Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38892351

ABSTRACT

Pulmonary arteriovenous malformations (PAVMs) are vascular anomalies resulting in abnormal connections between pulmonary arteries and veins. In 80% of cases, PAVMs are present from birth, but clinical manifestations are rarely seen in childhood. These congenital malformations are typically associated with Hereditary Hemorrhagic Telangiectasia (HHT), a rare disease that affects 1 in 5000/8000 individuals. HHT disease is frequently caused by mutations in genes involved in the TGF-ß pathway. However, approximately 15% of patients do not have a genetic diagnosis and, among the genetically diagnosed, more than 33% do not meet the Curaçao criteria. This makes clinical diagnosis even more challenging in the pediatric age group. Here, we introduce an 8-year-old patient bearing a severe phenotype of multiple diffuse PAVMs caused by an unknown mutation which ended in lung transplantation. Phenotypically, the case under study follows a molecular pattern which is HHT-like. Therefore, molecular- biological and cellular-functional analyses have been performed in primary endothelial cells (ECs) isolated from the explanted lung. The findings revealed a loss of functionality in lung endothelial tissue and a stimulation of endothelial-to-mesenchymal transition. Understanding the molecular basis of this transition could potentially offer new therapeutic strategies to delay lung transplantation in severe cases.


Subject(s)
Endothelial Cells , Pulmonary Artery , Pulmonary Veins , Telangiectasia, Hereditary Hemorrhagic , Humans , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Child , Pulmonary Artery/abnormalities , Pulmonary Artery/pathology , Pulmonary Veins/abnormalities , Pulmonary Veins/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Male , Mutation , Arteriovenous Malformations/genetics , Arteriovenous Malformations/pathology , Arteriovenous Malformations/metabolism , Epithelial-Mesenchymal Transition/genetics , Lung Transplantation , Arteriovenous Fistula/pathology , Arteriovenous Fistula/genetics , Lung/pathology , Lung/blood supply , Female
2.
Acta Neurochir (Wien) ; 165(12): 4241-4251, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37843608

ABSTRACT

PURPOSE: Von Hippel-Lindau (VHL) is a rare inherited disease mainly characterized by the growth of tumours, predominantly hemangioblastomas (Hbs) in the CNS and retina, and renal carcinomas. The natural history of VHL disease is variable, differing in the age of onset and its penetrance, even among relatives. Unfortunately, sometimes VHL shows more severe than average: the onset starts in adolescence, and surgeries are required almost every year. In these cases, the factor that triggers the appearance and growth of Hbs usually remains unknown, although additional mutations are suspected. METHODS: We present the case of a VHL patient whose first surgery was at 13 years of age. Then, along his next 8 years, he has undergone 5 surgeries for resection of 10 CNS Hbs. To clarify this severe VHL condition, DNA from a CNS Hb and white blood cells (WBC) was sequenced using next-generation sequencing technology. RESULTS: Massive DNA sequencing of the WBC (germ line) revealed a pathogenic mutation in CHEK2 and the complete loss of a VHL allele (both tumour suppressors). Moreover, in the tumour sample, several mutations, in BRAF1 and PTPN11 were found. Familiar segregation studies showed that CHEK2 mutation was in the maternal lineage, while VHL was inherited by paternal lineage. CONCLUSIONS: Finally, clinical history correlated to the different genotypes in the family, concluding that the severity of these VHL manifestations are due to both, VHL-and-CHEK2 mutations. This case report aims to notice the importance of deeper genetic analyses, in inherited rare diseases, to uncover non-expected mutations.


Subject(s)
Carcinoma, Renal Cell , Hemangioblastoma , Kidney Neoplasms , von Hippel-Lindau Disease , Male , Adolescent , Humans , Hemangioblastoma/genetics , Hemangioblastoma/surgery , Hemangioblastoma/pathology , Mutation/genetics , von Hippel-Lindau Disease/diagnosis , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology
3.
Cells ; 10(9)2021 09 04.
Article in English | MEDLINE | ID: mdl-34571962

ABSTRACT

Von Hippel-Lindau disease (VHL) is a rare hereditary disease characterized by the predisposal to develop different types of highly vascularized tumors. VHL patients carry a VHL mutation that causes partial lack of functional VHL protein (pVHL) in all cells, and a total lack thereof in cells harboring a second hit mutation. Absence of pVHL generates a prolonged state of pseudo-hypoxia in the cell due to accumulation of hypoxia inducible factor, an important transcription factor regulating pro-tumorigenic genes. The work here presented focuses on characterizing the endothelium of VHL patients, by means of blood outgrowth endothelial cells (BOECs). Transcriptome analysis of VHL-derived BOECs, further supported by in vitro assays, shows that these cells are at a disadvantage, as evidenced by loss of cell adhesion capacity, angiogenesis defects, and immune response and oxidative metabolic gene downregulation, which induce oxidative stress. These results suggest that the endothelium of VHL patients is functionally compromised and more susceptible to tumor development. These findings contribute to shedding light on the vascular landscape of VHL patients preceding the second hit mutation in the VHL gene. This knowledge could be useful in searching for new therapies for these patients and other vascular diseases.


