Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(4)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36835432

RESUMEN

The endothelial cells (EC) of established blood vessels in adults remain extraordinarily quiescent in the sense that they are not actively proliferating, but they fulfill the necessary role to control the permeability of their monolayer that lines the interior of blood vessels. The cell-cell junctions between ECs in the endothelium comprise tight junctions and adherens homotypic junctions, which are ubiquitous along the vascular tree. Adherens junctions are adhesive intercellular contacts that are crucial for the organization of the EC monolayer and its maintenance and regulation of normal microvascular function. The molecular components and underlying signaling pathways that control the association of adherens junctions have been described in the last few years. In contrast, the role that dysfunction of these adherens junctions has in contributing to human vascular disease remains an important open issue. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid mediator found at high concentrations in blood which has important roles in the control of the vascular permeability, cell recruitment, and clotting that follow inflammatory processes. This role of S1P is achieved through a signaling pathway mediated through a family of G protein-coupled receptors designated as S1PR1. This review highlights novel evidence for a direct linkage between S1PR1 signaling and the mediation of EC cohesive properties that are controlled by VE-cadherin.


Asunto(s)
Cadherinas , Células Endoteliales , Endotelio Vascular , Receptores de Esfingosina-1-Fosfato , Humanos , Uniones Adherentes/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/fisiología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo
2.
Int J Mol Sci ; 24(15)2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-37569899

RESUMEN

Long COVID-19 syndrome appears after Severe Acute Respiratory Syndrome-Corona Virus (SARS-CoV-2) infection with acute damage to microcapillaries, microthrombi, and endothelialitis. However, the mechanisms involved in these processes remain to be elucidated. All blood vessels are lined with a monolayer of endothelial cells called vascular endothelium, which provides a the major function is to prevent coagulation. A component of endothelial cell junctions is VE-cadherin, which is responsible for maintaining the integrity of the vessels through homophilic interactions of its Ca++-dependent adhesive extracellular domain. Here we provide the first evidence that VE-cadherin is a target in vitro for ACE2 cleavage because its extracellular domain (hrVE-ED) contains two amino acid sequences for ACE2 substrate recognition at the positions 256P-F257 and 321PMKP-325L. Indeed, incubation of hrVE-ED with the active ectopeptidase hrACE2 for 16 hrs in the presence of 10 µM ZnCl2 showed a dose-dependent (from 0.2 ng/µL to 2 ng/µL) decrease of the VE-cadherin immunoreactive band. In vivo, in the blood from patients having severe COVID-19 we detected a circulating form of ACE2 with an apparent molecular mass of 70 kDa, which was barely detectable in patients with mild COVID-19. Of importance, in the patients with severe COVID-19 disease, the presence of three soluble fragments of VE-cadherin (70, 62, 54 kDa) were detected using the antiEC1 antibody while only the 54 kDa fragment was present in patients with mild disease. Altogether, these data clearly support a role for ACE2 to cleave VE-cadherin, which leads to potential biomarkers of SARS-CoV-2 infection related with the vascular disease in "Long COVID-19".


Asunto(s)
COVID-19 , Células Endoteliales , Humanos , Células Endoteliales/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , Síndrome Post Agudo de COVID-19 , SARS-CoV-2/metabolismo , Cadherinas/metabolismo , Endotelio Vascular/metabolismo
3.
Int J Mol Sci ; 23(13)2022 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-35806017

RESUMEN

Intermittent hypoxia (IH), the major feature of obstructive sleep apnea syndrome (OSAS), induces atherosclerosis and elastic fiber alterations. VE-cadherin cleavage is increased in OSAS patients and in an IH-cellular model. It is mediated by HIF-1 and Src-tyr-kinases pathways and results in endothelial hyperpermeability. Our aim was to determine whether blocking VE-cadherin cleavage in vivo could be an efficient strategy to inhibit deleterious IH-induced vascular remodeling, elastic fiber defects and atherogenesis. VE-cadherin regulation, aortic remodeling and atherosclerosis were studied in IH-exposed C57Bl/6J or ApoE-/-mice treated or not with Src-tyr-kinases inhibitors (Saracatinib/Pazopanib) or a HIF-1 inhibitor (Acriflavine). Human aortic endothelial cells were exposed to IH and treated with the same inhibitors. LDL and the monocytes transendothelium passage were measured. In vitro, IH increased transendothelium LDL and monocytes passage, and the tested inhibitors prevented these effects. In mice, IH decreased VE-cadherin expression and increased plasmatic sVE level, intima-media thickness, elastic fiber alterations and atherosclerosis, while the inhibitors prevented these in vivo effects. In vivo inhibition of HIF-1 and Src tyr kinase pathways were associated with the prevention of IH-induced elastic fiber/lamella degradation and atherogenesis, which suggests that VE-cadherin could be an important target to limit atherogenesis and progression of arterial stiffness in OSAS.


