Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Basic Clin Pharmacol Toxicol ; 123 Suppl 5: 6-19, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29668117

RESUMEN

Vascular anomalies are localized defects of morphogenesis that can affect lymphatic and blood vessels. They are generally called birthmarks, typically observed soon after birth and occurring in up to 10% of children. Based on their clinical and histological characteristics, they are classified into vascular tumours and vascular malformations. The most common malformations are venous malformations (VMs) resulting in chronic vascular diseases that can be associated with significant morbidity necessitating often demanding and repeating clinical management. The current treatment is based on surgical resection and sclerotherapy, which can be impossible due to the size or location of lesions or ineffective due to the regrowth of malformed vessels. Therefore, medical therapies for VMs are highly desired. Recent studies have identified genetic defects that result in the constantly active endothelial cell receptor tyrosine kinase TIE2/phosphoinositide 3-kinase PI3K signalling pathway as a frequent cause for VMs. The first treatment to inhibit this pathway with sirolimus indicated that molecular treatment can be effective against VMs. In addition, certain VM 'hotspot' mutations have been previously found in tumours, providing the rationale for the exploration and repurposing of existing and investigational cancer drugs for VMs. Finally, discoveries of molecular and cellular abnormalities that characterize a large proportion of VMs and the generation of pre-clinical VM mouse models provide the necessary basis for the development of the targeted molecular treatment strategies we discuss in this MiniReview.


Asunto(s)
Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Enfermedades Vasculares/prevención & control , Malformaciones Vasculares/tratamiento farmacológico , Venas/anomalías , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Enfermedad Crónica/prevención & control , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Modelos Animales de Enfermedad , Humanos , Terapia Molecular Dirigida/métodos , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Enfermedades Vasculares/etiología , Malformaciones Vasculares/complicaciones , Malformaciones Vasculares/genética
2.
Clin Sci (Lond) ; 131(1): 87-103, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27941161

RESUMEN

Endothelial cells that form the inner layer of blood and lymphatic vessels are important regulators of vascular functions and centrally involved in the pathogenesis of vascular diseases. In addition to the vascular endothelial growth factor (VEGF) receptor pathway, the angiopoietin (Ang)-Tie system is a second endothelial cell specific ligand-receptor signalling system necessary for embryonic cardiovascular and lymphatic development. The Ang-Tie system also regulates postnatal angiogenesis, vessel remodelling, vascular permeability and inflammation to maintain vascular homoeostasis in adult physiology. This system is implicated in numerous diseases where the vasculature has an important contribution, such as cancer, sepsis, diabetes, atherosclerosis and ocular diseases. Furthermore, mutations in the TIE2 signalling pathway cause defects in vascular morphogenesis, resulting in venous malformations and primary congenital glaucoma. Here, we review recent advances in the understanding of the Ang-Tie signalling system, including cross-talk with the vascular endothelial protein tyrosine phosphatase (VE-PTP) and the integrin cell adhesion receptors, focusing on the Ang-Tie system in vascular development and pathogenesis of vascular diseases.


Asunto(s)
Angiopoyetinas/metabolismo , Sistema Cardiovascular/metabolismo , Sistema Linfático/metabolismo , Receptor TIE-1/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal , Angiopoyetinas/genética , Animales , Sistema Cardiovascular/enzimología , Sistema Cardiovascular/crecimiento & desarrollo , Humanos , Sistema Linfático/enzimología , Sistema Linfático/crecimiento & desarrollo , Receptor TIE-1/genética , Receptor TIE-2/genética
3.
J Invest Dermatol ; 137(1): 207-216, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27519652

RESUMEN

Blue rubber bleb nevus syndrome (Bean syndrome) is a rare, severe disorder of unknown cause, characterized by numerous cutaneous and internal venous malformations; gastrointestinal lesions are pathognomonic. We discovered somatic mutations in TEK, the gene encoding TIE2, in 15 of 17 individuals with blue rubber bleb nevus syndrome. Somatic mutations were also identified in five of six individuals with sporadically occurring multifocal venous malformations. In contrast to common unifocal venous malformation, which is most often caused by the somatic L914F TIE2 mutation, multifocal forms are predominantly caused by double (cis) mutations, that is, two somatic mutations on the same allele of the gene. Mutations are identical in all lesions from a given individual. T1105N-T1106P is recurrent in blue rubber bleb nevus, whereas Y897C-R915C is recurrent in sporadically occurring multifocal venous malformation: both cause ligand-independent activation of TIE2, and increase survival, invasion, and colony formation when expressed in human umbilical vein endothelial cells.


Asunto(s)
Neoplasias Gastrointestinales/genética , Predisposición Genética a la Enfermedad/epidemiología , Mutación , Nevo Azul/genética , Receptor TIE-2/genética , Neoplasias Cutáneas/genética , Malformaciones Vasculares/genética , Bélgica , Estudios de Cohortes , Femenino , Neoplasias Gastrointestinales/diagnóstico , Humanos , Incidencia , Masculino , Nevo Azul/diagnóstico , Enfermedades Raras , Neoplasias Cutáneas/diagnóstico , Malformaciones Vasculares/diagnóstico
4.
Am J Hum Genet ; 97(6): 914-21, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26637981

RESUMEN

Somatic mutations in TEK, the gene encoding endothelial cell tyrosine kinase receptor TIE2, cause more than half of sporadically occurring unifocal venous malformations (VMs). Here, we report that somatic mutations in PIK3CA, the gene encoding the catalytic p110α subunit of PI3K, cause 54% (27 out of 50) of VMs with no detected TEK mutation. The hotspot mutations c.1624G>A, c.1633G>A, and c.3140A>G (p.Glu542Lys, p.Glu545Lys, and p.His1047Arg), frequent in PIK3CA-associated cancers, overgrowth syndromes, and lymphatic malformation (LM), account for >92% of individuals who carry mutations. Like VM-causative mutations in TEK, the PIK3CA mutations cause chronic activation of AKT, dysregulation of certain important angiogenic factors, and abnormal endothelial cell morphology when expressed in human umbilical vein endothelial cells (HUVECs). The p110α-specific inhibitor BYL719 restores all abnormal phenotypes tested, in PIK3CA- as well as TEK-mutant HUVECs, demonstrating that they operate via the same pathogenic pathways. Nevertheless, significant genotype-phenotype correlations in lesion localization and histology are observed between individuals with mutations in PIK3CA versus TEK, pointing to gene-specific effects.


Asunto(s)
Mutación , Neovascularización Patológica/genética , Fosfatidilinositol 3-Quinasas/genética , Malformaciones Vasculares/genética , Alelos , Fosfatidilinositol 3-Quinasa Clase I , Regulación de la Expresión Génica , Frecuencia de los Genes , Estudios de Asociación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Transducción de Señal , Tiazoles/farmacología , Transfección , Malformaciones Vasculares/enzimología , Malformaciones Vasculares/patología , Venas/enzimología , Venas/patología
5.
Hum Mol Genet ; 24(22): 6374-89, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26319232

RESUMEN

Venous malformations (VMs) are localized defects in vascular morphogenesis frequently caused by mutations in the gene for the endothelial tyrosine kinase receptor TIE2. Here, we report the analysis of a comprehensive collection of 22 TIE2 mutations identified in patients with VM, either as single amino acid substitutions or as double-mutations on the same allele. Using endothelial cell (EC) cultures, mouse models and ultrastructural analysis of tissue biopsies from patients, we demonstrate common as well as mutation-specific cellular and molecular features, on the basis of which mutations cluster into categories that correlate with data from genetic studies. Comparisons of double-mutants with their constituent single-mutant forms identified the pathogenic contributions of individual changes, and their compound effects. We find that defective receptor trafficking and subcellular localization of different TIE2 mutant forms occur via a variety of mechanisms, resulting in attenuated response to ligand. We also demonstrate, for the first time, that TIE2 mutations cause chronic activation of the MAPK pathway resulting in loss of normal EC monolayer due to extracellular matrix (ECM) fibronectin deficiency and leading to upregulation of plasminogen/plasmin proteolytic pathway. Corresponding EC and ECM irregularities are observed in affected tissues from mouse models and patients. Importantly, an imbalance between plasminogen activators versus inhibitors would also account for high d-dimer levels, a major feature of unknown cause that distinguishes VMs from other vascular anomalies.


