transmissibility or immune evasion would be extremely valuable in development of broad-acting therapeutics and vaccines, and prioritising viral monitoring and containment. Here we use in vitro evolution to seek mutations in SARS-CoV-2 receptor binding domain (RBD) that would substantially increase binding to ACE2. We find a double mutation, S477N and Q498H, that increases affinity of RBD for ACE2 by 6.5-fold. This affinity gain is largely driven by the Q498H mutation. We determine the structure of the mutant-RBD:ACE2 complex by cryo-electron microscopy to reveal the mechanism for increased affinity. Addition of Q498H to SARS-CoV-2 RBD variants is found to boost binding affinity of the variants for human ACE2 and confer a new ability to bind rat ACE2 with high affinity. Surprisingly however, in the presence of the common N501Y mutation, Q498H inhibits binding, due to a clash between H498 and Y501 side chains. To achieve an intermolecular bonding network, affinity gain and cross-species binding similar to Q498H alone, RBD variants with the N501Y mutation must acquire instead the related Q498R mutation. Thus, SARS-CoV-2 RBD can access large affinity gains and cross-species binding via two alternative mutational routes involving Q498, with route selection determined by whether a variant already has the N501Y mutation. These mutations are now appearing in emerging SARS-CoV-2 variants where they have the potential to influence human-to-human and cross-species transmission.


Asunto(s)
COVID-19 , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/genética , Animales , COVID-19/genética , Microscopía por Crioelectrón , Humanos , Mutación , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Unión Proteica , Ratas , Receptores Virales/metabolismo , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
2.
J Biol Chem ; 297(2): 100888, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34153320

RESUMEN

Angiopoietins Ang1 and Ang2 are secreted ligands for the endothelial receptor tyrosine kinase Tie2 essential for vascular development and maintenance. Ang1 acts as an agonist to maintain normal vessel function, whereas Ang2 acts as a Tie2 antagonist. Ang2 is increased in macular edema, sepsis, and other conditions, in which it blocks Ang1-mediated signaling, causing vascular dysfunction and contributing to disease pathology. Therefore, Ang2 is an attractive therapeutic target. Previously, we reported a Tie2 ectodomain variant that selectively binds Ang2 and acts as soluble ligand trap to sequester Ang2; however, the mechanism of Ang2-binding selectivity is unknown. In the present study, we used directed protein evolution to enhance Ang2-binding affinity of this Tie2 ectodomain trap. We examined contributions of individual residues in the ligand-binding interface of Tie2 to Ang1 and Ang2 binding. Surprisingly, different combinations of Tie2 residues were found to bind each ligand, with hydrophobic residues binding both ligands and polar residues contributing selectively to either Ang1 or Ang2 binding. Our analysis also identified a single Tie2 residue, His168, with a pivotal role in both Ang1 and Ang2 binding, enabling competition between binding ligands. In summary, this study reports an enhanced-affinity Ang2-selective ligand trap with potential for therapeutic development and reveals the mechanism behind its selectivity. It also provides the first analysis of contributions of individual Tie2 residues to Ang1 and Ang2 binding and identifies selectivity-determining residues that could be targeted in the future design of small molecule and other inhibitors of Ang2 for the treatment of vascular dysfunction.


Asunto(s)
Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Evolución Molecular Dirigida/métodos , Receptor TIE-2/metabolismo , Angiopoyetina 1/química , Angiopoyetina 2/química , Células Cultivadas , Humanos , Ligandos , Neovascularización Fisiológica , Unión Proteica , Receptor TIE-2/química , Transducción de Señal
3.
Sci Rep ; 7(1): 3658, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28623351

RESUMEN

Angiopoietin-1 (Angpt1) is a glycoprotein ligand important for maintaining the vascular system. It signals via a receptor tyrosine kinase expressed on the surface on endothelial cells, Tie2. This receptor can undergo regulated ectodomain cleavage that releases the ligand-binding domain (sTie2) into the circulation. The concentration of sTie2 is increased in a range of conditions, including peripheral arterial disease and myocardial infarction, where it has been suggested to bind and block Angpt1 resulting in vascular dysfunction. Here we use a joint mathematical modelling and experimental approach to assess the potential impact of sTie2 on the ability of Angpt1 to signal. We find that the concentrations of sTie2 relative to Angpt1 required to suppress signalling by the ligand are more than ten-fold higher than those ever seen in normal or disease conditions. In contrast to the endogenous sTie2, an engineered form of sTie2, which presents dimeric ligand binding sites, inhibits Angpt1 signalling at seventy-fold lower concentrations. While loss of Tie2 ectodomain can suppress Angpt1 signalling locally in the cells in which the receptor is lost, our study shows that the resulting increase in circulating sTie2 is unlikely to affect Angpt1 activity elsewhere in the body.

