Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(20)2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34681942

ABSTRACT

Endoglin (Eng, CD105) is a type I membrane glycoprotein that functions in endothelial cells as an auxiliary receptor for transforming growth factor ß (TGF-ß)/bone morphogenetic protein (BMP) family members and as an integrin ligand, modulating the vascular pathophysiology. Besides the membrane-bound endoglin, there is a soluble form of endoglin (sEng) that can be generated by the action of the matrix metalloproteinase (MMP)-14 or -12 on the juxtamembrane region of its ectodomain. High levels of sEng have been reported in patients with preeclampsia, hypercholesterolemia, atherosclerosis and cancer. In addition, sEng is a marker of cardiovascular damage in patients with hypertension and diabetes, plays a pathogenic role in preeclampsia, and inhibits angiogenesis and tumor proliferation, migration, and invasion in cancer. However, the mechanisms of action of sEng have not yet been elucidated, and new tools and experimental approaches are necessary to advance in this field. To this end, we aimed to obtain a fluorescent form of sEng as a new tool for biological imaging. Thus, we cloned the extracellular domain of endoglin in the pEGFP-N1 plasmid to generate a fusion protein with green fluorescent protein (GFP), giving rise to pEGFP-N1/Eng.EC. The recombinant fusion protein was characterized by transient and stable transfections in CHO-K1 cells using fluorescence microscopy, SDS-PAGE, immunodetection, and ELISA techniques. Upon transfection with pEGFP-N1/Eng.EC, fluorescence was readily detected in cells, indicating that the GFP contained in the recombinant protein was properly folded into the cytosol. Furthermore, as evidenced by Western blot analysis, the secreted fusion protein yielded the expected molecular mass and displayed a specific fluorescent signal. The fusion protein was also able to bind to BMP9 and BMP10 in vitro. Therefore, the construct described here could be used as a tool for functional in vitro studies of the extracellular domain of endoglin.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Endoglin/metabolism , Green Fluorescent Proteins/metabolism , Growth Differentiation Factor 2/metabolism , Recombinant Fusion Proteins/metabolism , Animals , CHO Cells , Cricetulus , Endoglin/genetics , Green Fluorescent Proteins/genetics , Humans , Recombinant Fusion Proteins/genetics
2.
Gene ; 647: 85-92, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29305977

ABSTRACT

Hereditary Hemorrhagic Telangiectasia (HHT) is a rare disease, with an autosomal dominant inheritance and a worldwide incidence of about 1: 5000 individuals. In >80% of patients, HHT is caused by mutations in either ENG or ACVRL1, which code for ENDOGLIN and Activin A Receptor Type II-Like Kinase 1 (ALK1), belonging to the TGF-ß/BMP signalling pathway. Typical HHT clinical features are mucocutaneous telangiectases, arteriovenous malformations, spontaneous and recurrent epistaxis, as well as gastrointestinal bleedings. An additional, but less frequent, clinical manifestation in some HHT patients is the presence of Pulmonary Arterial Hypertension (PAH). The aim of this work is to describe the functional role of a novel ENG intronic variant found in a patient affected by both HHT and PAH, in order to assess whether it has a pathogenic role. We proved that the variant lies in a novel binding-site for the transcription factor Sp1, known to be involved in the regulation of ENG and ACVRL1 transcription. We confirmed a pathogenic role for this intronic variant, as it significantly reduces ENG transcription by affecting this novel Sp1 binding-site.


Subject(s)
Binding Sites/genetics , Endoglin/genetics , Genetic Variation/genetics , Sp1 Transcription Factor/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Activin Receptors, Type II/genetics , Female , Gene Expression Regulation/genetics , Humans , Male , Middle Aged , Protein Binding/genetics , Signal Transduction/genetics
3.
Cell Mol Life Sci ; 75(7): 1269-1284, 2018 04.
Article in English | MEDLINE | ID: mdl-29080903

