Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Clin Med ; 11(18)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36142926

ABSTRACT

Haploinsufficiency for Endoglin (ENG) and activin A receptor type II-like I (ACVRL1/ALK1) lead to the formation of weak and abnormal vessels in hereditary hemorrhagic telangiectasia (HHT). These cause epistaxis (nosebleeds) and/or gastrointestinal blood loss. In vitro in cultured endothelial cells, tacrolimus has been shown to increase ENG and ALK1 expression. It is, therefore, a potential treatment option. We report here a proof-of-concept study in patients with HHT and severe epistaxis and/or gastrointestinal bleeding who were treated daily with orally-administered tacrolimus for twenty weeks. Twenty-five patients with HHT (11 females (44%)) and median age of 59 years were enrolled. Five patients (20%) stopped the trial prematurely-four due to (serious) adverse events ((S)AE). Twenty patients were included in further analyses. Hemoglobin levels increased during tacrolimus treatment from 6.1 (IQR 5.2-6.9) mmol/L at baseline (9.8 g/dL) to 6.7 (6.5-7.1) mmol/L (10.8 g/dL), p = 0.003. The number of blood transfusions over the twenty weeks decreased from a mean of 5.0 (±9.2) to 1.9 (±3.5), p = 0.04. In 64% of the patients, at least one AE occurred. Oral tacrolimus, thus, significantly increased hemoglobin levels and decreased blood transfusion needs, epistaxis and/or gastrointestinal bleeding in patients with HHT. However, side-effects were common. Further investigation of the potential therapeutic benefit is justified by the outcome of the study.

2.
Stem Cell Reports ; 17(7): 1536-1545, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35777360

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is a genetic disease characterized by weak blood vessels. HHT1 is caused by mutations in the ENDOGLIN (ENG) gene. Here, we generated induced pluripotent stem cells (hiPSCs) from a patient with rare mosaic HHT1 with tissues containing both mutant (ENGc.1678C>T) and normal cells, enabling derivation of isogenic diseased and healthy hiPSCs, respectively. We showed reduced ENG expression in HHT1 endothelial cells (HHT1-hiPSC-ECs), reflecting haploinsufficiency. HHT1c.1678C>T-hiPSC-ECs and the healthy isogenic control behaved similarly in two-dimensional (2D) culture, forming functionally indistinguishable vascular networks. However, when grown in 3D organ-on-chip devices under microfluidic flow, lumenized vessels formed in which defective vascular organization was evident: interaction between inner ECs and surrounding pericytes was decreased, and there was evidence for vascular leakage. Organs on chip thus revealed features of HHT in hiPSC-derived blood vessels that were not evident in conventional 2D assays.


Subject(s)
Induced Pluripotent Stem Cells , Telangiectasia, Hereditary Hemorrhagic , Activin Receptors, Type II/genetics , Endoglin/genetics , Endoglin/metabolism , Endothelial Cells/metabolism , Humans , Mutation , Telangiectasia, Hereditary Hemorrhagic/complications , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/metabolism
3.
Cardiovasc Res ; 118(5): 1262-1275, 2022 03 25.
Article in English | MEDLINE | ID: mdl-33909875