4.
Cell Signal ; 26(7): 1379-84, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24642125

RESUMEN

Angiopoietin-1 (Ang1) is a ligand for the receptor tyrosine kinase Tie2 and has key roles in the development of the vascular system and vascular protection. In a screen to define signalling pathways regulated by Ang1 in endothelial cells we found the RNA-binding protein hnRNP-K to be phosphorylated in response to Ang1. The ligand stimulated both tyrosine phosphorylation of hnRNP-K and recruitment of the tyrosine kinase Src to the RNA-binding protein. In endothelial cells hnRNP-K was found bound to mRNA encoding the mitochondrial protein uncoupling protein-2 (UCP2). Ang1 stimulation of cells resulted in the release of UCP2 mRNA from hnRNP-K. Using in vitro assays we confirmed direct binding between hnRNP-K and UCP2 mRNA. Furthermore Src induced phosphorylation of purified hnRNP-K and prevented UCP2 mRNA binding. Tyrosine 458 in the RNA-binding protein was found to be required for suppression of UCP2 mRNA binding by Src phosphorylation. In addition to releasing UCP2 mRNA from hnRNP-K, Ang1 induced an increase in UCP2 protein expression in endothelial cells without affecting total UCP2 mRNA levels. Consistent with the known effects of UCP2 to suppress generation of reactive oxygen species, Ang1 limited ROS production in endothelium stimulated with tumour necrosis factor-α. Taken together these data suggest that UCP2 mRNA is present in endothelial cells bound to hnRNP-K, which holds it in a translationally inactive state, and that Ang1 stimulates Src interaction with hnRNP-K, phosphorylation of the RNA-binding protein, release of these transcripts and upregulation of UCP2 protein expression. This study demonstrates a new mechanism for post-transcriptional regulation of UCP2 by the vascular protective ligand Ang1. The ability to rapidly upregulate UCP2 protein expression may be important in protecting endothelial cells from excessive generation of potentially damaging reactive oxygen species.


Asunto(s)
Angiopoyetina 1/metabolismo , Endotelio Vascular/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Línea Celular , Clonación Molecular , Regulación de la Expresión Génica , Humanos , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/genética , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteína Desacopladora 2 , Familia-src Quinasas/metabolismo
5.
J Biol Chem ; 288(46): 33205-12, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24106271

RESUMEN

Tie2 is a receptor tyrosine kinase that is essential for the development and maintenance of blood vessels through binding the soluble ligands angiopoietin 1 (Ang1) and 2 (Ang2). Ang1 is constitutively produced by perivascular cells and is protective of the adult vasculature. Ang2 plays an important role in blood vessel formation and is normally expressed during development. However, its re-expression in disease states, including cancer and sepsis, results in destabilization of blood vessels contributing to the pathology of these conditions. Ang2 is thus an attractive therapeutic target. Here we report the directed evolution of a ligand trap for Ang2 by harnessing the B cell somatic hypermutation machinery and coupling this to selectable cell surface display of a Tie2 ectodomain. Directed evolution produced an unexpected combination of mutations resulting in loss of Ang1 binding but maintenance of Ang2 binding. A soluble form of the evolved ectodomain binds Ang2 but not Ang1. Furthermore, the soluble evolved ectodomain blocks Ang2 effects on endothelial cells without interfering with Ang1 activity. Our study has created a novel Ang2 ligand trap and provided proof of concept for combining surface display and exogenous gene diversification in B cells for evolution of a non-immunoglobulin target.