Subject(s)
Endothelial Cells/pathology , Neovascularization, Pathologic , von Hippel-Lindau Disease/pathology , Case-Control Studies , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Mutation , Neovascularization, Pathologic/genetics , Oxidative Stress , Phenotype , Signal Transduction , Transcriptome , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/immunology , von Hippel-Lindau Disease/metabolism
4.
J Clin Med ; 9(9)2020 Sep 03.
Article in English | MEDLINE | ID: mdl-32899377

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant, vascular disorder that presents with telangiectases and arteriovenous malformations. HHT is a genetically heterogeneous disorder, involving mutations in endoglin (ENG; HHT1) and activin receptor-like kinase 1 (ACVRL1/ALK1; HHT2) genes that account for over 85% of all HHT patients. The current diagnosis of HHT patients remains at the clinical level, but many suspected patients do not have a clear HHT diagnosis or do not show pathogenic mutations in HHT genes. This situation has prompted the search for biomarkers to help in the early diagnosis of the disease. We have analyzed the plasma levels in HHT patients of selected micro-RNAs (miRNAs), small single-stranded RNAs that regulate gene expression at the transcriptional level by interacting with specific RNA targets. A total of 16 HHT1 and 17 HHT2 plasma samples from clinically confirmed patients and 16 controls were analyzed in this study. Total RNA was purified from plasma, and three selected miRNAs (miRNA-10a, miRNA-214, and miRNA-370), related to the pathobiology of cardiovascular diseases and potentially targeting ENG or ALK1, were measured by quantitative polymerase chain reaction. Compared with controls, levels of miRNA-370, whose putative target is ENG, were significantly downregulated in HHT1, but not in HHT2, whereas the levels of miRNA-10a, whose putative target is ALK1, were significantly upregulated in HHT2, but not in HHT1. In addition, the levels of miRNA-214, potentially targeting ENG and ALK1, did not change in either HHT1 or HHT2 patients versus control samples. While further studies are warranted, these results suggest that dysregulated plasma levels of miRNA-370 or miRNA-10a could help to identify undiagnosed HHT1 or HHT2 patients, respectively.

5.
Int J Mol Sci ; 20(12)2019 Jun 25.
Article in English | MEDLINE | ID: mdl-31242676

ABSTRACT

Upon inflammation, monocyte-derived macrophages (MΦ) infiltrate blood vessels to regulate several processes involved in vascular pathophysiology. However, little is known about the mediators involved. Macrophage polarization is crucial for a fast and efficient initial response (GM-MΦ) and a good resolution (M-MΦ) of the inflammatory process. The functional activity of polarized MΦ is exerted mainly through their secretome, which can target other cell types, including endothelial cells. Endoglin (CD105) is a cell surface receptor expressed by endothelial cells and MΦ that is markedly upregulated in inflammation and critically involved in angiogenesis. In addition, a soluble form of endoglin with anti-angiogenic activity has been described in inflammation-associated pathologies. The aim of this work was to identify components of the MΦ secretome involved in the shedding of soluble endoglin. We find that the GM-MΦ secretome contains metalloprotease 12 (MMP-12), a GM-MΦ specific marker that may account for the anti-angiogenic activity of the GM-MΦ secretome. Cell surface endoglin is present in both GM-MΦ and M-MΦ, but soluble endoglin is only detected in GM-MΦ culture supernatants. Moreover, MMP-12 is responsible for the shedding of soluble endoglin in vitro and in vivo by targeting membrane-bound endoglin in both MΦ and endothelial cells. These data demonstrate a direct correlation between GM-MΦ polarization, MMP-12, and soluble endoglin expression and function. By targeting endothelial cells, MMP-12 may represent a novel mediator involved in vascular homeostasis.


Subject(s)
Endoglin/metabolism , Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Matrix Metalloproteinase 12/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Disease Susceptibility , Endoglin/genetics , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Mice , Models, Biological
6.
Dis Model Mech ; 11(9)2018 09 21.
Article in English | MEDLINE | ID: mdl-30108051