Asunto(s)
Receptor TIE-2/genética , Malformaciones Vasculares/genética , Sustitución de Aminoácidos , Animales , Movimiento Celular/genética , Células Endoteliales/metabolismo , Femenino , Productos de Degradación de Fibrina-Fibrinógeno , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ligandos , Ratones , Ratones SCID , Mutación , Fosforilación , Receptor TIE-2/metabolismo , Transducción de Señal , Esferoides Celulares , Malformaciones Vasculares/enzimología
6.
Hum Mol Genet ; 22(17): 3438-48, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23633549

RESUMEN

Mutations in the endothelial cell (EC) tyrosine kinase receptor TIE2 cause inherited and sporadic forms of venous malformation. The recurrent somatic mutation L914F and common germline mutation R849W differ in terms of phosphorylation level, as well as sub-cellular localization and trafficking of the receptor. Previous studies have shed light on certain pathogenic properties of R849W, but the mechanisms of action of L914F are unknown. We used global gene expression profiling to study the effects of L914F on ECs. We found that L914F strongly dysregulates genes involved in vascular development, cell migration and extracellular matrix processing, while R849W has weak effects. We also demonstrate, for the first time, that TIE2-mutant ECs are deficient in the production of PDGFB, both in vitro and ex vivo in patient tissues. This defect is mediated by the chronic, ligand-independent activation of AKT by the mutant receptors. Inadequate secretion of the major mural cell attractant likely plays an important role in the development of abnormal vascular channels, contributing to the characteristic paucity of surrounding vascular smooth muscle cells.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Malformaciones Vasculares/genética , Malformaciones Vasculares/metabolismo , Movimiento Celular/genética , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Mutación de Línea Germinal , Humanos , Músculo Liso Vascular/metabolismo , Fosforilación , Transducción de Señal/genética , Transducción de Señal/fisiología
7.
J Cell Sci ; 125(Pt 9): 2212-23, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22357955

RESUMEN

Angiopoietin 1 (Ang1) is an activating ligand for the endothelial receptor tyrosine kinase Tie2, whereas Ang2 acts as a context-dependent agonist or antagonist that has a destabilizing effect on the vasculature. The molecular mechanisms responsible for the versatile functions of Ang2 are poorly understood. We show here that Ang2, but not Ang1, induces Tie2 translocation to the specific cell-matrix contact sites located at the distal end of focal adhesions. The Ang2-specific Tie2 translocation was associated with distinct Tie2 activation and downstream signals which differed from those of Ang1, and led to impaired cell motility and weak cell-matrix adhesion. We demonstrate that the different oligomeric or multimeric forms of the angiopoietins induce distinct patterns of Tie2 trafficking; the lower oligomerization state of native Ang2 was crucial for the Ang2-specific Tie2 redistribution, whereas multimeric structures of Ang1 and Ang2 induced similar responses. The Ang2-specific Tie2 trafficking to cell-matrix contacts was also dependent on the cell substratum, α2ß1-integrin-containing cell-matrix adhesion sites and intact microtubules. Our data indicate that the different subcellular trafficking of Tie2-Ang2 and Tie2-Ang1 complexes generates ligand-specific responses in the angiopoietin-Tie signaling pathway, including modulation of cell-matrix interactions.


Asunto(s)
Angiopoyetina 1/química , Angiopoyetina 2/química , Endotelio Vascular/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Cuerpo Vítreo/irrigación sanguínea , Angiopoyetina 1/genética , Angiopoyetina 1/farmacología , Angiopoyetina 2/genética , Angiopoyetina 2/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Uniones Célula-Matriz/efectos de los fármacos , Uniones Célula-Matriz/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfa2beta1/genética , Integrina alfa2beta1/metabolismo , Inyecciones Intravítreas , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Células 3T3 NIH , Neovascularización Fisiológica , Multimerización de Proteína , Proteínas Tirosina Quinasas Receptoras/genética , Receptor TIE-2 , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...