Asunto(s)
Aterosclerosis , Apnea Obstructiva del Sueño , Animales , Antígenos CD , Aorta/metabolismo , Aterosclerosis/metabolismo , Cadherinas , Grosor Intima-Media Carotídeo , Tejido Elástico/metabolismo , Células Endoteliales/metabolismo , Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Apnea Obstructiva del Sueño/metabolismo
4.
Eur Respir J ; 58(4)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33737411

RESUMEN

BACKGROUND: Obstructive sleep apnoea (OSA) causes intermittent hypoxia that in turn induces endothelial dysfunction and atherosclerosis progression. We hypothesised that VE-cadherin cleavage, detected by its released extracellular fragment solubilised in the blood (sVE), may be an early indicator of emergent abnormal endothelial permeability. Our aim was to assess VE-cadherin cleavage in OSA patients and in in vivo and in vitro intermittent hypoxia models to decipher the cellular mechanisms and consequences. METHODS: Sera from seven healthy volunteers exposed to 14 nights of intermittent hypoxia, 43 OSA patients and 31 healthy control subjects were analysed for their sVE content. Human aortic endothelial cells (HAECs) were exposed to 6 h of intermittent hypoxia in vitro, with or without an antioxidant or inhibitors of hypoxia-inducible factor (HIF)-1, tyrosine kinases or vascular endothelial growth factor (VEGF) pathways. VE-cadherin cleavage and phosphorylation were evaluated, and endothelial permeability was assessed by measuring transendothelial electrical resistance (TEER) and fluorescein isothiocyanate (FITC)-dextran flux. RESULTS: sVE was significantly elevated in sera from healthy volunteers submitted to intermittent hypoxia and OSA patients before treatment, but conversely decreased in OSA patients after 6 months of continuous positive airway pressure treatment. OSA was the main factor accounting for sVE variations in a multivariate analysis. In in vitro experiments, cleavage and expression of VE-cadherin increased upon HAEC exposure to intermittent hypoxia. TEER decreased and FITC-dextran flux increased. These effects were reversed by all of the pharmacological inhibitors tested. CONCLUSIONS: We suggest that in OSA, intermittent hypoxia increases endothelial permeability in OSA by inducing VE-cadherin cleavage through reactive oxygen species production, and activation of HIF-1, VEGF and tyrosine kinase pathways.


Asunto(s)
Células Endoteliales , Apnea Obstructiva del Sueño , Antígenos CD , Cadherinas/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Humanos , Hipoxia , Permeabilidad , Factor A de Crecimiento Endotelial Vascular
5.
Parasite Immunol ; 42(10): e12771, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32602946

RESUMEN

AIMS: We evaluate whether the serum and aqueous humour (AH) level of IgG anti-Hsp70.1 antibodies improved the biological diagnosis of ocular toxoplasmosis. METHODS AND RESULTS: In this prospective cross-sectional and multicentre study, serum and AH were collected at the time of active uveitis. Anti-Hsp70.1-antibody levels were determined by ELISA. Patients with confirmed (Group A1, n = 21) or suspected ocular toxoplasmosis (group A2, n = 30) were enrolled, as well as a control group of patients with cataract (group B, n = 42). Serum IgG anti-Hsp70.1 antibody levels were not significantly different within the group of uveitis patients (A1, n = 21 vs A2, n = 30, P = .8) and were significantly associated with the affected retinal zone (P = .006) and with the size of the retinal lesion (P = .03). Serum anti-Hsp70.1 antibody level was positive in 10 out of the 18 patients of group A2. Significant anti-Hsp-70.1 antibody level in AH was reported in only three patients (3 eyes) with confirmed ocular toxoplasmosis. CONCLUSION: While the level of IgG anti-Hsp-70.1 antibody in AH did not improve the laboratory diagnosis of ocular toxoplasmosis, its level in serum was of major significance for retinal damage diagnosis.