ABSTRACT

Endoglin is a transmembrane glycoprotein expressed in vascular endothelium that plays a key role in angiogenesis. Mutations in the endoglin gene (ENG) cause hereditary hemorrhagic telangiectasia type 1 (HHT1), characterized by arteriovenous malformations (AVMs) in different organs. These vascular lesions derive from abnormal processes of angiogenesis, whereby aberrant vascular remodeling leads to focal loss of capillaries. Current treatments for HHT1 include antiangiogenic therapies. Interestingly, a circulating form of endoglin (also known as soluble endoglin, sEng), proteolytically released from the membrane-bound protein and displaying antiangiogenic activity, has been described in several endothelial-related pathological conditions. Using human and mouse endothelial cells, we find that sEng downregulates several pro-angiogenic and pro-migratory proteins involved in angiogenesis. However, this effect is much reduced in endothelial cells that lack endogenous transmembrane endoglin, suggesting that the antiangiogenic activity of sEng is dependent on the presence of endogenous transmembrane endoglin protein. In fact, sEng partially restores the phenotype of endoglin-silenced endothelial cells to that of normal endothelial cells. Moreover, using an established neonatal retinal model of HHT1 with depleted endoglin in the vascular endothelium, sEng treatment decreases the number of AVMs and has a normalizing effect on the vascular phenotype with respect to vessel branching, vascular density and migration of the vascular plexus towards the retinal periphery. Taken together, these data show that circulating sEng can influence vascular development and AVMs by modulating angiogenesis, and that its effect on endothelial cells depends on the expression of endogenous endoglin.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Arteriovenous Malformations/genetics , Arteriovenous Malformations/pathology , Endoglin/metabolism , Gene Expression Regulation , Neovascularization, Pathologic/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Animals , Biomarkers/metabolism , Cell Movement , Disease Models, Animal , Endoglin/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lung/pathology , Mice, Knockout , Models, Biological , Retina/pathology , Solubility , Wound Healing
7.
Arterioscler Thromb Vasc Biol ; 38(5): 1216-1229, 2018 05.
Article in English | MEDLINE | ID: mdl-29449337

ABSTRACT

OBJECTIVE: ALK1 (activin-receptor like kinase 1) is an endothelial cell-restricted receptor with high affinity for BMP (bone morphogenetic protein) 9 TGF-ß (transforming growth factor-ß) family member. Loss-of-function mutations in ALK1 cause a subtype of hereditary hemorrhagic telangiectasia-a rare disease characterized by vasculature malformations. Therapeutic strategies are aimed at reducing potential complications because of vascular malformations, but currently, there is no curative treatment for hereditary hemorrhagic telangiectasia. APPROACH AND RESULTS: In this work, we report that a reduction in ALK1 gene dosage (heterozygous ALK1+/- mice) results in enhanced retinal endothelial cell proliferation and vascular hyperplasia at the sprouting front. We found that BMP9/ALK1 represses VEGF (vascular endothelial growth factor)-mediated PI3K (phosphatidylinositol 3-kinase) by promoting the activity of the PTEN (phosphatase and tensin homolog). Consequently, loss of ALK1 function in endothelial cells results in increased activity of the PI3K pathway. These results were confirmed in cutaneous telangiectasia biopsies of patients with hereditary hemorrhagic telangiectasia 2, in which we also detected an increase in endothelial cell proliferation linked to an increase on the PI3K pathway. In mice, genetic and pharmacological inhibition of PI3K is sufficient to abolish the vascular hyperplasia of ALK1+/- retinas and in turn normalize the vasculature. CONCLUSIONS: Overall, our results indicate that the BMP9/ALK1 hub critically mediates vascular quiescence by limiting PI3K signaling and suggest that PI3K inhibitors could be used as novel therapeutic agents to treat hereditary hemorrhagic telangiectasia.


Subject(s)
Activin Receptors, Type II/genetics , Activin Receptors, Type I/genetics , Endothelial Cells/enzymology , Mutation , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinase/metabolism , Retinal Telangiectasis/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Activin Receptors, Type I/deficiency , Angiogenesis Inhibitors/pharmacology , Animals , Case-Control Studies , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Activation , Gene Deletion , Genetic Predisposition to Disease , Growth Differentiation Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Hyperplasia , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Retinal Telangiectasis/drug therapy , Retinal Telangiectasis/enzymology , Retinal Telangiectasis/pathology , Signal Transduction , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Telangiectasia, Hereditary Hemorrhagic/enzymology , Telangiectasia, Hereditary Hemorrhagic/pathology , Vascular Endothelial Growth Factor A/pharmacology
8.
Angiogenesis ; 21(1): 169-181, 2018 02.
Article in English | MEDLINE | ID: mdl-29147802

ABSTRACT

Hereditary hemorrhagic telangiectasia is an autosomal dominant trait affecting approximately 1 in 5000 people. A pathogenic DNA sequence variant in the ENG, ACVRL1 or SMAD4 genes, can be found in the majority of patients. The 12th International Scientific HHT Conference was held on June 8-11, 2017 in Dubrovnik, Croatia to present and discuss the latest scientific achievements, and was attended by over 200 scientific and clinical researchers. In total 174 abstracts were accepted of which 58 were selected for oral presentations. This article covers the basic science and clinical talks, and discussions from three theme-based workshops. We focus on significant emergent themes and unanswered questions. Understanding these topics and answering these questions will help to define the future of HHT research and therapeutics, and ultimately bring us closer to a cure.