Asunto(s)
Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Evolución Molecular Dirigida , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Células HEK293 , Humanos , Unión Proteica , Estructura Terciaria de Proteína
6.
PLoS One ; 7(1): e29319, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22235284

RESUMEN

Angiopoietin-1 (Ang1) signals via the receptor tyrosine kinase Tie2 which exists in complex with the related protein Tie1 at the endothelial cell surface. Tie1 undergoes regulated ectodomain cleavage in response to phorbol esters, vascular endothelial growth factor (VEGF) and tumour necrosis factor-α (TNFα). Recently phorbol esters and VEGF were found also to stimulate ectodomain cleavage of Tie2. Here we investigate for the first time the effects of factors activating ectodomain cleavage on both Tie1 and Tie2 within the same population of cells, and their impact on angiopoietin signalling. We find that phorbol ester and VEGF activated Tie1 cleavage within minutes followed by restoration to control levels by 24 h. However, several hours of PMA and VEGF treatment were needed to elicit a detectable decrease in cellular Tie2, with complete loss seen at 24 h of PMA treatment. TNFα stimulated Tie1 cleavage, and induced a sustained decrease in cellular Tie1 over 24 h whilst increasing cellular Tie2. These differential effects of agonists on Tie1 and Tie2 result in dynamic modulation of the cellular Tie2∶Tie1 ratio. To assess the impact of this on Ang1 signalling cells were stimulated with VEGF and TNFα for differing times and Ang1-induced Tie2 phosphorylation examined. Elevated Tie2∶Tie1, in response to acute VEGF treatment or chronic TNFα, was associated with increased Ang1-activated Tie2 in cells. These data demonstrate cellular levels of Tie1 and Tie2 are differentially regulated by pathophysiologically relevant agonists resulting in dynamic control of the cellular Tie2∶Tie1 balance and modulation of Ang1 signalling. These findings highlight the importance of regulation of signalling at the level of the receptor. Such control may be an important adaptation to allow modulation of cellular signalling responses in systems in which the activating ligand is normally present in excess or where the ligand provides a constitutive maintenance signal.


Asunto(s)
Angiopoyetina 1/metabolismo , Receptor TIE-1/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Cinética , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína , Proteolisis/efectos de los fármacos , Receptor TIE-1/química , Receptor TIE-2/química
7.
Biochem Soc Trans ; 39(6): 1592-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22103492

RESUMEN

The angiopoietins act through the endothelial receptor tyrosine kinase Tie2 to regulate vessel maturation in angiogenesis and control quiescence and stability of established vessels. The activating ligand, Ang1 (angiopoietin-1), is constitutively expressed by perivascular cells, and the ability of endothelial cells to respond to the ligand is controlled at the level of the Ang1 receptor. This receptor interacts with the related protein Tie1 on the cell surface, and Tie1 inhibits Ang1 signalling through Tie2. The responsiveness of endothelium to Ang1 is determined by the relative levels of Tie2 and the inhibitory co-receptor Tie1 in the cells. Tie1 undergoes regulated ectodomain cleavage which is stimulated by a range of factors including VEGF (vascular endothelial growth factor), inflammatory cytokines and changes in shear stress. Ectodomain cleavage of Tie1 relieves inhibition of Tie2 and enhances Ang1 signalling. This mechanism regulates Ang1 signalling without requiring changes in the level of the ligand and allows Ang1 signalling to be co-ordinated with other signals in the cellular environment. Regulation of signalling at the level of receptor responsiveness may be an important adaptation in systems in which an activating ligand is normally present in excess or where the ligand provides a constitutive maintenance signal.


Asunto(s)
Angiopoyetinas/metabolismo , Transducción de Señal , Animales , Humanos , Receptores TIE/metabolismo
8.
Eur J Cell Biol ; 89(9): 661-73, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20605055