Asunto(s)
Anticuerpos Antiprotozoarios/análisis , Humor Acuoso/inmunología , Proteínas HSP70 de Choque Térmico/inmunología , Inmunoglobulina G/análisis , Toxoplasmosis Ocular/inmunología , Adulto , Anticuerpos Antiprotozoarios/inmunología , Estudios Transversales , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Toxoplasma/inmunología , Toxoplasmosis Ocular/diagnóstico , Uveítis/diagnóstico , Uveítis/inmunología
6.
Br J Cancer ; 118(9): 1179-1188, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29563634

RESUMEN

BACKGROUND: Vascular endothelial (VE)-cadherin is an endothelial cell-specific protein responsible for endothelium integrity. Its adhesive properties are regulated by post-translational processing, such as tyrosine phosphorylation at site Y685 in its cytoplasmic domain, and cleavage of its extracellular domain (sVE). In hormone-refractory metastatic breast cancer, we recently demonstrated that sVE levels correlate to poor survival. In the present study, we determine whether kidney cancer therapies had an effect on VE-cadherin structural modifications and their clinical interest to monitor patient outcome. METHODS: The effects of kidney cancer biotherapies were tested on an endothelial monolayer model mimicking the endothelium lining blood vessels and on a homotypic and heterotypic 3D cell model mimicking tumour growth. sVE was quantified by ELISA in renal cell carcinoma patients initiating sunitinib (48 patients) or bevacizumab (83 patients) in the first-line metastatic setting (SUVEGIL and TORAVA trials). RESULTS: Human VE-cadherin is a direct target for sunitinib which inhibits its VEGF-induced phosphorylation and cleavage on endothelial monolayer and endothelial cell migration in the 3D model. The tumour cell environment modulates VE-cadherin functions through MMPs and VEGF. We demonstrate the presence of soluble VE-cadherin in the sera of mRCC patients (n = 131) which level at baseline, is higher than in a healthy donor group (n = 96). Analysis of sVE level after 4 weeks of treatment showed that a decrease in sVE level discriminates the responders vs. non-responders to sunitinib, but not bevacizumab. CONCLUSIONS: These data highlight the interest for the sVE bioassay in future follow-up of cancer patients treated with targeted therapies such as tyrosine-kinase inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores Farmacológicos , Cadherinas/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Endotelio Vascular/efectos de los fármacos , Neoplasias Renales/tratamiento farmacológico , Sunitinib/uso terapéutico , Biomarcadores Farmacológicos/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Células Cultivadas , Ensayos Clínicos como Asunto , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Terapia Molecular Dirigida/métodos , Metástasis de la Neoplasia , Estudios Retrospectivos , Resultado del Tratamiento
7.
Br J Cancer ; 116(3): 356-361, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28056463

RESUMEN

BACKGROUND: Patients with metastatic breast cancer (MBC) represent a heterogeneous group, with large differences in outcomes from individual patients. VE-cadherin, an endothelial-specific cadherin, was shown to promote tumour proliferation and angiogenesis. Soluble VE-cadherin has been recently associated to breast cancer progression. This study was designed to investigate the prognosis significance of soluble VE-cadherin in hormone-refractory MBC. METHODS: Between 2004 and 2007, 150 patients with a fully documented history of hormone-refractory MBC were included in the prospective SEMTOF study. Serum concentrations of VE-cadherin were measured at inclusion for 141 patients and 6 weeks after the beginning of chemotherapy, using a sandwich enzyme immunoassay. RESULTS: The presence of high levels of serum VE-cadherin was significantly correlated to a shorter progression-free (PFS) and overall survival (OS). In a multivariate analysis along with clinical and biologic prognostic parameters, high serum VE-cadherin level was an independent adverse prognostic variable for PFS (median PFS 9.7 (IC95: 8; 11.9) vs 5.8 (IC95: 4.1; 8) months P=0.0008) and OS (median OS 34 (IC95: 26.6; 47.1) vs 14.8 (IC95: 9.3; 21.4) months P=0.0007). Moreover, VE-cadherin decrease during chemotherapy was also associated with good prognosis. CONCLUSIONS: Serum VE-cadherin levels correlate to poorer survival in patients with hormone-refractory MBC. As sVE-cadherin reflects tumour angiogenesis, this could have therapeutic implications for antiangiogenic treatment.