Subject(s)
Telangiectasia, Hereditary Hemorrhagic , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Arteriovenous Malformations/genetics , Arteriovenous Malformations/metabolism , Arteriovenous Malformations/pathology , Arteriovenous Malformations/therapy , Croatia , Endoglin/genetics , Endoglin/metabolism , Epistaxis/genetics , Epistaxis/metabolism , Genetic Variation , Humans , Smad4 Protein/genetics , Smad4 Protein/metabolism , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/metabolism , Telangiectasia, Hereditary Hemorrhagic/pathology , Telangiectasia, Hereditary Hemorrhagic/therapy
9.
Expert Opin Ther Targets ; 21(10): 933-947, 2017 10.
Article in English | MEDLINE | ID: mdl-28796572

ABSTRACT

INTRODUCTION: Hereditary Haemorrhagic Telangiectasia (HHT) is as an autosomal dominant trait characterized by frequent nose bleeds, mucocutaneous telangiectases, arteriovenous malformations (AVMs) of the lung, liver and brain, and gastrointestinal bleedings due to telangiectases. HHT is originated by mutations in genes whose encoded proteins are involved in the transforming growth factor ß (TGF-ß) family signalling of vascular endothelial cells. In spite of the great advances in the diagnosis as well as in the molecular, cellular and animal models of HHT, the current treatments remain just at the palliative level. Areas covered: Pathogenic mutations in genes coding for the TGF-ß receptors endoglin (ENG) (HHT1) or the activin receptor-like kinase-1 (ACVRL1 or ALK1) (HHT2), are responsible for more than 80% of patients with HHT. Therefore, ENG and ALK1 are the main potential therapeutic targets for HHT and the focus of this review. The current status of the preclinical and clinical studies, including the anti-angiogenic strategy, have been addressed. Expert opinion: Endoglin and ALK1 are attractive therapeutic targets in HHT. Because haploinsufficiency is the pathogenic mechanism in HHT, several therapeutic approaches able to enhance protein expression and/or function of endoglin and ALK1 are keys to find novel and efficient treatments for the disease.


Subject(s)
Activin Receptors, Type II/genetics , Endoglin/genetics , Molecular Targeted Therapy , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Drug Design , Endothelial Cells/metabolism , Humans , Mutation , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/physiopathology , Transforming Growth Factor beta/metabolism
10.
BMC Med Genet ; 18(1): 20, 2017 Feb 23.
Article in English | MEDLINE | ID: mdl-28231770

ABSTRACT

BACKGROUND: Hereditary hemorrhagic telangiectasia (HHT) is a vascular multi-organ system disorder. Its diagnostic criteria include epistaxis, telangiectases in mucocutaneous sites, arteriovenous malformations (AVMs), and familial inheritance. HHT is transmitted as an autosomal dominant condition, caused in 85% of cases by mutations in either Endoglin (ENG) or Activin receptor-like kinase (ACVRL1/ACVRL1/ALK1) genes. Pathogenic mutations have been described in exons, splice junctions and, in a few cases with ENG mutations, in the proximal promoter, which creates a new ATG start site. However, no mutations affecting transcription regulation have been described to date in HHT, and this type of mutation is rarely identified in the literature on rare diseases. METHODS: Sequencing data from a family with HHT lead to single nucleotide change, c.-58G > A. The functionality and pathogenicity of this change was analyzed by in vitro mutagenesis, quantitative PCR and Gel shift assay. Student t test was used for statistical significance. RESULTS: A single nucleotide change, c.-58G > A, in the proximal ENG promoter co-segregated with HHT clinical features in an HHT family. This mutation was present in the proband and in 2 other symptomatic members, whereas 2 asymptomatic relatives did not harbor the mutation. Analysis of RNA from activated monocytes from the probands and the healthy brother revealed reduced ENG mRNA expression in the HHT patient (p = 0.005). Site-directed mutagenesis of the ENG promoter resulted in a three-fold decrease in luciferase activity of the mutant c.-58A allele compared to wild type (p = 0.005). Finally, gel shift assay identified a DNA-protein specific complex. CONCLUSIONS: The novel ENG c.-58G > A substitution in the ENG promoter co-segregates with HHT symptoms in a family and appears to affect the transcriptional regulation of the gene, resulting in reduced ENG expression. ENG c.-58G > A may therefore be a pathogenic HHT mutation leading to haploinsufficiency of Endoglin and HHT symptoms. To the best of our knowledge, this is the first report of a pathogenic mutation in HHT involving the binding site for a transcription factor in the promoter of ENG.


Subject(s)
Endoglin/genetics , Promoter Regions, Genetic/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Activin Receptors, Type II/genetics , Alleles , Base Sequence , Cell Line , DNA/chemistry , DNA/isolation & purification , DNA/metabolism , Endoglin/metabolism , Exons , Genes, Reporter , Genotype , Humans , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Mutation , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Protein Binding , Telangiectasia, Hereditary Hemorrhagic/pathology , Transcription Factors/chemistry , Transcription Factors/metabolism
11.
PLoS Genet ; 12(3): e1005935, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27010826