ABSTRACT

Complex interactions between platelets and activated endothelium occur during the thrombo-inflammatory reaction at sites of vascular injuries and during vascular hemostasis. The endothelial receptor endoglin is involved in inflammation through integrin-mediated leukocyte adhesion and transmigration; and heterozygous mutations in the endoglin gene cause hereditary hemorrhagic telangiectasia type 1. This vascular disease is characterized by a bleeding tendency that is postulated to be a consequence of telangiectasia fragility rather than a platelet defect, since platelets display normal functions in vitro in this condition. Here, we hypothesize that endoglin may act as an adhesion molecule involved in the interaction between endothelial cells and platelets through integrin recognition. We find that the extracellular domain of human endoglin promotes specific platelet adhesion under static conditions and confers resistance of adherent platelets to detachment upon exposure to flow. Also, platelets adhere to confluent endothelial cells in an endoglin-mediated process. Remarkably, Chinese hamster ovary cells ectopically expressing the human αIIbß3 integrin acquire the capacity to adhere to myoblast transfectants expressing human endoglin, whereas platelets from Glanzmann's thrombasthenia patients lacking the αIIbß3 integrin are defective for endoglin-dependent adhesion to endothelial cells. Furthermore, the bleeding time, but not the prothrombin time, is significantly prolonged in endoglin-haplodeficient (Eng +/-) mice compared to Eng +/+ animals. These results suggest a new role for endoglin in αIIbß3 integrin-mediated adhesion of platelets to the endothelium, and may provide a better understanding on the basic cellular mechanisms involved in hemostasis and thrombo-inflammatory events.


Subject(s)
Blood Platelets/metabolism , Cell Communication , Endoglin/metabolism , Endothelial Cells/metabolism , Animals , Blood Platelets/cytology , CHO Cells , Cell Adhesion , Cell Line , Cells, Cultured , Cricetinae , Cricetulus , Endoglin/genetics , Endothelial Cells/cytology , Humans , Mice, Inbred C57BL , Mice, Knockout , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
4.
Biochim Biophys Acta ; 1862(9): 1801-14, 2016 09.
Article in English | MEDLINE | ID: mdl-27321931

ABSTRACT

Transforming growth factor beta 1 (TGF-ß1) is one of the most studied cytokines involved in renal tubulo-interstitial fibrosis, which is characterized by myofibroblast abundance and proliferation, and high buildup of extracellular matrix in the tubular interstitium leading to organ failure. Endoglin (Eng) is a 180-kDa homodimeric transmembrane protein that regulates a great number of TGF-ß1 actions in different biological processes, including ECM synthesis. High levels of Eng have been observed in experimental models of renal fibrosis or in biopsies from patients with chronic kidney disease. In humans and mice, two Eng isoforms are generated by alternative splicing, L-Eng and S-Eng that differ in the length and composition of their cytoplasmic domains. We have previously described that L-Eng overexpression promotes renal fibrosis after unilateral ureteral obstruction (UUO). However, the role of S-Eng in renal fibrosis is unknown and its study would let us analyze the possible function of the cytoplasmic domain of Eng in this process. For this purpose, we have generated a mice strain that overexpresses S-Eng (S-ENG(+)) and we have performed an UUO in S-ENG(+) and their wild type (WT) control mice. Our results indicate that obstructed kidney of S-ENG(+) mice shows lower levels of tubulo-interstitial fibrosis, less inflammation and less interstitial cell proliferation than WT littermates. Moreover, S-ENG(+) mice show less activation of Smad1 and Smad2/3 pathways. Thus, S-Eng overexpression reduces UUO-induced renal fibrosis and some associated mechanisms. As L-Eng overexpression provokes renal fibrosis we conclude that Eng-mediated induction of renal fibrosis in this model is dependent on its cytoplasmic domain.