Asunto(s)
Antígenos CD/sangre , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/diagnóstico , Cadherinas/sangre , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/sangre , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Pronóstico , Análisis de Supervivencia
9.
Int Arch Allergy Immunol ; 170(2): 108-14, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27475458

RESUMEN

BACKGROUND: The aetiology of angio-oedema (AE) is difficult to determine; however, it is essential in emergency situations when two major contexts may be present: mast cell-mediated AE and bradykinin-mediated AE. Different forms of AE are currently distinguished based on clinical criteria (spontaneous duration of the attack, presence of concomitant or late-appearing superficial urticaria, history of atopy, and others), but specific biomarkers could improve patient management. OBJECTIVE: In this prospective study, potential biomarkers have been identified, and their statistical characteristics were examined. METHODS: Samples were taken on day 0 (D0) and D7 for 3 patient groups (n = 11 each): bradykinin-mediated AE [peripheral site of attack, ear, nose, throat (ENT), and abdominal involvement], mast cell-mediated AE, and non-bradykinin-mediated abdominal pain. RESULTS: Assay of the potential biomarkers revealed no significant differences in C1 inhibitor and C4 levels. In contrast, D-dimer levels peaked during bradykinin-mediated AE attacks (median 2.2 mg/l at D0 vs. 0.52 mg/l at D7; p < 10-3) as well as during mast cell-mediated AE attacks (1.97 vs. 0.65 mg/l; p = 0.04) and were high in bradykinin-mediated AE compared to the control group (0.69 mg/l; p = 0.01). A threshold value of 0.62 mg/l was found to have a negative predictive value of 100% for bradykinin-mediated AE compared to other causes of abdominal pain (group 3). Circulating VE-cadherin levels were also increased during an attack (1,990 at D0 vs. 1,566 ng/ml at D7; p = 0.01), but could not distinguish between bradykinin-mediated and mast cell-mediated AE, like D-dimers. CONCLUSIONS: Exploration of changes in fibrinolysis-related markers (particularly D-dimers) is thus promising for the diagnosis of AE attacks in difficult-to-diagnose abdominal forms, although it was not able to differentiate between bradykinin and mast cell-mediated AE.


Asunto(s)
Angioedema/diagnóstico , Angioedema/etiología , Biomarcadores , Bradiquinina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/sangre , Cadherinas/sangre , Proteína Inhibidora del Complemento C1 , Proteínas del Sistema Complemento , Femenino , Productos de Degradación de Fibrina-Fibrinógeno , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Tiempo , Adulto Joven
10.
Vasc Med ; 20(6): 557-65, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26129735

RESUMEN

Vascular endothelial-cadherin is the most important transmembrane component of endothelial adherens junctions, exclusively expressed by endothelial cells in all types of vessels. Targeting either the extracellular domain or the cytoplasmic tail deleteriously affects the junctional strength and leads to vascular permeability. Recently, cytokine-induced phosphorylation of the vascular endothelial-cadherin cytoplasmic domain was reported to trigger cleavage of its extracellular domain, producing the soluble form of the protein - soluble vascular endothelial-cadherin. Hence, the presence of soluble vascular endothelial-cadherin or auto-antibodies to human vascular endothelial-cadherin in human serum could signalize the presence of vascular abnormalities. This systematic review covers many human studies reporting increased levels of soluble vascular endothelial-cadherin, as well as auto-antibodies to human vascular endothelial-cadherin, which could be promising biomarkers of endothelial dysfunction in a large panel of diseases.