ABSTRACT

Endoglin is an auxiliary receptor for members of the TGF-ß superfamily and plays an important role in the homeostasis of the vessel wall. Mutations in endoglin gene (ENG) or in the closely related TGF-ß receptor type I ACVRL1/ALK1 are responsible for a rare dominant vascular dysplasia, the Hereditary Hemorrhagic Telangiectasia (HHT), or Rendu-Osler-Weber syndrome. Endoglin is also expressed in human macrophages, but its role in macrophage function remains unknown. In this work, we show that endoglin expression is triggered during the monocyte-macrophage differentiation process, both in vitro and during the in vivo differentiation of blood monocytes recruited to foci of inflammation in wild-type C57BL/6 mice. To analyze the role of endoglin in macrophages in vivo, an endoglin myeloid lineage specific knock-out mouse line (Eng(fl/fl)LysMCre) was generated. These mice show a predisposition to develop spontaneous infections by opportunistic bacteria. Eng(fl/fl)LysMCre mice also display increased survival following LPS-induced peritonitis, suggesting a delayed immune response. Phagocytic activity is impaired in peritoneal macrophages, altering one of the main functions of macrophages which contributes to the initiation of the immune response. We also observed altered expression of TGF-ß1 target genes in endoglin deficient peritoneal macrophages. Overall, the altered immune activity of endoglin deficient macrophages could help to explain the higher rate of infectious diseases seen in HHT1 patients.


Subject(s)
Activin Receptors, Type I/genetics , Immunity, Innate/genetics , Inflammation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Transforming Growth Factor beta/genetics , Activin Receptors, Type I/biosynthesis , Activin Receptors, Type II , Animals , Endoglin , Flow Cytometry , Gene Expression Regulation , Humans , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Opportunistic Infections/genetics , Opportunistic Infections/pathology , Phagocytosis/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology
12.
Angiogenesis ; 19(2): 155-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26850053

ABSTRACT

After endothelial injury, the transcription factor Krüppel-like factor 6 (KLF6) translocates into the cell nucleus to regulate a variety of target genes involved in angiogenesis, vascular repair and remodeling, including components of the membrane transforming growth factor beta (TGF-ß) receptor complex such as endoglin and activin receptor-like kinase 1. The membrane metalloproteinase 14 (MMP14 or MT1-MMP) targets endoglin to release soluble endoglin and is involved in vascular inflammation and endothelial tubulogenesis. However, little is known about the regulation of MMP14 expression during vascular wounding. In vitro denudation of monolayers of human endothelial cell monolayers leads to an increase in the KLF6 gene transcriptional rate, followed by an upregulation of MMP14 and release of soluble endoglin. Concomitant with this process, MMP14 co-localizes with endoglin in the sprouting endothelial cells surrounding the wound border. MMP14 expression at mRNA and protein levels is increased by ectopic KLF6 and downregulated by KLF6 suppression in cultured endothelial cells. Moreover, after wire-induced endothelial denudation, Klf6 (+/-) mice show lower levels of MMP14 in their vasculature compared with their wild-type siblings. Ectopic cellular expression of KLF6 results in an increased transcription rate of MMP14, and chromatin immunoprecipitation assays show that KLF6 interacts with MMP14 promoter in ECs, this interaction being enhanced during wound healing. Furthermore, KLF6 markedly increases the transcriptional activity of different reporter constructs of MMP14 gene promoter. These results suggest that KLF6 regulates MMP14 transcription and is a critical player of the gene expression network triggered during endothelial repair.


Subject(s)
Endoglin/metabolism , Kruppel-Like Transcription Factors/metabolism , Matrix Metalloproteinase 14/genetics , Proto-Oncogene Proteins/metabolism , Up-Regulation , Vascular System Injuries/enzymology , Vascular System Injuries/genetics , Animals , Base Sequence , Computer Simulation , Endoglin/genetics , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kruppel-Like Factor 6 , Matrix Metalloproteinase 14/metabolism , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solubility , Transcription, Genetic , Up-Regulation/genetics , Vascular System Injuries/pathology , Wound Healing
13.
Thromb Haemost ; 115(6): 1167-77, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26818701

ABSTRACT

Hereditary haemorrhagic telangiectasia (HHT), or Rendu-Osler-Weber syndrome, is a dominant genetic vascular disorder. In HHT, blood vessels are weak and prone to bleeding, leading to epistaxis and anaemia, severely affecting patients' quality of life. Development of vascular malformations in HHT patients is originated mainly by mutations in ACVRL1/ALK1 (activin receptor-like kinase type I) or Endoglin (ENG) genes. These genes encode proteins of the TGF-ß signalling pathway in endothelial cells, controlling angiogenesis. Haploinsufficiency of these proteins is the basis of HHT pathogenicity. It was our objective to study the efficiency of Bazedoxifene, a selective estrogen receptor modulator (SERM) in HHT, looking for a decrease in epistaxis, and understanding the underlying molecular mechanism. Plasma samples of five HHT patients were collected before, and after 1 and 3 months of Bazedoxifene treatment. ENG and ALK1 expression in activated mononuclear cells derived from blood, as well as VEGF plasma levels, were measured. Quantification of Endoglin and ALK1 mRNA was done in endothelial cells derived from HHT and healthy donors, after in vitro treatment with Bazedoxifene. Angiogenesis was also measured by tubulogenesis and wound healing assays. Upon Bazedoxifene treatment, haemoglobin levels of HHT patients increased and the quantity and frequency of epistaxis decreased. Bazedoxifene increased Endoglin and ALK1 mRNA levels, in cells derived from blood samples and in cultured endothelial cells, promoting tube formation. In conclusion, Bazedoxifene seems to decrease bleeding in HHT by partial compensation of haploinsufficiency. The results shown here are the basis of a new orphan drug designation for HHT by the European Medicine Agency (EMA).