Subject(s)
Endoglin/genetics , Endoglin/metabolism , Kidney/metabolism , Kidney/pathology , Nephritis/prevention & control , Ureteral Obstruction/metabolism , Animals , Cell Proliferation , Collagen Type I/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Biological , Myofibroblasts/metabolism , Myofibroblasts/pathology , Nephritis/metabolism , Nephritis/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
5.
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
6.
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
7.
Carcinogenesis ; 36(2): 212-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25503931

ABSTRACT

Increased levels of soluble endoglin (Sol-Eng) correlate with poor outcome in human cancer. We have previously shown that shedding of membrane endoglin, and concomitant release of Sol-Eng is a late event in chemical mouse skin carcinogenesis associated with the development of undifferentiated spindle cell carcinomas (SpCCs). In this report, we show that mouse skin SpCCs exhibit a high expression of hepatocyte growth factor (HGF) and an elevated ratio of its active tyrosine kinase receptor Met versus total Met levels. We have evaluated the effect of Sol-Eng in spindle carcinoma cells by transfection of a cDNA encoding most of the endoglin ectodomain or by using purified recombinant Sol-Eng. We found that Sol-Eng inhibited both mitogen-activated protein kinase (MAPK) activity and cell growth in vitro and in vivo. Sol-Eng also blocked MAPK activation by transforming growth factor-ß1 (TGF-ß1) and impaired both basal and HGF-induced activation of Met and downstream MAPK. Moreover, Sol-Eng strongly reduced basal and HGF-stimulated spindle cell migration and invasion. Both Sol-Eng and full-length endoglin were shown to interact with Met by coimmunoprecipitation experiments. However, full-length endoglin expressed at the plasma membrane of spindle carcinoma cells had no effect on Met signaling activity, and was unable to inhibit HGF-induced cell migration/invasion. These results point to a paradoxical suppressor role for Sol-Eng in carcinogenesis.


Subject(s)
Antigens, CD/metabolism , Carcinogenesis/metabolism , Hepatocyte Growth Factor/biosynthesis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Sarcoma/metabolism , Skin Neoplasms/metabolism , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Antigens, CD/genetics , Carcinogenesis/pathology , Cell Movement/genetics , Cell Proliferation/genetics , DNA, Complementary/genetics , Endoglin , Enzyme Activation , Female , HEK293 Cells , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Prognosis , Receptors, Cell Surface/genetics , Sarcoma/pathology , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Transforming Growth Factor beta1/antagonists & inhibitors , Tumor Cells, Cultured
8.
PLoS One ; 9(10): e110365, 2014.
Article in English | MEDLINE | ID: mdl-25313562

ABSTRACT

Transforming growth factor-ß (TGF-ß) plays a pivotal role in renal fibrosis. Endoglin, a 180 KDa membrane glycoprotein, is a TGF-ß co-receptor overexpressed in several models of chronic kidney disease, but its function in renal fibrosis remains uncertain. Two membrane isoforms generated by alternative splicing have been described, L-Endoglin (long) and S-Endoglin (short) that differ from each other in their cytoplasmic tails, being L-Endoglin the most abundant isoform. The aim of this study was to assess the effect of L-Endoglin overexpression in renal tubulo-interstitial fibrosis. For this purpose, a transgenic mouse which ubiquitously overexpresses human L-Endoglin (L-ENG+) was generated and unilateral ureteral obstruction (UUO) was performed in L-ENG+ mice and their wild type (WT) littermates. Obstructed kidneys from L-ENG+ mice showed higher amounts of type I collagen and fibronectin but similar levels of α-smooth muscle actin (α-SMA) than obstructed kidneys from WT mice. Smad1 and Smad3 phosphorylation were significantly higher in obstructed kidneys from L-ENG+ than in WT mice. Our results suggest that the higher increase of renal fibrosis observed in L-ENG+ mice is not due to a major abundance of myofibroblasts, as similar levels of α-SMA were observed in both L-ENG+ and WT mice, but to the higher collagen and fibronectin synthesis by these fibroblasts. Furthermore, in vivo L-Endoglin overexpression potentiates Smad1 and Smad3 pathways and this effect is associated with higher renal fibrosis development.