ABSTRACT

AIMS: Hepatic capillaries are lined with specialized liver sinusoidal endothelial cells (LSECs) which support macromolecule passage to hepatocytes and prevent fibrosis by keeping hepatic stellate cells (HSCs) quiescent. LSEC specialization is co-determined by transcription factors. The zinc-finger E-box-binding homeobox (Zeb)2 transcription factor is enriched in LSECs. Here, we aimed to elucidate the endothelium-specific role of Zeb2 during maintenance of the liver and in liver fibrosis. METHODS AND RESULTS: To study the role of Zeb2 in liver endothelium we generated EC-specific Zeb2 knock-out (ECKO) mice. Sequencing of liver EC RNA revealed that deficiency of Zeb2 results in prominent expression changes in angiogenesis-related genes. Accordingly, the vascular area was expanded and the presence of pillars inside ECKO liver vessels indicated that this was likely due to increased intussusceptive angiogenesis. LSEC marker expression was not profoundly affected and fenestrations were preserved upon Zeb2 deficiency. However, an increase in continuous EC markers suggested that Zeb2-deficient LSECs are more prone to dedifferentiation, a process called 'capillarization'. Changes in the endothelial expression of ligands that may be involved in HSC quiescence together with significant changes in the expression profile of HSCs showed that Zeb2 regulates LSEC-HSC communication and HSC activation. Accordingly, upon exposure to the hepatotoxin carbon tetrachloride (CCl4), livers of ECKO mice showed increased capillarization, HSC activation, and fibrosis compared to livers from wild-type littermates. The vascular maintenance and anti-fibrotic role of endothelial Zeb2 was confirmed in mice with EC-specific overexpression of Zeb2, as the latter resulted in reduced vascularity and attenuated CCl4-induced liver fibrosis. CONCLUSION: Endothelial Zeb2 preserves liver angioarchitecture and protects against liver fibrosis. Zeb2 and Zeb2-dependent genes in liver ECs may be exploited to design novel therapeutic strategies to attenuate hepatic fibrosis.


Subject(s)
Endothelial Cells , Liver Cirrhosis , Animals , Biomarkers/metabolism , Endothelial Cells/metabolism , Endothelium , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/prevention & control , Mice
4.
Int J Mol Sci ; 22(16)2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34445652

ABSTRACT

Hereditary Hemorrhagic Telangiectasia type 1 (HHT1) is an autosomal dominant inherited disease characterized by arteriovenous malformations and hemorrhage. HHT1 is caused by mutations in ENDOGLIN, which encodes an ancillary receptor for Transforming Growth Factor-ß/Bone Morphogenetic Protein-9 expressed in all vascular endothelial cells. Haploinsufficiency is widely accepted as the underlying mechanism for HHT1. However, it remains intriguing that only some, but not all, vascular beds are affected, as these causal gene mutations are present in vasculature throughout the body. Here, we have examined the endoglin expression levels in the blood vessels of multiple organs in mice and in humans. We found a positive correlation between low basal levels of endoglin and the general prevalence of clinical manifestations in selected organs. Endoglin was found to be particularly low in the skin, the earliest site of vascular lesions in HHT1, and even undetectable in the arteries and capillaries of heterozygous endoglin mice. Endoglin levels did not appear to be associated with organ-specific vascular functions. Instead, our data revealed a critical endoglin threshold compatible with the haploinsufficiency model, below which endothelial cells independent of their tissue of origin exhibited abnormal responses to Vascular Endothelial Growth Factor. Our results support the development of drugs promoting endoglin expression as potentially protective.


Subject(s)
Endoglin/physiology , Endothelium, Vascular/pathology , Mutation , Telangiectasia, Hereditary Hemorrhagic/complications , Vascular Diseases/pathology , Animals , Endothelium, Vascular/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Vascular Diseases/etiology , Vascular Diseases/metabolism
5.
Cells ; 10(5)2021 05 02.
Article in English | MEDLINE | ID: mdl-34063200

ABSTRACT

Renal microvascular rarefaction plays a pivotal role in progressive kidney disease. Therefore, modalities to visualize the microcirculation of the kidney will increase our understanding of disease mechanisms and consequently may provide new approaches for evaluating cell-based therapy. At the moment, however, clinical practice is lacking non-invasive, safe, and efficient imaging modalities to monitor renal microvascular changes over time in patients suffering from renal disease. To emphasize the importance, we summarize current knowledge of the renal microcirculation and discussed the involvement in progressive kidney disease. Moreover, an overview of available imaging techniques to uncover renal microvascular morphology, function, and behavior is presented with the associated benefits and limitations. Ultimately, the necessity to assess and investigate renal disease based on in vivo readouts with a resolution up to capillary level may provide a paradigm shift for diagnosis and therapy in the field of nephrology.