Subject(s)
Indoles/therapeutic use , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Activin Receptors, Type II/genetics , Aged , Cells, Cultured , Endoglin/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Hemorrhage/blood , Hemorrhage/drug therapy , Hemorrhage/etiology , Humans , Middle Aged , Neovascularization, Physiologic/drug effects , Orphan Drug Production , Pilot Projects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Telangiectasia, Hereditary Hemorrhagic/complications , Telangiectasia, Hereditary Hemorrhagic/genetics , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics , Wound Healing/drug effects
14.
Cell Mol Life Sci ; 73(8): 1715-39, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26646071

ABSTRACT

The circulatory system is walled off by different cell types, including vascular mural cells and podocytes. The interaction and interplay between endothelial cells (ECs) and mural cells, such as vascular smooth muscle cells or pericytes, play a pivotal role in vascular biology. Endoglin is an RGD-containing counter-receptor for ß1 integrins and is highly expressed by ECs during angiogenesis. We find that the adhesion between vascular ECs and mural cells is enhanced by integrin activators and inhibited upon suppression of membrane endoglin or ß1-integrin, as well as by addition of soluble endoglin (SolEng), anti-integrin α5ß1 antibody or an RGD peptide. Analysis of different endoglin mutants, allowed the mapping of the endoglin RGD motif as involved in the adhesion process. In Eng (+/-) mice, a model for hereditary hemorrhagic telangectasia type 1, endoglin haploinsufficiency induces a pericyte-dependent increase in vascular permeability. Also, transgenic mice overexpressing SolEng, an animal model for preeclampsia, show podocyturia, suggesting that SolEng is responsible for podocytes detachment from glomerular capillaries. These results suggest a critical role for endoglin in integrin-mediated adhesion of mural cells and provide a better understanding on the mechanisms of vessel maturation in normal physiology as well as in pathologies such as preeclampsia or hereditary hemorrhagic telangiectasia.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion/physiology , Endothelium, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Podocytes/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, CD/genetics , Cell Line, Tumor , Disease Models, Animal , Endoglin , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Integrin beta1/genetics , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/pathology , Pregnancy , Protein Binding , RNA Interference , RNA, Small Interfering , Receptors, Cell Surface/genetics , Retina/metabolism , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology
15.
J Cell Physiol ; 230(4): 947-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25216259

ABSTRACT

Endoglin is an auxiliary cell surface receptor for TGF-ß family members. Two different alternatively spliced isoforms, long (L)-endoglin and short (S)-endoglin, have been reported. S-endoglin and L-endoglin proteins vary from each other in their cytoplasmic tails that contain 14 and 47 amino acids, respectively. A critical role for endoglin in vascular development has primarily been studied in endothelial cells. In addition, endoglin expression is upregulated during monocyte-to-macrophage differentiation; however, little is known about its role in this myeloid context. To investigate the function of endoglin in monocytes, stable transfectants expressing the two endoglin isoforms in the promonocytic human cell line U937 were generated. The differential gene expression fingerprinting of these endoglin transfectants using DNA microarrays and further bioinformatics analysis showed a clear alteration in essential biological functions, mainly those related to "Cellular Movement", including cell adhesion and transmigration. Interestingly, these cellular functions are highly dependent on adhesion molecules, including integrins α1 (CD49a, ITGA1 gene), αL (CD11a, ITGAL gene), αM (CD11b, ITGAM gene) and ß2 (CD18, ITGB2 gene) and the chemokine receptor CCR2 (CD192, CCR2 gene), which are downregulated in endoglin transfectants. Moreover, activin A (INHBA gene), a TGF-ß superfamily member involved in macrophage polarization, was distinctly affected in each endoglin transfectant, and may contribute to the regulated expression of integrins. These data were confirmed by quantitative PCR, flow cytometry and functional tests. Taken together, these results provide new insight into endoglin function in monocytes.