RESUMEN

Talin binds to and activates integrins and is thought to couple them to cytoskeletal actin. However, functional studies on talin have been restricted by the fact that most cells express two talin isoforms. Here we show that human umbilical vein endothelial cells (HUVEC) express only talin1, and that talin1 knockdown inhibited focal adhesion (FA) assembly preventing the cells from maintaining a spread morphology, a phenotype that was rescued by GFP-mouse talin1. Thus HUVEC offer an ideal model system in which to conduct talin structure/function studies. Talin contains an N-terminal FERM domain (comprised of F1, F2 and F3 domains) and a C-terminal flexible rod. The F3 FERM domain binds beta-integrin tails, and mutations in F3 that inhibited integrin binding (W359A) or activation (L325R) severely compromised the ability of GFP-talin1 to rescue the talin1 knockdown phenotype despite the presence of a second integrin-binding site in the talin rod. The talin rod contains several actin-binding sites (ABS), and mutations in the C-terminal ABS that reduced actin-binding impaired talin1 function, whereas those that increased binding resulted in more stable FAs. The results show that both the N-terminal integrin and C-terminal actin-binding functions of talin are essential to cell spreading and FA assembly. Finally, mutations that relieve talin auto-inhibition resulted in the rapid and excessive production of FA, highlighting the importance of talin regulation within the cell.


Asunto(s)
Células Endoteliales/metabolismo , Integrinas/metabolismo , Talina/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Adhesiones Focales/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Integrinas/química , Integrinas/genética , Ratones , Fenotipo , Talina/química , Talina/genética , Transfección , Venas Umbilicales/citología , Regulación hacia Arriba
9.
Microvasc Res ; 79(2): 121-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20079751

RESUMEN

Pre-diabetes is characterized by hyperglycemia and dyslipidemia; it is associated with increased cardiovascular disease and endothelial dysfunction. Angiopoietin-1 (Ang1), a ligand for endothelial receptor, is a potent vascular protective factor important in maintaining normal endothelial function. The aim of the study was to examine the influence of elevated glucose and fatty acid concentrations on angiopoietin signaling in human cardiac microvascular endothelial cells. Incubation with 30 mM glucose caused 50% suppression in the ability of Ang1 to activate Tie2-receptor phosphorylation without any decrease in Tie2 expression or increased internalization in microvascular endothelial cells. Examination of downstream signaling revealed inhibition of Ang1-dependent Akt phosphorylation. By contrast, Ang1 activation of Erk1/2 signaling was not affected by hyperglycemia. Similar suppression of Ang1-dependent activation of Akt by hyperglycemia was observed in large vessel human endothelial cells. Incubation of microvascular endothelial cells with 200 microM palmitic acid significantly inhibited Ang1-dependent Akt phosphorylation without affecting phosphorylation of the Tie-2 receptor or of ERK1/2. Therefore, contrary to hyperglycemia, palmitate acted exclusively downstream of the receptor. The present findings suggest a mechanism by which increased glucose or fatty acids may suppress vascular protection by Ang1 and predispose to endothelial dysfunction and vascular disease.


Asunto(s)
Angiopoyetina 1/farmacología , Endotelio Vascular/efectos de los fármacos , Glucosa/farmacología , Palmitatos/farmacología , Células Cultivadas , Vasos Coronarios/citología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor TIE-1/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal/efectos de los fármacos , Venas Umbilicales/citología
10.
Cell Signal ; 22(3): 527-32, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19922791

RESUMEN

Angiopoietin-1 (Ang1) and Ang2 are ligands for the receptor tyrosine kinase Tie2. Structural data suggest that the two ligands bind Tie2 similarly. However, in endothelial cells Ang1 activates Tie2 whereas Ang2 can act as an apparent antagonist. In addition, each ligand exhibits distinct kinetics of release following binding. These observations suggest that additional factors influence function and binding of angiopoietins with receptors in the cellular context. Previous work has shown that Ang1 binding and activation of Tie2 are inhibited by Tie1, a related receptor that complexes with Tie2 in cells. In this study we have investigated binding of Ang1 and Ang2 to Tie2 in endothelial cells. In contrast to Ang1, binding of Ang2 to Tie2 was found to be not affected by Tie1. Neither PMA-induced Tie1 ectodomain cleavage nor suppression of Tie1 expression by siRNA affected the ability of Ang2 to bind Tie2. Analysis of the level of Tie1 co-immunoprecipitating with angiopoietin-bound Tie2 demonstrated that Ang2 can bind Tie2 in Tie2:Tie1 complexes whereas Ang1 preferentially binds non-complexed Tie2. Stimulation of Tie1 ectodomain cleavage did not increase the agonist activity of Ang2 for Tie2. Similarly, the Tie2-agonist activity of Ang2 was not affected by siRNA suppression of Tie1 expression. Consistent with previous reports, loss of Tie1 ectodomain enhanced the agonist activity of Ang1 for Tie2. Importantly, Ang2 was still able to antagonize the elevated Ang1-activation of Tie2 that occurs on Tie1 ectodomain loss. Together these data demonstrate that Ang1 and Ang2 bind differently to Tie2 at the cell surface and this is controlled by Tie1. This differential regulation of angiopoietin binding allows control of Tie2 activation response to Ang1 without affecting Ang2 agonist activity and maintains the ability of Ang2 to antagonize even the enhanced Ang1 activation of Tie2 that occurs on loss of Tie1 ectodomain. This provides a mechanism by which signalling through Tie2 can be modified by stimuli in the cellular microenvironment.