Asunto(s)
Antígenos CD/sangre , Autoanticuerpos/sangre , Enfermedades Autoinmunes/sangre , Cadherinas/sangre , Endotelio Vascular/metabolismo , Enfermedades Vasculares/sangre , Secuencia de Aminoácidos , Animales , Antígenos CD/inmunología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Biomarcadores/sangre , Cadherinas/inmunología , Permeabilidad Capilar , Endotelio Vascular/inmunología , Endotelio Vascular/fisiopatología , Humanos , Datos de Secuencia Molecular , Valor Predictivo de las Pruebas , Pronóstico , Enfermedades Vasculares/diagnóstico , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/fisiopatología
11.
Am J Physiol Heart Circ Physiol ; 307(3): H448-54, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24858855

RESUMEN

We previously reported that vascular endothelial growth factor induced vascular endothelial (VE)-cadherin tyrosine phosphorylation at Y685 in a Src-dependent manner in vitro. Here, we studied the occurrence of Y685 phosphorylation in vivo in the female reproductive tract because it is a unique model of physiological vascular remodeling dependent on vascular endothelial growth factor. We first developed and characterized an anti-phospho-specific antibody against the site Y685 of VE-cadherin to monitor VE-cadherin phosphorylation along the four phases of mouse estrous cycle, termed proestrus, estrus, metestrus, and diestrus. A dynamic profile of tyrosine phosphorylated proteins was observed in both uterus and ovary throughout mouse estrous cycle, including kinase Src, which was found highly active at the estrus phase. The extent of tyrosine phosphorylated VE-cadherin was low at proestrus but strongly increased at estrus and returned to baseline at metestrus and diestrus, suggesting a potent hormonal regulation of this specific process. Indeed, C57Bl/6 female mice treatment with pregnant mare serum gonadotropin and human chorionic gonadotropin confirmed a significant increase in phosphoY685-VE-cadherin compared with that in untreated mice. These results demonstrate that VE-cadherin tyrosine phosphorylation at Y685 is a physiological and hormonally regulated process in female reproductive organs. In addition, this process was concomitant with the early steps of vascular remodeling taking place at estrus stage, suggesting that phosphoY685-VE-cadherin is a biomarker of endothelial cell activation in vivo.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Ciclo Estral/metabolismo , Ovario/irrigación sanguínea , Ovario/metabolismo , Útero/metabolismo , Animales , Gonadotropina Coriónica/farmacología , Ciclo Estral/efectos de los fármacos , Femenino , Gonadotropinas Equinas/farmacología , Ratones Endogámicos C57BL , Ovario/efectos de los fármacos , Fosforilación , Factores de Tiempo , Tirosina , Útero/efectos de los fármacos , Remodelación Vascular , Familia-src Quinasas/metabolismo
12.
Am J Physiol Heart Circ Physiol ; 307(3): H455-63, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24858856

RESUMEN

Covalent modifications such as tyrosine phosphorylation are associated with the breakdown of endothelial cell junctions and increased vascular permeability. We previously showed that vascular endothelial (VE)-cadherin was tyrosine phosphorylated in vivo in the mouse reproductive tract and that Y685 was a target site for Src in response to vascular endothelial growth factor in vitro. In the present study, we aimed to understand the implication of VE-cadherin phosphorylation at site Y685 in cyclic angiogenic organs. To achieve this aim, we generated a knock-in mouse carrying a tyrosine-to-phenylalanine point mutation of VE-cadherin Y685 (VE-Y685F). Although homozygous VE-Y685F mice were viable and fertile, the nulliparous knock-in female mice exhibited enlarged uteri with edema. This phenotype was observed in 30% of females between 4 to 14 mo old. Histological examination of longitudinal sections of the VE-Y685F uterus showed an extensive disorganization of myometrium and endometrium with highly edematous uterine glands, numerous areas with sparse cells, and increased accumulation of collagen fibers around blood vessels, indicating a fibrotic state. Analysis of cross section of ovaries showed the appearance of spontaneous cysts, which suggested increased vascular hyperpermeability. Electron microscopy analysis of capillaries in the ovary showed a slight but significant increase in the gap size between two adjacent endothelial cell membranes in the junctions of VE-Y685F mice (wild-type, 11.5 ± 0.3, n = 78; and VE-Y685F, 12.48 ± 0.3, n = 65; P = 0.045), as well as collagen fiber accumulation around capillaries. Miles assay revealed that either basal or vascular endothelial growth factor-stimulated permeability in the skin was increased in VE-Y685F mice. Since edema and fibrotic appearance have been identified as hallmarks of initial increased vascular permeability, we conclude that the site Y685 in VE-cadherin is involved in the physiological regulation of capillary permeability. Furthermore, this knock-in mouse model is of potential interest for further studies of diseases that are associated with abnormal vascular permeability.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Capilares/metabolismo , Permeabilidad Capilar , Técnicas de Sustitución del Gen , Neovascularización Fisiológica , Ovario/irrigación sanguínea , Útero/irrigación sanguínea , Animales , Antígenos CD/genética , Cadherinas/genética , Capilares/fisiopatología , Capilares/ultraestructura , Edema/metabolismo , Edema/patología , Edema/fisiopatología , Femenino , Genotipo , Homocigoto , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Fosforilación , Tirosina
13.
Arthritis Rheum ; 64(1): 77-87, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21905018