Subject(s)
Antigens, CD/genetics , Intracellular Signaling Peptides and Proteins/genetics , Monocytes/metabolism , Receptors, Cell Surface/genetics , Transcription, Genetic , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Endoglin , Endothelial Cells/metabolism , Genome-Wide Association Study , Humans , Integrins/metabolism , Mice , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , U937 Cells
16.
J Cell Sci ; 127(Pt 12): 2723-35, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24777481

ABSTRACT

Endoglin plays a crucial role in pathophysiological processes such as hereditary hemorrhagic telangiectasia (HHT), preeclampsia and cancer. Endoglin expression is upregulated during the monocyte-to-macrophage transition, but little is known about its regulation and function in these immune cells. Two different alternatively spliced isoforms of endoglin have been reported, L-endoglin and S-endoglin. Although L-endoglin is the predominant variant, here, we found that there was an increased expression of the S-endoglin isoform during senescence of the myeloid lineage in human and murine models. We performed a stable isotope labelling of amino acids in cell culture (SILAC) analysis of both L-endoglin and S-endoglin transfectants in the human promonocytic cell line U937. Analysis of differentially expressed protein clusters allowed the identification of cellular activities affected during aging. S-endoglin expression led to decreased cellular proliferation and a decreased survival response to granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced apoptosis, as well as increased oxidative stress. Gene expression and functional studies suggested that there was a non-redundant role for each endoglin isoform in monocyte biology. In addition, we found that S-endoglin impairs the monocytic differentiation into the pro-inflammatory M1 phenotype and contributes to the compromised status of macrophage functions during aging.


Subject(s)
Antigens, CD/metabolism , Macrophages/physiology , Receptors, Cell Surface/metabolism , Alternative Splicing , Antigens, CD/genetics , Cell Differentiation , Cell Line , Cell Lineage , Cell Polarity , Cellular Senescence , Endoglin , Gene Expression , Humans , Monocytes/physiology , Oxidative Stress , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Cell Surface/genetics
17.
BMC Med Genet ; 14: 121, 2013 Nov 25.
Article in English | MEDLINE | ID: mdl-24267784

ABSTRACT

BACKGROUND: The hereditary hemorrhagic telangiectasia syndrome (HHT), also known as the Rendu-Osler-Weber syndrome is a multiorganic vascular disorder inherited as an autosomal dominant trait. Diagnostic clinical criteria include: epistaxis, telangiectases in mucocutaneous and gastrointestinal sites, arteriovenous malformations (AVMs) most commonly found in pulmonary, hepatic and cerebral circulations, and familial inheritance. HHT is transmitted in 90% of the cases as an autosomal dominant condition due to mutations in either endoglin (ENG), or activin receptor-like kinase 1 (ACVRL1/ALK1) genes (HHT type 1 and 2, respectively). METHODS: We have carried out a genetic analysis of four independent Spanish families with HHT clinical criteria, which has permitted the identification of new large deletions in ENG. These mutations were first detected using the MLPA technique and subsequently, the deletion breakpoints were mapped using a customized copy number variation (CNV) microarray. The array was designed to cover the ENG gene and surrounding areas. RESULTS: All tested families carried large deletions ranging from 3-kb to 100-kb, involving the ENG gene promoter, several ENG exons, and the two downstream genes FGSH and CDK9. Interestingly, common breakpoints coincident with Alu repetitive sequences were found among these families. CONCLUSIONS: The systematic hybridization of DNA from HHT families, with deletions or duplications, to custom designed microarrays, could allow the mapping of breakpoints, coincident with repetitive Alu sequences that might act as "hot spots" in the development of chromosomal anomalies.


Subject(s)
Antigens, CD/genetics , Receptors, Cell Surface/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , White People/genetics , Chromosome Mapping , Cyclin-Dependent Kinase 9/genetics , DNA Copy Number Variations , Endoglin , Exons , Gene Deletion , Genetic Association Studies , Genetic Loci , Genotype , Humans , Multiplex Polymerase Chain Reaction , Phenotype , Promoter Regions, Genetic , Spain , Telangiectasia, Hereditary Hemorrhagic/pathology
18.
Am J Hum Genet ; 93(3): 530-7, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23972370

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT), the most common inherited vascular disorder, is caused by mutations in genes involved in the transforming growth factor beta (TGF-ß) signaling pathway (ENG, ACVRL1, and SMAD4). Yet, approximately 15% of individuals with clinical features of HHT do not have mutations in these genes, suggesting that there are undiscovered mutations in other genes for HHT and possibly vascular disorders with overlapping phenotypes. The genetic etiology for 191 unrelated individuals clinically suspected to have HHT was investigated with the use of exome and Sanger sequencing; these individuals had no mutations in ENG, ACVRL1, and SMAD4. Mutations in BMP9 (also known as GDF2) were identified in three unrelated probands. These three individuals had epistaxis and dermal lesions that were described as telangiectases but whose location and appearance resembled lesions described in some individuals with RASA1-related disorders (capillary malformation-arteriovenous malformation syndrome). Analyses of the variant proteins suggested that mutations negatively affect protein processing and/or function, and a bmp9-deficient zebrafish model demonstrated that BMP9 is involved in angiogenesis. These data confirm a genetic cause of a vascular-anomaly syndrome that has phenotypic overlap with HHT.