Asunto(s)
Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Células Endoteliales/metabolismo , Receptor TIE-2/metabolismo , Línea Celular , Inhibidores Enzimáticos/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Inmunoprecipitación , ARN Interferente Pequeño/metabolismo , Receptor TIE-1/genética , Receptor TIE-1/metabolismo
11.
Biochem Soc Trans ; 37(Pt 4): 717-21, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19614582

RESUMEN

Growth factors provide key instructive cues for tissue formation and repair. However, many natural growth factors are limited in their usefulness for tissue engineering and regenerative applications by their poor retention at desired sites of action, short half-lives in vivo, pleiotropic actions and other features. In the present article, we review approaches to rational design of synthetic growth factors based on mechanisms of receptor activation. Such synthetic molecules can function as simplified ligands with potentially tunable specificity and action. Rational and combinatorial protein engineering techniques allow introduction of additional features into these synthetic growth molecules, as well as natural growth factors, which significantly enhance their therapeutic utility.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ingeniería de Proteínas/métodos , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Animales , Humanos , Péptidos y Proteínas de Señalización Intercelular/síntesis química , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Modelos Teóricos
12.
Cell Signal ; 21(8): 1346-50, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19376222

RESUMEN

The ability of cells to respond appropriately to changes in their environment requires integration and cross-talk between relevant signalling pathways. The vascular endothelial growth factor (VEGF) and angiopoietin families of ligands are key regulators of blood vessel formation. VEGF binds to receptor tyrosine kinases of the VEGF-receptor family to activate signalling pathways leading to endothelial migration, proliferation and survival whereas the angiopoietins interact with the Tie receptor tyrosine kinases to control vessel stability, survival and maturation. Here we show that VEGF can also activate the angiopoietin receptor Tie2. Activation of human endothelial cells with VEGF caused a four-fold stimulation of tyrosine phosphorylation of Tie2. This stimulation was not due to VEGF-induction of Tie2 ligands as soluble ligand binding domain of Tie2 failed to inhibit VEGF activation of the receptor. Immunoprecipitation analysis demonstrated no physical interaction between VEGF receptors and Tie2. However Tie2 does interact with the related receptor tyrosine kinase Tie1 and this receptor was found to be essential for VEGF activation of Tie2. VEGF stimulated proteolytic cleavage of Tie1 generating a truncated Tie1 intracellular domain. Similarly, phorbol ester also both stimulated Tie1 truncation and activated Tie2 phosphorylation. Inhibition of Tie1 cleavage with the metalloprotease inhibitor TAPI-2 suppressed VEGF- and phorbol ester-induced phosphorylation of Tie2. Truncated Tie1 formed in response to VEGF was also found to be tyrosine phosphorylated and this was independent of Tie2, though Tie2 could enhance Tie1 intracellular domain phosphorylation. Together these data demonstrate that VEGF activates Tie2 via a mechanism involving proteolytic cleavage of the associated tyrosine kinase Tie1 leading to trans-phosphorylation of Tie2. This novel mechanism of receptor tyrosine kinase activation is likely to be important in integrating signalling between two of the key receptor groups regulating angiogenesis.


Asunto(s)
Receptores TIE/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Células Cultivadas , Humanos , Ácidos Hidroxámicos/farmacología , Fosforilación , Receptor TIE-2/metabolismo , Transducción de Señal
13.
Microvasc Res ; 77(2): 187-91, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18848573

RESUMEN

Angiopoietin-1 (Ang1) has key roles in development and maintenance of the vascular system. The ligand is a potent inhibitor of vascular leakage and suppresses endothelial apoptosis and vessel regression. Ang1 was originally identified as a ligand for the receptor tyrosine kinase Tie2. Recently however Ang1 has also been found to activate the related tyrosine kinase Tie1. The contribution of Tie1 to mediating the effects of Ang1 on endothelial function is not known. In this study we used an siRNA approach to investigate the relative importance of Tie1 and Tie2 in transducing the effects of Ang1 on monolayer permeability and induction of apoptosis in human endothelial cells. siRNA directed against either Tie1 or Tie2 suppressed expression of each respective receptor by more than 90%. Ang1 inhibited endothelial monolayer permeability and this effect was prevented by suppression of Tie2 expression. In contrast, Ang1 inhibition of permeability was not affected by suppression of Tie1 expression. The ability of Ang1 to inhibit induction of apoptosis in response to serum deprivation was completely blocked by suppression of Tie2 expression, but not diminished by suppression of Tie1 expression. Taken together these data demonstrate that Tie2 mediates the inhibitory effects of Ang1 on endothelial permeability and apoptosis. The data also demonstrates that Tie1 does not transduce anti-apoptotic or anti-permeability effects of Ang1 in endothelial cells.


Asunto(s)
Angiopoyetina 1/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Receptor TIE-1/metabolismo , Receptor TIE-2/metabolismo , Angiopoyetina 1/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Secuencia de Bases , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Receptor TIE-1/antagonistas & inhibidores , Receptor TIE-1/genética , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/genética , Proteínas Recombinantes/farmacología
14.
Curr Neurovasc Res ; 5(4): 236-45, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18991658

RESUMEN

Vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang1) were originally identified as endothelial-specific ligands regulating key functions of the vasculature important in stroke. There is increasing evidence that these ligands also exert effects on neurons. Here we review the neuronal effects of VEGF and Ang1 and highlight their potential for therapeutic manipulation in stroke. VEGF stimulates angiogenesis whereas Ang1 suppresses leakage, inflammation and regression of microvessels. Expression of both ligands change dramatically in the brain in experimental stroke, correlating with increased vascular leakage and inflammation. In addition to vascular effects, VEGF can stimulate survival, migration and proliferation of neurons suggesting roles in neural protection and possible therapeutic applications, an idea supported by preclinical studies. Recent reports now demonstrate that Ang1 can also act directly on neurons and enhance neural repair. The realization that VEGF and Ang1 have effects on both neural and vascular compartments impacted by stroke provides new opportunities for therapeutic manipulation to promote neuroprotection and extend the thrombolytic window, as well as stimulating neurogenesis and revascularization.


Asunto(s)
Angiopoyetina 1/metabolismo , Neovascularización Fisiológica , Regeneración Nerviosa/fisiología , Accidente Cerebrovascular/fisiopatología , Factores de Crecimiento Endotelial Vascular/metabolismo , Angiopoyetina 2/metabolismo , Angiopoyetinas/metabolismo , Animales , Encéfalo/irrigación sanguínea , Humanos
15.
Int J Oncol ; 31(4): 893-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17786322

RESUMEN

The receptor tyrosine kinase Tie1 is expressed primarily in vascular endothelial cells. The receptor has also been detected in epithelial tumours in breast, thyroid and gastric cancers and in tumour cell lines where it appears as a 45 kDa truncated receptor fragment. In this study, we show that in addition to truncated Tie1, breast and colon tumour cell lines express a full-length Tie1 holoreceptor. In contrast to the situation in endothelial cells, Tie1 truncation is not activated by phorbol esters and generation of truncated Tie1 does not occur via a metalloprotease-inhibitor sensitive mechanism. Examination of the phosphorylation status of Tie1 revealed both the holoreceptor and truncated receptor to be constitutively activated in MCF-7 cells. These data indicate that Tie1 expressed in epithelial tumour cell lines is present in holoreceptor and truncated forms, and in MCF-7 cells both forms are constitutively phosphorylated and competent to signal. Our findings suggest therefore that anti-angiogenic strategies targeting the angiopoietin/Tie system in tumour microvasculature could also have additional direct effects on the tumour epithelial cells within those tumours in which there is also extravascular expression of the Tie1 receptor tyrosine kinase.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias del Colon/enzimología , Endotelio Vascular/enzimología , Receptor TIE-1/metabolismo , Venas Umbilicales/enzimología , Neoplasias de la Mama/patología , Células Cultivadas , Neoplasias del Colon/patología , Endotelio Vascular/citología , Activación Enzimática , Humanos , Immunoblotting , Inmunoprecipitación , Fosforilación , Eliminación de Secuencia , Venas Umbilicales/citología
16.
J Biol Chem ; 282(42): 30509-17, 2007 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17728252