RESUMEN

OBJECTIVE: Rheumatoid arthritis (RA) is a chronic, systemic inflammatory disorder that principally attacks synovial joints. However, accelerated atherosclerosis and increased cardiovascular morbidity and mortality are major clinical consequences of endothelial dysfunction in RA patients. Tumor necrosis factor α (TNFα) is the major mediator of inflammation in RA, related to vascular injury by targeting VE-cadherin, an endothelium-specific adhesion molecule of vital importance for endothelium integrity and angiogenesis. We undertook this study to examine the mechanisms regulating VE-cadherin processing by TNFα and their occurrence in RA. METHODS: Human umbilical vein endothelial cells were used in primary culture and treated with recombinant TNFα to study VE-cadherin cleavage. Cell lysates and conditioned media were analyzed by Western blotting for VE-cadherin cytoplasmic domain and extracellular domain (VE-90) generation, respectively. VE-90 was analyzed at baseline and at the 1-year followup in sera from 63 RA patients (from the Very Early Rheumatoid Arthritis cohort) with disease duration of <6 months. RESULTS: TNFα induced a time-dependent shedding of VE-90 in cell media. This effect was prevented by tyrosine kinase inhibitors (genistein and PP2) or by knocking down Src kinase. In contrast, tyrosine phosphatase blockade enhanced VE-cadherin cleavage, confirming the requirement of tyrosine phosphorylation processes. In addition, using the matrix metalloproteinase (MMP) activator APMA and the MMP inhibitor GM6001, we demonstrated that MMPs are involved in TNFα-induced VE-cadherin cleavage. Of major importance, VE-90 was detected in sera from the 63 RA patients and was positively correlated with the Disease Activity Score at baseline and after 1-year followup. CONCLUSION: These findings provide the first evidence of VE-cadherin proteolysis upon TNFα stimulation and suggest potential clinical relevance of soluble VE-cadherin in management of RA.


Asunto(s)
Antígenos CD/metabolismo , Artritis Reumatoide/metabolismo , Cadherinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Adulto , Anciano , Artritis Reumatoide/patología , Artritis Reumatoide/fisiopatología , Células Cultivadas , Medios de Cultivo Condicionados/química , Femenino , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Proteínas Recombinantes , Factores de Tiempo , Transfección , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
14.
Anticancer Res ; 42(10): 4689-4700, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36191998

RESUMEN

BACKGROUND/AIM: A growing body of research is contributing to the development of three-dimensional (3D) tissue models to close the gap between two-dimensional (2D) cell culture and animal models. Here, we report fundamental studies to confirm the modification of vascular endothelial (VE)-cadherin by a tumor microenvironment using 2D and 3D in vitro models of triple-negative breast cancer cells co-cultured with endothelial cells. MATERIALS AND METHODS: Breast cancer cells were cultivated as a monolayer (2D) on plates for 5 days or as microtumor spheroids (3D) with endothelial cells for up to 6 days. Phosphotyrosine-containing protein panels were analyzed in both cell types and upon co-culture. Microtumor spheroid size was evaluated via phase contrast microscopy. The content of VE-cadherin and phospho-VE-cadherin was determined. The effect of microtumor spheroid on the capillary network formed by endothelial cells was quantified by ImageJ Angiogenesis Analyzer. Sunitinib was used to determine drug efficacy in this model. RESULTS: The activity of signaling pathways in endothelial cells, including phosphorylation of Y685-VE-cadherin, was increased by the presence of breast cancer cells. In the 3D co-culture system, we established a ratio of the two cell types which allowed viability for 6 days. As a proof-of-concept of the 3D co-culture system for the process of drug discovery and development, we used the system to quantify the efficacy of sunitinib on the phosphorylation of VE-cadherin. CONCLUSION: In summary, we established 2D and 3D breast cancer-endothelial cell test systems compatible for detection of minimally tyrosine-phosphorylated proteins including VE-cadherin. The systems are capable of quantifying the effect of drugs on a tissue model of angiogenesis. This is a step towards developing tools for drug-efficacy testing that do not rely on live animals.