Subject(s)
Diagnostic Imaging/methods , Kidney/blood supply , Kidney/diagnostic imaging , Microcirculation , Capillaries , Cell- and Tissue-Based Therapy , Disease Progression , Endothelium, Vascular/pathology , Fibroblasts/metabolism , Humans , Kidney Diseases , Magnetic Resonance Imaging , Pericytes , Signal Transduction , Ultrasonography , X-Ray Microtomography
6.
J Vis Exp ; (171)2021 05 11.
Article in English | MEDLINE | ID: mdl-34057453

ABSTRACT

Recent advances in induced pluripotent stem cells (iPSC) and gene editing technologies enable the development of novel human cell-based disease models for phenotypic drug discovery (PDD) programs. Although these novel devices could predict the safety and efficacy of investigational drugs in humans more accurately, their development to the clinic still strongly rely on mammalian data, notably the use of mouse disease models. In parallel to human organoid or organ-on-chip disease models, the development of relevant in vitro mouse models is therefore an unmet need for evaluating direct drug efficacy and safety comparisons between species and in vivo and in vitro conditions. Here, a vascular sprouting assay that utilizes mouse embryonic stem cells differentiated into embryoid bodies (EBs) is described. Vascularized EBs cultured onto 3D-collagen gel develop new blood vessels that expand, a process called sprouting angiogenesis. This model recapitulates key features of in vivo sprouting angiogenesis-formation of blood vessels from a pre-existing vascular network-including endothelial tip cell selection, endothelial cell migration and proliferation, cell guidance, tube formation, and mural cell recruitment. It is amenable to screening for drugs and genes modulating angiogenesis and shows similarities with recently described three-dimensional (3D) vascular assays based on human iPSC technologies.


Subject(s)
Induced Pluripotent Stem Cells , Neovascularization, Physiologic , Pharmaceutical Preparations , Vascular Diseases , Animals , Cell Differentiation , Humans , Mice , Mouse Embryonic Stem Cells , Neovascularization, Pathologic
7.
J Am Soc Nephrol ; 30(10): 1886-1897, 2019 10.
Article in English | MEDLINE | ID: mdl-31308073

ABSTRACT

BACKGROUND: A glycocalyx envelope consisting of proteoglycans and adhering proteins covers endothelial cells, both the luminal and abluminal surface. We previously demonstrated that short-term loss of integrity of the luminal glycocalyx layer resulted in perturbed glomerular filtration barrier function. METHODS: To explore the role of the glycocalyx layer of the endothelial extracellular matrix in renal function, we generated mice with an endothelium-specific and inducible deletion of hyaluronan synthase 2 (Has2), the enzyme that produces hyaluronan, the main structural component of the endothelial glycocalyx layer. We also investigated the presence of endothelial hyaluronan in human kidney tissue from patients with varying degrees of diabetic nephropathy. RESULTS: Endothelial deletion of Has2 in adult mice led to substantial loss of the glycocalyx structure, and analysis of their kidneys and kidney function showed vascular destabilization, characterized by mesangiolysis, capillary ballooning, and albuminuria. This process develops over time into glomerular capillary rarefaction and glomerulosclerosis, recapitulating the phenotype of progressive human diabetic nephropathy. Using a hyaluronan-specific probe, we found loss of glomerular endothelial hyaluronan in association with lesion formation in tissue from patients with diabetic nephropathy. We also demonstrated that loss of hyaluronan, which harbors a specific binding site for angiopoietin and a key regulator of endothelial quiescence and maintenance of EC barrier function results in disturbed angiopoietin 1 Tie2. CONCLUSIONS: Endothelial loss of hyaluronan results in disturbed glomerular endothelial stabilization. Glomerular endothelial hyaluronan is a previously unrecognized key component of the extracelluar matrix that is required for glomerular structure and function and lost in diabetic nephropathy.