RESUMEN

Regulated ectodomain shedding followed by intramembrane proteolysis has recently been recognized as important in cell signaling and for degradation of several type I transmembrane proteins. The receptor-tyrosine kinase Tie1 is known to undergo ectodomain cleavage generating a membrane-tethered endodomain. Here we show Tie1 is a substrate for regulated intramembrane proteolysis. After Tie1 ectodomain cleavage the newly formed 45-kDa endodomain undergoes additional proteolytic processing mediated by gamma-secretase to generate an amino-terminal-truncated 42-kDa fragment that is subsequently degraded by proteasomal activity. This sequential processing occurs constitutively and is stimulated by phorbol ester and vascular endothelial growth factor. To assess the biological significance of regulated Tie1 processing, we analyzed its effects on angiopoietin signaling. Activation of ectodomain cleavage causes loss of phosphorylated Tie1 holoreceptor and generation of phosphorylated receptor fragments in the presence of cartilage oligomeric protein angiopoietin 1. A key function of gamma-secretase is in preventing accumulation of these phosphorylated fragments. We also find that regulated Tie1 processing modulates ligand responsiveness of the Tie-1-associated receptor Tie2. Activation of Tie1 ectodomain cleavage increases cartilage oligomeric protein angiopoietin 1 activation of Tie2. This correlates with increased ability of Tie2 to bind ligand after shedding of the Tie1 extracellular domain. A similar enhancement of ligand activation of Tie2 is seen when Tie1 expression is suppressed by RNA interference. Together these data indicate that Tie1, via its extracellular domain, limits the ability of ligand to bind and activate Tie2. Furthermore the data suggest that regulated processing of Tie1 may be an important mechanism for controlling signaling by Tie2.


Asunto(s)
Angiopoyetina 1/metabolismo , Células Endoteliales/enzimología , Procesamiento Proteico-Postraduccional/fisiología , Receptor TIE-1/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal/fisiología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Carcinógenos/farmacología , Células Cultivadas , Células Endoteliales/citología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Ligandos , Ésteres del Forbol/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología
17.
J Vasc Surg ; 45(1): 155-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17210401

RESUMEN

BACKGROUND: Instability and rupture of carotid atherosclerotic plaques leads to thromboemboli and ischemic symptoms. Angiogenesis occurs within atherosclerotic plaques, and plaque vulnerability and symptomatic carotid disease have been associated with increased numbers of microvessels. In addition to microvessel number, it is possible that the phenotypes of intraplaque vessels could influence plaque stability. To test this, the morphology and maturity of vessels within plaques from symptomatic and asymptomatic patients was determined. METHODS: Carotid plaques were collected after endarterectomy from a cohort of 13 asymptomatic patients and 30 symptomatic patients. Plaques were sectioned and immunostained for the presence of endothelial cells, vascular smooth muscle cells, macrophages, and vascular endothelial growth factor. Sections were assessed for microvessel morphology, maturity as judged by smooth muscle cell cover, and the presence of vascular endothelial growth factor and macrophages. RESULTS: Two types of vascular structure were observed within plaques, microvessels and dilated, highly irregular multilobular vessels. These irregular dysmorphic vessels were found almost exclusively in plaques from symptomatic patients. The dysmorphic vessels lacked smooth muscle cells and were highly immature. Plaques also contained vascular endothelial growth factor, and this was observed adjacent to the dysmorphic vessels. This growth factor was found colocalized with macrophages. CONCLUSIONS: Symptomatic carotid plaques contain abnormal, immature microvessels similar to those found in tumors and healing wounds. Such vessels could contribute to plaque instability by acting as sites of vascular leakage by inflammatory cell recruitment. The immature vessels within plaques may be therapeutic targets for promoting plaque stabilization.


Asunto(s)
Aterosclerosis/complicaciones , Isquemia Encefálica/etiología , Enfermedades de las Arterias Carótidas/complicaciones , Trombosis Intracraneal/complicaciones , Anciano , Anciano de 80 o más Años , Aterosclerosis/metabolismo , Aterosclerosis/cirugía , Isquemia Encefálica/prevención & control , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/cirugía , Progresión de la Enfermedad , Endarterectomía Carotidea , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Trombosis Intracraneal/prevención & control , Macrófagos/patología , Persona de Mediana Edad , Pronóstico , Factores de Riesgo , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Biochem Biophys Res Commun ; 346(1): 335-8, 2006 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16756945

RESUMEN

Tie2 is a receptor tyrosine kinase expressed predominantly in endothelial cells. A missense mutation in the intracellular domain of Tie2 resulting in an arginine to tryptophan substitution causes an inherited form of vascular dysmorphogenesis, venous malformation (VM). The signalling pathways activated by mutant Tie2 and responsible for formation and maintenance of the abnormal vessels in VM are not known. In this study, we have sought to define these pathways by identifying phosphoproteins interacting with mutant Tie2 expressed in endothelial cells. We find R849W Tie2 is constitutively active in endothelium and recruits and phosphorylates a 52 kDa protein. This protein is identified as p52 ShcA. We show endothelial cells expressing VM-mutant Tie2 are protected from cell death and expression of dominant-negative ShcA inhibits the anti-apoptotic activity of the mutant receptor. Suppression of this pro-survival signalling could be a therapeutic option for inducing regression of lesional vessels.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Transducción de Señal/genética , Venas/anomalías , Células Cultivadas , Endotelio Vascular/citología , Humanos , Mutación Missense , Fosforilación , Mutación Puntual , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
19.
Circ Res ; 98(8): 1014-23, 2006 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-16645151

RESUMEN

Angiopoietin-1 (Ang1) has powerful vascular protective effects: suppressing plasma leakage, inhibiting vascular inflammation, and preventing endothelial death. Preclinical studies indicate that Ang1 may be therapeutically useful in a number of situations, including treatment of edema, endotoxemia, and transplant arteriosclerosis. However, the ligand has also been implicated in vessel remodeling, induction of angiogenesis and pulmonary hypertension, indicating that strategies to minimize any deleterious effects while optimizing vessel protection are likely to be needed. This review surveys the published data on vascular protective effects of Ang1 and highlights the therapeutic potential of this ligand, as well as possible limitations to its use. We also consider the data on Ang1 receptors and speculate on how to maximize therapeutic benefit by targeting the Tie receptors.


Asunto(s)
Angiopoyetina 1/fisiología , Cardiotónicos/uso terapéutico , Angiopoyetina 1/farmacología , Angiopoyetina 1/uso terapéutico , Animales , Arteriosclerosis/terapia , Vasos Sanguíneos/citología , Vasos Sanguíneos/fisiología , Permeabilidad Capilar , Supervivencia Celular , Modelos Animales de Enfermedad , Humanos , Transducción de Señal
20.
Biochem Biophys Res Commun ; 336(2): 392-6, 2005 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-16129411

RESUMEN

The factors controlling recruitment of endogenous and transplanted endothelial progenitor cells (EPC) to areas of neovascularization are largely unknown. In this study, we have examined the possibility that EPC migration and adhesion could be regulated by angiopoietin-2 (Ang2), a soluble ligand expressed by endothelial cells at sites of vessel remodelling and angiogenesis. We show for the first time that Ang2 causes a marked stimulation of EPC migration. This was specific for EPC as the ligand failed to affect endothelial cell migration. Ang2-stimulated EPC migration was inhibited by soluble Tie2 ectodomain. Furthermore, the ligand stimulated adhesion between EPC and endothelial monolayers.


Asunto(s)
Angiopoyetina 2/farmacología , Adhesión Celular/efectos de los fármacos , Comunicación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/fisiología , Endotelio Vascular/metabolismo , Células Madre/fisiología , Adhesión Celular/fisiología , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Células Madre/citología , Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...