Asunto(s)
Cadherinas , Células Endoteliales , Animales , Antígenos CD , Cadherinas/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Neovascularización Patológica/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Sunitinib/farmacología , Tirosina/metabolismo
16.
Sci Rep ; 10(1): 16040, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32994463

RESUMEN

Aqueous humor drainage is essential for the regulation of intraocular pressure (IOP), a major risk factor for glaucoma. The Schlemm's canal and the non-conventional uveoscleral pathway are known to drain aqueous humor from the eye anterior chamber. It has recently been reported that lymphatic vessels are involved in this process, and that the Schlemm's canal responds to some lymphatic regulators. We have previously shown a critical role for bone morphogenetic protein 9 (BMP9) in lymphatic vessel maturation and valve formation, with repercussions in drainage efficiency. Here, we imaged eye lymphatic vessels and analyzed the consequences of Bmp9 (Gdf2) gene invalidation. A network of lymphatic vessel hyaluronan receptor 1 (LYVE-1)-positive lymphatic vessels was observed in the corneolimbus and the conjunctiva. In contrast, LYVE-1-positive cells present in the ciliary bodies were belonging to the macrophage lineage. Although enlarged conjunctival lymphatic trunks and a reduced valve number were observed in Bmp9-KO mice, there were no morphological differences in the Schlemm's canal compared to wild type animals. Moreover, there were no functional consequences on IOP in both basal control conditions and after laser-induced ocular hypertonia. Thus, the BMP9-activated signaling pathway does not constitute a wise target for new glaucoma therapeutic strategies.


Asunto(s)
Factor 2 de Diferenciación de Crecimiento/metabolismo , Presión Intraocular/fisiología , Vasos Linfáticos/metabolismo , Animales , Cámara Anterior/fisiología , Humor Acuoso/metabolismo , Glaucoma/metabolismo , Linfangiogénesis/fisiología , Vasos Linfáticos/fisiología , Masculino , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Esclerótica/fisiología , Tonometría Ocular/métodos , Malla Trabecular/fisiología
18.
Biotechnol J ; 13(12): e1800463, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30457706

RESUMEN

A major problem for the detection of cancer biomarkers in plasma or serum is that common clinical practice does not require the patient to be in a fasting state. Considering that lipoproteins are the main population affected by food intake, the authors hypothesized that biomarkers could be embedded in lipid particles and thereby opens a new avenue for detection. Using the recently published biomarker, soluble VE-cadherin (sVE), the authors tested our hypothesis using techniques of biophysics, biochemistry and the tools of nanobiotechnology on serum samples from kidney cancer patients (n = 106). Optical density as well as contact angle measurements of serum revealed heterogeneity in the particle content of the serum samples. Isolation of the lipidic moieties by ultracentrifugation showed that sVE was detected in this compartment. Further, isolation of lipoprotein subclasses by precipitation with sodium phosphotungstate and MgCl2 , showed that HDL carried the majority of sVE. Immunoprecipitation of sVE confirmed that it was associated with Apolipoprotein A1, a major compound of HDL. Using a biomimetic lipid bilayer membrane coupled with impedance spectroscopy the authors quantified, in real-time, that the sVE adsorbed to the lipid bilayer membrane without altering its structure. Taken together, these results show for the first time a direct interaction of a cancer biomarker with lipids. The authors anticipate these results to prompt fasting for future blood tests for large-scale studies in the biomarkers research field.