Subject(s)
Antigens, CD/genetics , Gene Expression , Kidney Diseases/etiology , Kidney Diseases/pathology , Receptors, Cell Surface/genetics , Ureteral Obstruction/complications , Animals , Collagen/metabolism , Disease Models, Animal , Endoglin , Extracellular Matrix/metabolism , Fibronectins , Fibrosis , Humans , Kidney Diseases/metabolism , Mice , Mice, Transgenic , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
9.
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
10.
PLoS One ; 8(1): e54591, 2013.
Article in English | MEDLINE | ID: mdl-23336009

ABSTRACT

Endoglin is a transmembrane auxiliary receptor for transforming growth factor-beta (TGF-beta) that is predominantly expressed on proliferating endothelial cells. It plays a wide range of physiological roles but its importance on energy balance or insulin sensitivity has been unexplored. Endoglin deficient mice die during midgestation due to cardiovascular defects. Here we report for first time that heterozygous endoglin deficiency in mice decreases high fat diet-induced hepatic triglyceride content and insulin levels. Importantly, these effects are independent of changes in body weight or adiposity. At molecular level, we failed to detect relevant changes in the insulin signalling pathway at basal levels in liver, muscle or adipose tissues that could explain the insulin-dependent effect. However, we found decreased triglyceride content in the liver of endoglin heterozygous mice fed a high fat diet in comparison to their wild type littermates. Overall, our findings indicate that endoglin is a potentially important physiological mediator of insulin levels and hepatic lipid metabolism.


Subject(s)
Diet, High-Fat , Heterozygote , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Liver/metabolism , Triglycerides/metabolism , Animals , Body Weight/genetics , Endoglin , Energy Metabolism , Glucose/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Male , Mice , Mice, Knockout , Phenotype , Signal Transduction
11.
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
12.
Cancer Immunol Immunother ; 62(3): 541-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23076642

ABSTRACT

TGF-beta superfamily co-receptors are emerging as targets for cancer therapy, acting both directly on cells and indirectly on the tumour neovasculature. Endoglin (CD105), an accessory component of the TGF-beta receptor complex, is expressed in certain melanoma cell lines and the endothelial cells of tumour neovessels. Targeting endoglin with immunotoxins is an attractive approach for actively suppressing the blood supply to tumours. Here, we report evidence indicating that endoglin is expressed in mouse melanoma B16MEL4A5 and mouse fibroblast L929 cell lines. We prepared an immunotoxin to target endoglin by coupling the rat anti-mouse MJ7/18 (IgG2a) monoclonal antibody (mAb) to the non-toxic type 2 ribosome-inactivating protein nigrin b (Ngb) with N-succinimidyl 3-(2-pyridyldithio)-propionate (SPDP) as a linker with a molar nigrin b at a MJ7/18 stoichiometry of 2:1. The MJ7-Ngb immunotoxin generated killed both cell lines, with IC50 values of 4.2 × 10(-9) M for B16MEL4A5 and 7.7 × 10(-11) M for L929 cells. For in vivo assays of the immunotoxin, B16MEL4A5 cells were injected subcutaneously into the right flanks of 6-week-old C57BL/6 J mice. When the animals developed palpable solid tumours, they were subjected to treatment with the immunotoxin. While treatment with either MJ7/18 mAb or Ngb did not affect tumour development, treatment with the immunotoxin completely and steadily blocked tumour growth up to 7 days, after which some tumours re-grew. Thus, vascular-targeting therapy with this anti-vascular immunotoxin could promote the destruction of newly created tumour vessels at early stages of B16MEL4A5 tumour development and readily accessible CD105+ B16MEL4A5 melanoma cells.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Immunotoxins/therapeutic use , Melanoma, Experimental/therapy , Plant Proteins/administration & dosage , Receptors, Cell Surface/immunology , Ribosome Inactivating Proteins/administration & dosage , Animals , Antibodies, Monoclonal/pharmacology , Cell Line , Cell Line, Tumor , Endoglin , Immunotoxins/pharmacology , Melanoma, Experimental/blood supply , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy
13.
Circulation ; 126(22): 2612-24, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23110859

ABSTRACT

BACKGROUND: Ischemia in the placenta is considered the base of the pathogenesis of preeclampsia, a pregnancy-specific syndrome in which soluble endoglin (sEng) is a prognostic marker and plays a pathogenic role. Here, we investigated the effects of hypoxia and the downstream pathways in the release of sEng. METHODS AND RESULTS: Under hypoxic conditions, the trophoblast-like cell line JAR showed an increase in sEng parallel to an elevated formation of reactive oxygen species. Because reactive oxygen species are related to the formation of oxysterols, we assessed the effect of 22-(R)-hydroxycholesterol, a natural ligand of the liver X receptor (LXR), and the LXR synthetic agonist T0901317. Treatment of JAR cells or human placental explants with 22-(R)-hydroxycholesterol or T0901317 resulted in a clear increase in sEng that was dependent on LXR. These LXR agonists induced an increased matrix metalloproteinase-14 expression and activity and a significant reduction of its endogenous inhibitor, tissue inhibitor of metalloproteinase-3. In addition, mice treated with LXR agonists underwent an increase in the plasma sEng levels, concomitant with an increase in arterial pressure. Moreover, transgenic mice overexpressing sEng displayed high blood pressure. Finally, administration of an endoglin peptide containing the consensus matrix metalloproteinase-14 cleavage site G-L prevented the oxysterol-dependent increase in arterial pressure and sEng levels in mice. CONCLUSIONS: These studies provide a clue to the involvement of the LXR pathway in sEng release and its pathogenic role in vascular disorders such as preeclampsia.


Subject(s)
Antigens, CD/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ischemia/metabolism , Placenta Diseases/metabolism , Pre-Eclampsia/metabolism , Receptors, Cell Surface/metabolism , Animals , Anticholesteremic Agents/pharmacology , Blood Pressure/physiology , Cell Line, Tumor , Choriocarcinoma , Endoglin , Female , Human Umbilical Vein Endothelial Cells , Humans , Hydrocarbons, Fluorinated/pharmacology , Hydroxycholesterols/pharmacology , Ischemia/pathology , Liver X Receptors , Male , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orphan Nuclear Receptors/metabolism , Placenta Diseases/pathology , Pre-Eclampsia/pathology , Pregnancy , Sulfonamides/pharmacology , Tissue Inhibitor of Metalloproteinase-3/metabolism , Uterine Neoplasms
14.
Gene ; 491(1): 31-9, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21982971

ABSTRACT

Endoglin is a TGF-ß co-receptor expressed in endothelial cells, where it plays a crucial role in angiogenesis, cardiovascular development and vascular remodeling. In humans, mutations in the endoglin gene give rise to Hereditary Hemorrhagic Telangiectasia type 1 (HHT1), an autosomal dominant disorder associated with vascular lesions in skin, mucosa and internal organs. So far, endoglin cDNA has been sequenced in several species from mammals, amphibians and birds. While in mammals the characterization of endoglin protein expression and function is well documented, little is known about the protein homologue in birds. In silico analysis by multiple sequences alignment showed a low homology score of 30-33 between the full length chicken endoglin protein and several mammalian homologues. However, a high homology score (80-85) was observed with the cytoplasmic and transmembrane regions and the overall structure of the zona pellucida (ZP) and orphan domains of the extracellular region appear to be conserved. Transient expression of chicken endoglin allowed the identification of a 180-kDa disulfide linked homodimer similar to the mammalian homologues. To further characterize its tissue expression, the novel specific monoclonal antibody (mAb) 7H5A8 was generated against chicken endoglin transfectant cells. The mAb 7H5A8 specifically recognized chicken endoglin by western blot, immunoprecipitation, immunofluorescence flow cytometry as well as immunofluorescence microscopy assays and displayed a positive staining of the endothelium in veins and arteries from frozen tissue sections of lung and bursa of Fabricius. These results may help to further understand the endoglin expression in vertebrates.


Subject(s)
Chickens/genetics , Animals , Antigens, CD , Tissue Distribution , Zona Pellucida/metabolism
15.
Orphanet J Rare Dis ; 6: 85, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22192717

ABSTRACT

BACKGROUND: Hereditary hemorrhagic telangiectasia (HHT) is a vascular disorder characterized by epistaxis, arteriovenous malformations, and telangiectases. The majority of the patients have a mutation in the coding region of the activin A receptor type II-like 1 (ACVRL1) or Endoglin (ENG) gene. However, in approximately 15% of cases, sequencing analysis and deletion/duplication testing fail to identify mutations in the coding regions of these genes. Knowing its vital role in transcription and translation control, we were prompted to investigate the 5'untranslated region (UTR) of ENG. METHODS AND RESULTS: We sequenced the 5'UTR of ENG for 154 HHT patients without mutations in ENG or ACVRL1 coding regions. We found a mutation (c.-127C > T), which is predicted to affect translation initiation and alter the reading frame of endoglin. This mutation was found in a family with linkage to the ENG, as well as in three other patients, one of which had an affected sibling with the same mutation. In vitro expression studies showed that a construct with the c.-127C > T mutation alters the translation and decreases the level of the endoglin protein. In addition, a c.-9G > A mutation was found in three patients, one of whom was homozygous for this mutation. Expression studies showed decreased protein levels suggesting that the c.-9G > A is a hypomorphic mutation. CONCLUSIONS: Our results emphasize the need for the inclusion of the 5'UTR region of ENG in clinical testing for HHT.


Subject(s)
5' Untranslated Regions/genetics , Antigens, CD/genetics , Mutation/genetics , Receptors, Cell Surface/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Adolescent , Adult , Aged , Animals , Antigens, CD/chemistry , Base Sequence , COS Cells , Child , Child, Preschool , Endoglin , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Receptors, Cell Surface/chemistry , Sequence Analysis, DNA , Transfection
16.
BMC Mol Biol ; 11: 51, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20587022

ABSTRACT

BACKGROUND: Activin receptor-like kinase 1 (ALK1) is a Transforming Growth Factor-beta (TGF-beta) receptor type I, mainly expressed in endothelial cells that plays a pivotal role in vascular remodelling and angiogenesis. Mutations in the ALK1 gene (ACVRL1) give rise to Hereditary Haemorrhagic Telangiectasia, a dominant autosomal vascular dysplasia caused by a haploinsufficiency mechanism. In spite of its patho-physiological relevance, little is known about the transcriptional regulation of ACVRL1. Here, we have studied the different origins of ACVRL1 transcription, we have analyzed in silico its 5'-proximal promoter sequence and we have characterized the role of Sp1 in the transcriptional regulation of ACVRL1. RESULTS: We have performed a 5'Rapid Amplification of cDNA Ends (5'RACE) of ACVRL1 transcripts, finding two new transcriptional origins, upstream of the one previously described, that give rise to a new exon undiscovered to date. The 5'-proximal promoter region of ACVRL1 (-1,035/+210) was analyzed in silico, finding that it lacks TATA/CAAT boxes, but contains a remarkably high number of GC-rich Sp1 consensus sites. In cells lacking Sp1, ACVRL1 promoter reporters did not present any significant transcriptional activity, whereas increasing concentrations of Sp1 triggered a dose-dependent stimulation of its transcription. Moreover, silencing Sp1 in HEK293T cells resulted in a marked decrease of ACVRL1 transcriptional activity. Chromatin immunoprecipitation assays demonstrated multiple Sp1 binding sites along the proximal promoter region of ACVRL1 in endothelial cells. Furthermore, demethylation of CpG islands, led to an increase in ACVRL1 transcription, whereas in vitro hypermethylation resulted in the abolishment of Sp1-dependent transcriptional activation of ACVRL1. CONCLUSIONS: Our results describe two new transcriptional start sites in ACVRL1 gene, and indicate that Sp1 is a key regulator of ACVRL1 transcription, providing new insights into the molecular mechanisms that contribute to the expression of ACVRL1 gene. Moreover, our data show that the methylation status of CpG islands markedly modulates the Sp1 regulation of ACVRL1 gene transcriptional activity.


Subject(s)
Activin Receptors, Type II/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , 5' Untranslated Regions , Activin Receptors, Type II/metabolism , Animals , Base Sequence , Binding Sites , Cattle , Chromatin Immunoprecipitation , CpG Islands , DNA Methylation , Dogs , Horses , Humans , Macaca mulatta , Mice , Molecular Sequence Data , Pongo , Protein Binding , Rats , Sequence Alignment , Transcription Initiation Site , Transcriptional Activation
17.
Circ Res ; 103(12): 1383-92, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18974388

ABSTRACT

Senescence of endothelial cells (ECs) may contribute to age-associated cardiovascular diseases, including atherosclerosis and hypertension. The functional and gene expression changes associated with cellular senescence are poorly understood. Here, we have analyzed the expression, during EC senescence, of 2 different isoforms (L, long; S, short) of endoglin, an auxiliary transforming growth factor (TGF)-beta receptor involved in vascular remodeling and angiogenesis. As evidenced by RT-PCR, the S/L ratio of endoglin isoforms was increased during senescence of human ECs in vitro, as well as during aging of mice in vascularized tissues. Next, the effect of S-endoglin protein on the TGF-beta receptor complex was studied. As revealed by coimmunoprecipitation assays, S-endoglin was able to interact with both TGF-beta type I receptors, ALK5 and ALK1, although the interaction with ALK5 was stronger than with ALK1. S-endoglin conferred a lower proliferation rate to ECs and behaved differently from L-endoglin in relation to TGF-beta-responsive reporters with ALK1 or ALK5 specificities, mimicking the behavior of the endothelial senescence markers Id1 and plasminogen activator inhibitor-1. In situ hybridization studies demonstrated the expression of S-endoglin in the endothelium from human arteries. Transgenic mice overexpressing S-endoglin in ECs showed hypertension, decreased hypertensive response to NO inhibition, decreased vasodilatory response to TGF-beta(1) administration, and decreased endothelial nitric oxide synthase expression in lungs and kidneys, supporting the involvement of S-endoglin in the NO-dependent vascular homeostasis. Taken together, these results suggest that S-endoglin is induced during endothelial senescence and may contribute to age-dependent vascular pathology.


Subject(s)
Antigens, CD/biosynthesis , Cellular Senescence/physiology , Endothelial Cells/physiology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Gene Expression Regulation/physiology , Receptors, Cell Surface/biosynthesis , Animals , Antigens, CD/genetics , Antigens, CD/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Endoglin , Endothelial Cells/pathology , Humans , Mice , Mice, Transgenic , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Vascular Diseases/blood , Vascular Diseases/genetics , Vascular Diseases/pathology
18.
J Cell Physiol ; 204(2): 574-84, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15702480

ABSTRACT

Transforming growth factor-beta (TGF-beta) signaling in endothelial cells is able to modulate angiogenesis and vascular remodeling, although the underlying molecular mechanisms remain poorly understood. Endoglin and ALK-1 are components of the TGF-beta receptor complex, predominantly expressed in endothelial cells, and mutations in either endoglin or ALK-1 genes are responsible for the vascular dysplasia known as hereditary hemorrhagic telangiectasia. Here we find that the extracellular and cytoplasmic domains of the auxiliary TGF-beta receptor endoglin interact with ALK-1 (a type I TGF-beta receptor). In addition, endoglin potentiates TGF-beta/ALK1 signaling, with the extracellular domain of endoglin contributing to this functional cooperation between endoglin and ALK-1. By contrast, endoglin appears to interfere with TGF-beta/ALK-5 signaling. These results suggest that the functional association of endoglin with ALK-1 is critical for the endothelial responses to TGF-beta.


Subject(s)
Activin Receptors, Type I/metabolism , Endothelial Cells/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Activin Receptors, Type II , Animals , Antigens, CD , Cell Line , Cytoplasm/metabolism , Drug Interactions , Endoglin , Extracellular Space/metabolism , Gene Expression Regulation , Humans , Inhibitor of Differentiation Protein 1 , Promoter Regions, Genetic , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Receptor, Transforming Growth Factor-beta Type I , Receptors, Cell Surface , Repressor Proteins/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transfection , Vascular Cell Adhesion Molecule-1/genetics
19.
Matrix Biol ; 22(7): 561-72, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14996436