Subject(s)
Hyaluronic Acid/biosynthesis , Kidney Glomerulus/anatomy & histology , Kidney Glomerulus/physiology , Animals , Endothelium/metabolism , Humans , Kidney Glomerulus/metabolism , Mice , Urothelium
8.
Adv Exp Med Biol ; 1147: 215-246, 2019.
Article in English | MEDLINE | ID: mdl-31147880

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is a genetic disorder characterized by multi-systemic vascular dysplasia affecting 1 in 5000 people worldwide. Individuals with HHT suffer from many complications including nose and gastrointestinal bleeding, anemia, iron deficiency, stroke, abscess, and high-output heart failure. Identification of the causative gene mutations and the generation of animal models have revealed that decreased transforming growth factor-ß (TGF-ß)/bone morphogenetic protein (BMP) signaling and increased vascular endothelial growth factor (VEGF) signaling activity in endothelial cells are responsible for the development of the vascular malformations in HHT. Perturbations in these key pathways are thought to lead to endothelial cell activation resulting in mural cell disengagement from the endothelium. This initial instability state causes the blood vessels to response inadequately when they are exposed to angiogenic triggers resulting in excessive blood vessel growth and the formation of vascular abnormalities that are prone to bleeding. Drugs promoting blood vessel stability have been reported as effective in preclinical models and in clinical trials indicating possible interventional targets based on a normalization approach for treating HHT. Here, we will review how disturbed TGF-ß and VEGF signaling relates to blood vessel destabilization and HHT development and will discuss therapeutic opportunities based on the concept of vessel normalization to treat HHT.


Subject(s)
Pericytes , Telangiectasia, Hereditary Hemorrhagic , Animals , Endothelial Cells , Humans , Transforming Growth Factor beta , Vascular Endothelial Growth Factor A
9.
Circulation ; 138(23): 2698-2712, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30571259

ABSTRACT

BACKGROUND: Hereditary Hemorrhagic Telangiectasia type 2 (HHT2) is an inherited genetic disorder characterized by vascular malformations and hemorrhage. HHT2 results from ACVRL1 haploinsufficiency, the remaining wild-type allele being unable to contribute sufficient protein to sustain endothelial cell function. Blood vessels function normally but are prone to respond to angiogenic stimuli, leading to the development of telangiectasic lesions that can bleed. How ACVRL1 haploinsufficiency leads to pathological angiogenesis is unknown. METHODS: We took advantage of Acvrl1+/- mutant mice that exhibit HHT2 vascular lesions and focused on the neonatal retina and the airway system after Mycoplasma pulmonis infection, as physiological and pathological models of angiogenesis, respectively. We elucidated underlying disease mechanisms in vitro by generating Acvrl1+/- mouse embryonic stem cell lines that underwent sprouting angiogenesis and performed genetic complementation experiments. Finally, HHT2 plasma samples and skin biopsies were analyzed to determine whether the mechanisms evident in mice are conserved in humans. RESULTS: Acvrl1+/- retinas at postnatal day 7 showed excessive angiogenesis and numerous endothelial "tip cells" at the vascular front that displayed migratory defects. Vascular endothelial growth factor receptor 1 (VEGFR1; Flt-1) levels were reduced in Acvrl1+/- mice and HHT2 patients, suggesting similar mechanisms in humans. In sprouting angiogenesis, VEGFR1 is expressed in stalk cells to inhibit VEGFR2 (Flk-1, KDR) signaling and thus limit tip cell formation. Soluble VEGFR1 (sVEGFR1) is also secreted, creating a VEGF gradient that promotes orientated sprout migration. Acvrl1+/- embryonic stem cell lines recapitulated the vascular anomalies in Acvrl1+/- (HHT2) mice. Genetic insertion of either the membrane or soluble form of VEGFR1 into the ROSA26 locus of Acvrl1+/- embryonic stem cell lines prevented the vascular anomalies, suggesting that high VEGFR2 activity in Acvrl1+/- endothelial cells induces HHT2 vascular anomalies. To confirm our hypothesis, Acvrl1+/- mice were infected by Mycoplasma pulmonis to induce sustained airway inflammation. Infected Acvrl1+/- tracheas showed excessive angiogenesis with the formation of multiple telangiectases, vascular defects that were prevented by VEGFR2 blocking antibodies. CONCLUSIONS: Our findings demonstrate a key role of VEGFR1 in HHT2 pathogenesis and provide mechanisms explaining why HHT2 blood vessels respond abnormally to angiogenic signals. This supports the case for using anti-VEGF therapy in HHT2.


Subject(s)
Telangiectasia, Hereditary Hemorrhagic/pathology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type II , Adult , Animals , Antibodies/administration & dosage , Antibodies/immunology , Arteriovenous Malformations/etiology , Disease Models, Animal , Female , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mycoplasma pulmonis/physiology , Neovascularization, Physiologic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Retinal Vessels/physiology , Signal Transduction , Skin/pathology , Telangiectasia, Hereditary Hemorrhagic/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/immunology
10.
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
11.
Stem Cells Dev ; 26(23): 1682-1694, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28922973

ABSTRACT

Neural crest (NC) cells are a migratory, multipotent population giving rise to numerous lineages in the embryo. Their plasticity renders attractive their use in tissue engineering-based therapies, but further knowledge on their in vivo behavior is required before clinical transfer may be envisioned. We here describe the isolation and characterization of a new mouse embryonic stem (ES) line derived from Wnt1-CRE-R26 RosaTomatoTdv blastocyst and show that it displays the characteristics of typical ES cells. Further, these cells can be efficiently directed toward an NC stem cell-like phenotype as attested by concomitant expression of NC marker genes and Tomato fluorescence. As native NC progenitors, they are capable of differentiating toward typical derivative phenotypes and interacting with embryonic tissues to participate in the formation of neo-structures. Their specific fluorescence allows purification and tracking in vivo. This cellular tool should facilitate a better understanding of the mechanisms driving NC fate specification and help identify the key interactions developed within a tissue after in vivo implantation. Altogether, this novel model may provide important knowledge to optimize NC stem cell graft conditions, which are required for efficient tissue repair.


Subject(s)
Embryonic Stem Cells/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Neurogenesis , Animals , Cell Line , Cells, Cultured , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Integrases/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Neural Crest/embryology , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
12.
Arterioscler Thromb Vasc Biol ; 36(4): 707-17, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26821948

ABSTRACT

OBJECTIVE: To determine the role of Gja5 that encodes for the gap junction protein connexin40 in the generation of arteriovenous malformations in the hereditary hemorrhagic telangiectasia type 2 (HHT2) mouse model. APPROACH AND RESULTS: We identified GJA5 as a target gene of the bone morphogenetic protein-9/activin receptor-like kinase 1 signaling pathway in human aortic endothelial cells and importantly found that connexin40 levels were particularly low in a small group of patients with HHT2. We next took advantage of the Acvrl1(+/-) mutant mice that develop lesions similar to those in patients with HHT2 and generated Acvrl1(+/-); Gja5(EGFP/+) mice. Gja5 haploinsufficiency led to vasodilation of the arteries and rarefaction of the capillary bed in Acvrl1(+/-) mice. At the molecular level, we found that reduced Gja5 in Acvrl1(+/-) mice stimulated the production of reactive oxygen species, an important mediator of vessel remodeling. To normalize the altered hemodynamic forces in Acvrl1(+/-); Gja5(EGFP/+) mice, capillaries formed transient arteriovenous shunts that could develop into large malformations when exposed to environmental insults. CONCLUSIONS: We identified GJA5 as a potential modifier gene for HHT2. Our findings demonstrate that Acvrl1 haploinsufficiency combined with the effects of modifier genes that regulate vessel caliber is responsible for the heterogeneity and severity of the disease. The mouse models of HHT have led to the proposal that 3 events-heterozygosity, loss of heterozygosity, and angiogenic stimulation-are necessary for arteriovenous malformation formation. Here, we present a novel 3-step model in which pathological vessel caliber and consequent altered blood flow are necessary events for arteriovenous malformation development.


Subject(s)
Activin Receptors, Type II/metabolism , Activin Receptors, Type I/metabolism , Arteriovenous Malformations/enzymology , Connexins/metabolism , Endothelial Cells/enzymology , Retinal Vessels/enzymology , Telangiectasia, Hereditary Hemorrhagic/enzymology , Activin Receptors, Type I/genetics , Activin Receptors, Type II/genetics , Animals , Arteriovenous Malformations/genetics , Arteriovenous Malformations/pathology , Cells, Cultured , Connexins/genetics , Disease Models, Animal , Genetic Predisposition to Disease , Haploinsufficiency , Humans , Mice, Mutant Strains , Mice, Transgenic , Neovascularization, Pathologic , Phenotype , RNA Interference , Reactive Oxygen Species/metabolism , Retinal Vessels/pathology , Signal Transduction , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Transfection , Vascular Remodeling , Gap Junction alpha-5 Protein
13.
Front Genet ; 6: 37, 2015.
Article in English | MEDLINE | ID: mdl-25763012

ABSTRACT

Defective paracrine Transforming Growth Factor-ß (TGF-ß) signaling between endothelial cells and the neighboring mural cells have been thought to lead to the development of vascular lesions that are characteristic of Hereditary Hemorrhagic Telangiectasia (HHT). This review highlights recent progress in our understanding of TGF-ß signaling in mural cell recruitment and vessel stabilization and how perturbed TGF-ß signaling might contribute to defective endothelial-mural cell interaction affecting vessel functionalities. Our recent findings have provided exciting insights into the role of thalidomide, a drug that reduces both the frequency and the duration of epistaxis in individuals with HHT by targeting mural cells. These advances provide opportunities for the development of new therapies for vascular malformations.

14.
PLoS One ; 9(6): e100310, 2014.
Article in English | MEDLINE | ID: mdl-24956016

ABSTRACT

Mutations affecting transforming growth factor-beta (TGF-ß) superfamily receptors, activin receptor-like kinase (ALK)-1, and endoglin (ENG) occur in patients with pulmonary arterial hypertension (PAH). To determine whether the TGF-ß/ALK1/ENG pathway was involved in PAH, we investigated pulmonary TGF-ß, ALK1, ALK5, and ENG expressions in human lung tissue and cultured pulmonary-artery smooth-muscle-cells (PA-SMCs) and pulmonary endothelial cells (PECs) from 14 patients with idiopathic PAH (iPAH) and 15 controls. Seeing that ENG was highly expressed in PEC, we assessed the effects of TGF-ß on Smad1/5/8 and Smad2/3 activation and on growth factor production by the cells. Finally, we studied the consequence of ENG deficiency on the chronic hypoxic-PH development by measuring right ventricular (RV) systolic pressure (RVSP), RV hypertrophy, and pulmonary arteriolar remodeling in ENG-deficient (Eng+/-) and wild-type (Eng+/+) mice. We also evaluated the pulmonary blood vessel density, macrophage infiltration, and cytokine expression in the lungs of the animals. Compared to controls, iPAH patients had higher serum and pulmonary TGF-ß levels and increased ALK1 and ENG expressions in lung tissue, predominantly in PECs. Incubation of the cells with TGF-ß led to Smad1/5/8 phosphorylation and to a production of FGF2, PDGFb and endothelin-inducing PA-SMC growth. Endoglin deficiency protected mice from hypoxic PH. As compared to wild-type, Eng+/- mice had a lower pulmonary vessel density, and no change in macrophage infiltration after exposure to chronic hypoxia despite the higher pulmonary expressions of interleukin-6 and monocyte chemoattractant protein-1. The TGF-ß/ALK1/ENG signaling pathway plays a key role in iPAH and experimental hypoxic PH via a direct effect on PECs leading to production of growth factors and inflammatory cytokines involved in the pathogenesis of PAH.


Subject(s)
Activin Receptors, Type II/metabolism , Endothelium, Vascular/metabolism , Familial Primary Pulmonary Hypertension/metabolism , Hypertension, Pulmonary/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Pulmonary Artery/metabolism , Transforming Growth Factor beta/metabolism , Activin Receptors, Type II/genetics , Animals , Blotting, Western , Case-Control Studies , Cell Proliferation , Cells, Cultured , Endoglin , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Familial Primary Pulmonary Hypertension/genetics , Familial Primary Pulmonary Hypertension/pathology , Female , Follow-Up Studies , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Prognosis , Pulmonary Artery/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transforming Growth Factor beta/genetics
15.
PLoS One ; 9(1): e86273, 2014.
Article in English | MEDLINE | ID: mdl-24489709

ABSTRACT

ENDOGLIN (ENG) is a co-receptor for transforming growth factor-ß (TGF-ß) family members that is highly expressed in endothelial cells and has a critical function in the development of the vascular system. Mutations in Eng are associated with the vascular disease known as hereditary hemorrhagic telangiectasia type l. Using mouse embryonic stem cells we observed that angiogenic factors, including vascular endothelial growth factor (VEGF), induce vasculogenesis in embryoid bodies even when Eng deficient cells or cells depleted of Eng using shRNA are used. However, ENG is required for the stem cell-derived endothelial cells to organize effectively into tubular structures. Consistent with this finding, fetal metatarsals isolated from E17.5 Eng heterozygous mouse embryos showed reduced VEGF-induced vascular network formation. Moreover, shRNA-mediated depletion and pharmacological inhibition of ENG in human umbilical vein cells mitigated VEGF-induced angiogenesis. In summary, we demonstrate that ENG is required for efficient VEGF-induced angiogenesis.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Endoglin , Flow Cytometry , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Neovascularization, Physiologic/genetics
16.
Eur J Dermatol ; 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23608678

ABSTRACT

The (pro)renin receptor (PRR) is a component of the renin-angiotensin system (RAS) that is believed to control blood pressure and salt homeostasis in mammals by favouring tissue activation of RAS. Genetic studies have recently provided novel and exciting insights into how PRR regulates embryonic development in Drosophila and Xenopus through RAS independent functions. By interacting with the H+ vacuolar ATPase (V-ATPase), PRR modulates Wnt signalling pathways. Signalling by Wnt family members governs many aspects of embryonic development and tissue homeostasis. In particular, in mammals, Wnt signalling plays essential roles in the control of stem cell fate decision and lineage commitment in tissues with high self-renewal capacities such as the intestine and the skin, in which we have found PRR to be strongly expressed. Here, we review recent data on how PRR is thought to function during development and place it in the broader context of wnt signalling in skin in general.

17.
Nat Med ; 16(4): 420-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20364125

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an inherited disorder characterized by vascular malformations. Many affected individuals develop recurrent nosebleeds, which can severely affect their quality of life and are clinically difficult to treat. We report here that treatment with thalidomide reduced the severity and frequency of nosebleeds (epistaxis) in the majority of a small group of subjects with HHT tested. The blood hemoglobin levels of the treated individuals rose as a result of reduced hemorrhage and enhanced blood vessel stabilization. In mice heterozygous for a null mutation in the Eng gene (encoding endoglin), an experimental model of HHT, thalidomide treatment stimulated mural cell coverage and thus rescued vessel wall defects. Thalidomide treatment increased platelet-derived growth factor-B (PDGF-B) expression in endothelial cells and stimulated mural cell activation. The effects of thalidomide treatment were partially reversed by pharmacological or genetic interference with PDGF signaling from endothelial cells to pericytes. Biopsies of nasal epithelium from individuals with HHT treated or not with thalidomide showed that similar mechanisms may explain the effects of thalidomide treatment in humans. Our findings demonstrate the ability of thalidomide to induce vessel maturation, which may be useful as a therapeutic strategy for the treatment of vascular malformations.