Asunto(s)
Biomarcadores de Tumor/sangre , Materiales Biomiméticos , Colesterol/sangre , Antígenos CD/sangre , Apolipoproteína A-I/sangre , Biotecnología , Cadherinas/sangre , Células HEK293 , Humanos , Inmunoprecipitación , Neoplasias Renales/sangre , Lipoproteínas/sangre , Modelos Teóricos , Nanotecnología
19.
Circ Res ; 96(3): 384-91, 2005 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-15662029

RESUMEN

Vascular endothelial-cadherin (VE-cadherin) plays a key role in angiogenesis and in vascular permeability. The regulation of its biological activity may be a central mechanism in normal or pathological angiogenesis. VE-cadherin has been shown to be phosphorylated on tyrosine in vitro under various conditions, including stimulation by VEGF. In the present study, we addressed the question of the existence of a tyrosine phosphorylated form of VE-cadherin in vivo, in correlation with the quiescent versus angiogenic state of adult tissues. Phosphorylated VE-cadherin was detected in mouse lung, uterus, and ovary but not in other tissues unless mice were injected with peroxovanadate to block protein phosphatases. Remarkably, VE-cadherin tyrosine phosphorylation was dramatically increased in uterus and ovary, and not in other organs, during PMSG/hCG-induced angiogenesis. In parallel, we observed an increased association of VE-cadherin with Flk1 (VEGF receptor 2) during hormonal angiogenesis. Additionally, Src kinase was constitutively associated with VE-cadherin in both quiescent and angiogenic tissues and increased phosphorylation of VE-cadherin-associated Src was detected in uterus and ovary after hormonal treatment. Src-VE-cadherin association was detected in cultured endothelial cells, independent of VE-cadherin phosphorylation state and Src activation level. In this model, Src inhibition impaired VEGF-induced VE-cadherin phosphorylation, indicating that VE-cadherin phosphorylation was dependent on Src activation. We conclude that VE-cadherin is a substrate for tyrosine kinases in vivo and that its phosphorylation, together with that of associated Src, is increased by angiogenic stimulation. Physical association between Flk1, Src, and VE-cadherin may thus provide an efficient mechanism for amplification and perpetuation of VEGF-stimulated angiogenic processes.


Asunto(s)
Cadherinas/química , Cadherinas/metabolismo , Endotelio Vascular/química , Endotelio Vascular/metabolismo , Neovascularización Fisiológica/fisiología , Fosfotirosina/metabolismo , Animales , Extractos Celulares/química , Línea Celular , Células Endoteliales/química , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Femenino , Gonadotropinas Equinas/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Ovario/efectos de los fármacos , Ovario/metabolismo , Fosforilación/efectos de los fármacos , Proteínas/química , Proteínas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Venas Umbilicales/citología , Útero/efectos de los fármacos , Útero/metabolismo , Vanadatos/administración & dosificación , Vanadatos/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
Trends Cardiovasc Med ; 16(2): 55-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16473763

RESUMEN

Because angiogenesis is a key step in a number of pathologic processes, including tumor growth and atherosclerosis, many research studies have investigated the regulatory signals active at various stages of vascular invasion. The differential activities of the endothelial junction protein vascular endothelial (VE)-cadherin reflect the versatile behavior of endothelial cells between vascular quiescence and angiogenesis. VE-cadherin function and signaling are deeply modified in proliferating cells, and this conversion is accompanied by phosphorylation of the protein on tyrosine residues and enhanced transcription of its gene. Recent advances in the complex interplay between protein tyrosine kinases and phosphatases regulating VE-cadherin phosphorylation and function are discussed in this review.


Asunto(s)
Antígenos CD/fisiología , Aterosclerosis/patología , Cadherinas/fisiología , Endotelio Vascular/fisiología , Neovascularización Patológica/etiología , Antígenos CD/genética , Antígenos CD/metabolismo , Aterosclerosis/etiología , Cadherinas/genética , Cadherinas/metabolismo , Permeabilidad Capilar , Ciclo Celular/fisiología , Células Endoteliales/fisiología , Endotelio Vascular/patología , Regulación de la Expresión Génica/fisiología , Humanos , Neoplasias/patología , Fosforilación , Tirosina/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA