Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Cell Mol Life Sci ; 81(1): 173, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38597967

ABSTRACT

Heterozygous mutations in any of three major genes, BRCA1, BRCA2 and PALB2, are associated with high-risk hereditary breast cancer susceptibility frequently seen as familial disease clustering. PALB2 is a key interaction partner and regulator of several vital cellular activities of BRCA1 and BRCA2, and is thus required for DNA damage repair and alleviation of replicative and oxidative stress. Little is however known about how PALB2-deficiency affects cell function beyond that, especially in the three-dimensional setting, and also about its role during early steps of malignancy development. To answer these questions, we have generated biologically relevant MCF10A mammary epithelial cell lines with mutations that are comparable to certain clinically important PALB2 defects. We show in a non-cancerous background how both mono- and biallelically PALB2-mutated cells exhibit gross spontaneous DNA damage and mitotic aberrations. Furthermore, PALB2-deficiency disturbs three-dimensional spheroid morphology, increases the migrational capacity and invasiveness of the cells, and broadly alters their transcriptome profiles. TGFß signaling and KRT14 expression are enhanced in PALB2-mutated cells and their inhibition and knock down, respectively, lead to partial restoration of cell functions. KRT14-positive cells are also more abundant with DNA damage than KRT14-negative cells. The obtained results indicate comprehensive cellular changes upon PALB2 mutations, even in the presence of half dosage of wild type PALB2 and demonstrate how PALB2 mutations may predispose their carriers to malignancy.


Subject(s)
Neoplasms , Signal Transduction , Humans , DNA Repair , Epithelial Cells , Breast , Fanconi Anemia Complementation Group N Protein/genetics
2.
Angiogenesis ; 25(2): 259-274, 2022 05.
Article in English | MEDLINE | ID: mdl-34997404

ABSTRACT

Hypoxia plays an important regulatory role in the vasculature to adjust blood flow to meet metabolic requirements. At the level of gene transcription, the responses are mediated by hypoxia-inducible factor (HIF) the stability of which is controlled by the HIF prolyl 4-hydroxylase-2 (PHD2). In the lungs hypoxia results in vasoconstriction, however, the pathophysiological relevance of PHD2 in the major arterial cell types; endothelial cells (ECs) and arterial smooth muscle cells (aSMCs) in the adult vasculature is incompletely characterized. Here, we investigated PHD2-dependent vascular homeostasis utilizing inducible deletions of PHD2 either in ECs (Phd2∆ECi) or in aSMCs (Phd2∆aSMC). Cardiovascular function and lung pathologies were studied using echocardiography, Doppler ultrasonography, intraventricular pressure measurement, histological, ultrastructural, and transcriptional methods. Cell intrinsic responses were investigated in hypoxia and in conditions mimicking hypertension-induced hemodynamic stress. Phd2∆ECi resulted in progressive pulmonary disease characterized by a thickened respiratory basement membrane (BM), alveolar fibrosis, increased pulmonary artery pressure, and adaptive hypertrophy of the right ventricle (RV). A low oxygen environment resulted in alterations in cultured ECs similar to those in Phd2∆ECi mice, involving BM components and vascular tone regulators favoring the contraction of SMCs. In contrast, Phd2∆aSMC resulted in elevated RV pressure without alterations in vascular tone regulators. Mechanistically, PHD2 inhibition in aSMCs involved  actin polymerization -related tension development via activated cofilin. The results also indicated that hemodynamic stress, rather than PHD2-dependent hypoxia response alone, potentiates structural remodeling of the extracellular matrix in the pulmonary microvasculature and respiratory failure.


Subject(s)
Hypertension, Pulmonary , Animals , Arteries/metabolism , Endothelial Cells/metabolism , Fibrosis , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mice , Myocytes, Smooth Muscle/pathology , Prolyl Hydroxylases/metabolism
3.
Hum Mol Genet ; 24(22): 6374-89, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26319232

ABSTRACT

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.


Subject(s)
Receptor, TIE-2/genetics , Vascular Malformations/genetics , Amino Acid Substitution , Animals , Cell Movement/genetics , Endothelial Cells/metabolism , Female , Fibrin Fibrinogen Degradation Products , Human Umbilical Vein Endothelial Cells , Humans , Ligands , Mice , Mice, SCID , Mutation , Phosphorylation , Receptor, TIE-2/metabolism , Signal Transduction , Spheroids, Cellular , Vascular Malformations/enzymology
4.
Hum Mol Genet ; 22(17): 3438-48, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23633549

ABSTRACT

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.


Subject(s)
Amino Acid Transport Systems, Neutral/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Vascular Malformations/genetics , Vascular Malformations/metabolism , Cell Movement/genetics , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Germ-Line Mutation , Humans , Muscle, Smooth, Vascular/metabolism , Phosphorylation , Signal Transduction/genetics , Signal Transduction/physiology
5.
J Cell Sci ; 125(Pt 9): 2212-23, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22357955

ABSTRACT

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.