ABSTRACT

Endoglin (CD105) is a homodimeric membrane glycoprotein, which acts as a TGF-beta coreceptor in the vasculature and plays an important role in cardiovascular development and vascular remodelling. To isolate putative genes regulated by endoglin expression, a PCR-based RNA fingerprinting technique was carried out. Myoblasts stably transfected with endoglin showed a decrease in the expression of lumican both at the RNA and protein levels. Lumican is a proteoglycan of the extracellular matrix, belonging to the SLRP (Small Leucine-Rich Repeat Proteoglycans) family. Lumican down-regulation by endoglin appeared to be controlled, at least in part, at the transcriptional level, as indicated by RT-PCR, and transient transfection experiments using a lumican promoter reporter based vector. This inverse correlation between endoglin and lumican expression was substantiated by immunohistochemical staining of vessels from human tissues. Thus, cells belonging to the high endothelia, such as tonsil, express a large amount of endoglin, and the lumican content of their matrix is considerably reduced. Conversely, in resting endothelia, such as that of large vessels, the expression of endoglin is reduced whereas the amount of lumican is greatly increased. The inverse regulation in the expression of endoglin and lumican was also evident after TGF-beta treatments since endoglin was up-regulated, whereas lumican was down-regulated by this cytokine. This report describes for the first time a relationship between endoglin and lumican expression.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Keratan Sulfate/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Antigens, CD , Cells, Cultured , Chondroitin Sulfate Proteoglycans/genetics , Down-Regulation , Endoglin , Gene Expression/drug effects , Humans , Keratan Sulfate/genetics , Lumican , Rabbits , Rats , Receptors, Cell Surface , Tissue Distribution , Transcription, Genetic/physiology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Vascular Cell Adhesion Molecule-1/genetics
20.
Anticancer Res ; 23(2B): 1189-96, 2003.
Article in English | MEDLINE | ID: mdl-12820370

ABSTRACT

The vascular endothelium participates in angiogenesis, inflammation and the immune response, which are modulated by vasoactive cytokines such as tumour necrosis factor-alpha (TNF alpha) and transforming growth factor-beta 1 (TGF beta 1). CD105 is a component of the TGF beta receptor complex and is abundantly expressed in activated/injured endothelium where it is implicated in multiple cellular processes. Up-regulation of CD105 in synovial cells of rheumatoid arthritis and psoriatic lesions implies a possible role in the pathogenesis of such inflammatory disorders. The pro-inflammatory cytokine, TNF alpha, and anti-inflammatory cytokine, TGF beta 1, regulate multiple cellular processes such as proliferation, differentiation and apoptosis. Our hypothesis is that CD105 gene expression in endothelial cells is regulated by the multifunctional cytokines TNF alpha and TGF beta 1. By using human dermal microvascular endothelial cells the present study has shown that long-term treatment with TNF alpha (0.1-5 ng/ml) elicited a concentration- and time-dependent significant suppression (over 50% reduction) in CD105 protein levels. The observations that no significant alterations in the CD105 mRNA levels or in the CD105 promoter activity were found and that the potent inhibitor of NF kappa B, PDTC, did not affect the TNF alpha action suggest that CD105 down-regulation by TNF alpha is not at the transcriptional level. In contrast to TNF alpha, TGF beta 1 significantly elevated CD105 protein and mRNA expression (approximately 2-fold increase) through activation of its promoter activity. From these data we conclude that TNF alpha and TGF beta 1 exert opposing effects on CD105 expression in human vascular endothelial cells and that CD105 is enmeshed in the network of signal pathways modulating multiple cellular functions.


Subject(s)
Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Proline/analogs & derivatives , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Antigens, CD , Blotting, Northern , Blotting, Western , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Endoglin , Endothelium, Vascular/metabolism , Genes, Reporter , Humans , Luciferases/analysis , Luciferases/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Proline/pharmacology , Promoter Regions, Genetic/drug effects , RNA, Messenger/biosynthesis , Receptors, Cell Surface , Thiocarbamates/pharmacology , Transcription, Genetic/drug effects , Transforming Growth Factor beta1 , Vascular Cell Adhesion Molecule-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...