Subject(s)
Blood Vessels/abnormalities , Growth Differentiation Factors/genetics , Mutation/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Adolescent , Adult , Amino Acid Substitution/genetics , Animals , Female , Genetic Predisposition to Disease , Growth Differentiation Factor 2 , Humans , Ligands , Male , Mice , Mutation, Missense/genetics , Phenotype , Protein Binding , Protein Processing, Post-Translational , Signal Transduction/genetics , Syndrome , Transforming Growth Factor beta/genetics , Zebrafish/genetics
19.
Circ Res ; 112(1): 113-27, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23048070

ABSTRACT

RATIONALE: Activin receptor-like kinase-1 (ALK1) is an endothelial transforming growth factor ß receptor involved in angiogenesis. ALK1 expression is high in the embryo vasculature, becoming less detectable in the quiescent endothelium of adult stages. However, ALK1 expression becomes rapidly increased after angiogenic stimuli such as vascular injury. OBJECTIVE: To characterize the molecular mechanisms underlying the regulation of ALK1 on vascular injury. METHODS AND RESULTS: Alk1 becomes strongly upregulated in endothelial (EC) and vascular smooth muscle cells of mouse femoral arteries after wire-induced endothelial denudation. In vitro denudation of monolayers of human umbilical vein ECs also leads to an increase in ALK1. Interestingly, a key factor in tissue remodeling, Krüppel-like factor 6 (KLF6) translocates to the cell nucleus during wound healing, concomitantly with an increase in the ALK1 gene transcriptional rate. KLF6 knock down in human umbilical vein ECs promotes ALK1 mRNA downregulation. Moreover, Klf6(+/-) mice have lower levels of Alk1 in their vasculature compared with their wild-type siblings. Chromatin immunoprecipitation assays show that KLF6 interacts with ALK1 promoter in ECs, and this interaction is enhanced during wound healing. We demonstrate that KLF6 is transactivating ALK1 gene, and this transactivation occurs by a synergistic cooperative mechanism with specificity protein 1. Finally, Alk1 levels in vascular smooth muscle cells are not directly upregulated in response to damage, but in response to soluble factors, such as interleukin 6, released from ECs after injury. CONCLUSIONS: ALK1 is upregulated in ECs during vascular injury by a synergistic cooperative mechanism between KLF6 and specificity protein 1, and in vascular smooth muscle cells by an EC-vascular smooth muscle cell paracrine communication during vascular remodeling.


Subject(s)
Activin Receptors, Type II/metabolism , Activin Receptors, Type I/metabolism , Endothelial Cells/metabolism , Femoral Artery/metabolism , Kruppel-Like Transcription Factors/metabolism , Proto-Oncogene Proteins/metabolism , Vascular System Injuries/metabolism , Wound Healing , Activin Receptors, Type I/genetics , Activin Receptors, Type II/genetics , Animals , Binding Sites , Disease Models, Animal , Endothelial Cells/pathology , Femoral Artery/injuries , Femoral Artery/pathology , Gene Expression Regulation , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-6/metabolism , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Paracrine Communication , Promoter Regions, Genetic , Protein Transport , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Messenger/metabolism , Sp1 Transcription Factor/metabolism , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Vascular System Injuries/genetics , Vascular System Injuries/pathology
20.
Blood ; 121(2): 403-15, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23074273

ABSTRACT

Human endoglin is an RGD-containing transmembrane glycoprotein identified in vascular endothelial cells. Although endoglin is essential for angiogenesis and its expression is up-regulated in inflammation and at sites of leukocyte extravasation, its role in leukocyte trafficking is unknown. This function was tested in endoglin heterozygous mice (Eng(+/-)) and their wild-type siblings Eng(+/+) treated with carrageenan or LPS as inflammatory agents. Both stimuli showed that inflammation-induced leukocyte transendothelial migration to peritoneum or lungs was significantly lower in Eng(+/-) than in Eng(+/+) mice. Leukocyte transmigration through cell monolayers of endoglin transfectants was clearly enhanced in the presence of endoglin. Coating transwells with the RGD-containing extracellular domain of endoglin, enhanced leukocyte transmigration, and this increased motility was inhibited by soluble endoglin. Leukocytes stimulated with CXCL12, a chemokine involved in inflammation, strongly adhered to endoglin-coated plates and to endoglin-expressing endothelial cells. This endoglin-dependent adhesion was abolished by soluble endoglin, RGD peptides, the anti-integrin α5ß1 inhibitory antibody LIA1/2 and the chemokine receptor inhibitor AMD3100. These results demonstrate for the first time that endothelial endoglin interacts with leukocyte integrin α5ß1 via its RGD motif, and this adhesion process is stimulated by the inflammatory chemokine CXCL12, suggesting a regulatory role for endoglin in transendothelial leukocyte trafficking.


Subject(s)
Antigens, CD/metabolism , Chemotaxis, Leukocyte/physiology , Endothelial Cells/metabolism , Inflammation/metabolism , Receptors, Cell Surface/metabolism , Transendothelial and Transepithelial Migration/physiology , Animals , Cell Adhesion/physiology , Cell Migration Assays, Leukocyte , Chemokine CXCL12/metabolism , Endoglin , Flow Cytometry , Humans , Integrin alpha5beta1/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Fluorescence , Transcellular Cell Migration/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...