Subject(s)
Angiopoietin-1/chemistry , Angiopoietin-2/chemistry , Endothelium, Vascular/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Vitreous Body/blood supply , Angiopoietin-1/genetics , Angiopoietin-1/pharmacology , Angiopoietin-2/genetics , Angiopoietin-2/pharmacology , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell-Matrix Junctions/drug effects , Cell-Matrix Junctions/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gene Expression/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha2beta1/genetics , Integrin alpha2beta1/metabolism , Intravitreal Injections , Mice , Microtubules/drug effects , Microtubules/metabolism , NIH 3T3 Cells , Neovascularization, Physiologic , Protein Multimerization , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-2 , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects
6.
Basic Clin Pharmacol Toxicol ; 123 Suppl 5: 6-19, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29668117

ABSTRACT

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.


Subject(s)
Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Vascular Diseases/prevention & control , Vascular Malformations/drug therapy , Veins/abnormalities , Animals , Biomarkers/analysis , Biomarkers/metabolism , Chronic Disease/prevention & control , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Humans , Molecular Targeted Therapy/methods , Mutation , Protein Kinase Inhibitors/pharmacology , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Vascular Diseases/etiology , Vascular Malformations/complications , Vascular Malformations/genetics
7.
Elife ; 72018 11 16.
Article in English | MEDLINE | ID: mdl-30444491

ABSTRACT

The maintenance of fluid homeostasis is necessary for function of the neural retina; however, little is known about the significance of potential fluid management mechanisms. Here, we investigated angiopoietin-4 (Angpt4, also known as Ang3), a poorly characterized ligand for endothelial receptor tyrosine kinase Tie2, in mouse retina model. By using genetic reporter, fate mapping, and in situ hybridization, we found Angpt4 expression in a specific sub-population of astrocytes at the site where venous morphogenesis occurs and that lower oxygen tension, which distinguishes peripheral and venous locations, enhances Angpt4 expression. Correlating with its spatiotemporal expression, deletion of Angpt4 resulted in defective venous development causing impaired venous drainage and defects in neuronal cells. In vitro characterization of angiopoietin-4 proteins revealed both ligand-specific and redundant functions among the angiopoietins. Our study identifies Angpt4 as the first growth factor for venous-specific development and its importance in venous remodeling, retinal fluid clearance and neuronal function.


Subject(s)
Angiopoietins/metabolism , Neovascularization, Physiologic , Regional Blood Flow , Retina/physiology , Veins/physiology , Angiopoietins/genetics , Animals , Astrocytes/enzymology , Cells, Cultured , Gene Deletion , Mice, Inbred C57BL , Mice, Knockout
8.
J Invest Dermatol ; 137(1): 207-216, 2017 01.
Article in English | MEDLINE | ID: mdl-27519652

ABSTRACT

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.


Subject(s)
Gastrointestinal Neoplasms/genetics , Genetic Predisposition to Disease/epidemiology , Mutation , Nevus, Blue/genetics , Receptor, TIE-2/genetics , Skin Neoplasms/genetics , Vascular Malformations/genetics , Belgium , Cohort Studies , Female , Gastrointestinal Neoplasms/diagnosis , Humans , Incidence , Male , Nevus, Blue/diagnosis , Rare Diseases , Skin Neoplasms/diagnosis , Vascular Malformations/diagnosis
9.
J Clin Invest ; 125(9): 3491-504, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26258417

ABSTRACT

Venous malformations (VMs) are composed of ectatic veins with scarce smooth muscle cell coverage. Activating mutations in the endothelial cell tyrosine kinase receptor TIE2 are a common cause of these lesions. VMs cause deformity, pain, and local intravascular coagulopathy, and they expand with time. Targeted pharmacological therapies are not available for this condition. Here, we generated a model of VMs by injecting HUVECs expressing the most frequent VM-causing TIE2 mutation, TIE2-L914F, into immune-deficient mice. TIE2-L914F-expressing HUVECs formed VMs with ectatic blood-filled channels that enlarged over time. We tested both rapamycin and a TIE2 tyrosine kinase inhibitor (TIE2-TKI) for their effects on murine VM expansion and for their ability to inhibit mutant TIE2 signaling. Rapamycin prevented VM growth, while TIE2-TKI had no effect. In cultured TIE2-L914F-expressing HUVECs, rapamycin effectively reduced mutant TIE2-induced AKT signaling and, though TIE2-TKI did target the WT receptor, it only weakly suppressed mutant-induced AKT signaling. In a prospective clinical pilot study, we analyzed the effects of rapamycin in 6 patients with difficult-to-treat venous anomalies. Rapamycin reduced pain, bleeding, lesion size, functional and esthetic impairment, and intravascular coagulopathy. This study provides a VM model that allows evaluation of potential therapeutic strategies and demonstrates that rapamycin provides clinical improvement in patients with venous malformation.


Subject(s)
Immunosuppressive Agents/administration & dosage , Mutation, Missense , Receptor, TIE-2 , Signal Transduction , Sirolimus/administration & dosage , Vascular Malformations , Adolescent , Adult , Animals , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Male , Mice , Mice, Nude , Middle Aged , Pilot Projects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Vascular Malformations/drug therapy , Vascular Malformations/genetics , Vascular Malformations/metabolism , Vascular Malformations/pathology , Veins
SELECTION OF CITATIONS
SEARCH DETAIL