Subject(s)
Blood Vessels/drug effects , Epistaxis/drug therapy , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Thalidomide/therapeutic use , Aged , Animals , Blood Vessels/growth & development , Blood Vessels/physiology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/physiology , Hemoglobins/analysis , Humans , Mice , Mice, Mutant Strains , Middle Aged , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins c-sis/biosynthesis , Thalidomide/pharmacology
18.
Stroke ; 40(2): 439-47, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074480

ABSTRACT

BACKGROUND AND PURPOSE: Unstable atherosclerotic plaques are characterized by increased macrophages and reduced smooth muscle cells (SMCs) and collagen content. Endoglin, an accessory transforming growth factor-beta (TGFbeta) receptor, is a modulator of TGFbeta signaling recently found to be expressed on SMCs in atherosclerotic plaques. Its function in plaque SMCs and plaque development is unknown. Early growth response-1 (EGR-1), a transcription factor downstream of TGFbeta, stimulates SMC proliferation and collagen synthesis. In atherosclerotic lesions, it is mainly expressed by SMCs. Therefore, we studied the TGFbeta, endoglin, and EGR-1 pathway in advanced atherosclerotic plaques in relation to plaque phenotype. METHODS: Human carotid atherosclerotic plaques (n=103) were collected from patients undergoing carotid endarterectomy. Histologically, plaques were analyzed for plaque characteristics, ie, collagen, macrophage and SMC content, and intraplaque thrombus. Intraplaque endoglin, pSmad (indicative for TGFbeta signaling), EGR-1, and TGFbeta levels were analyzed using Western blots and enzyme-linked immunosorbent assays, respectively. RESULTS: Higher endoglin and EGR-1 protein levels correlated positively with increased plaque collagen levels, increased smooth muscle cell content, and decreased intraplaque thrombi as well as TGFbeta signaling (pSmad). Although EGR-1 overexpression in vitro stimulated collagen synthesis, inhibiting endoglin resulted in lower EGR-1 levels, decreased SMC proliferation, and decreased collagen content. CONCLUSIONS: TGFbeta in human atherosclerotic plaques is active and signals through the TGFbeta/Smad pathway. For the first time, we show a strong association between endoglin and EGR-1, increased collagen and SMCs expression, decreased levels of intraplaque thrombosis, and a stable plaque phenotype.


Subject(s)
Antigens, CD/biosynthesis , Early Growth Response Protein 1/biosynthesis , Intracranial Arteriosclerosis/metabolism , Receptors, Cell Surface/biosynthesis , Signal Transduction/physiology , Transforming Growth Factor beta/biosynthesis , Aged , Blotting, Western , Cell Line , Cell Proliferation , Collagen/biosynthesis , Collagen/genetics , Collagen Type I , Endoglin , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Profiling , Genes, Reporter/genetics , Humans , Immunohistochemistry , Intracranial Arteriosclerosis/physiopathology , Male , Matrix Metalloproteinases/biosynthesis , Myocytes, Smooth Muscle/physiology , Phenotype , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
19.
Circ Res ; 103(4): 432-40, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18617693

ABSTRACT

Apelin has been identified as the endogenous ligand of the human orphan G protein-coupled receptor APJ. This peptide exerts a variety of cardiovascular effects and particularly acts as an activator of angiogenesis. Importantly, hypoxia has been reported to regulate apelin expression but the molecular mechanism underlying hypoxia-induced apelin expression and the relationship with the physiological response of the apelin/APJ system are still not established. Here, we demonstrate that apelin expression is induced by hypoxia in cultured endothelial and vascular smooth muscle cells as well as in lung from mice exposed to acute hypoxia. Transient transfection experiments show that hypoxia-inducible transcriptional activation of apelin requires an intact hypoxia-responsive element (+813/+826) located within the first intron of the human apelin gene. Chromatin immunoprecipitation assay reveals that hypoxia-inducible factor-1alpha binds to the endogenous hypoxia-responsive element site of the apelin gene. Moreover, overexpression of hypoxia-inducible factor-1alpha increases the transcriptional activity of a reporter construct containing this hypoxia-responsive element, whereas small interfering RNA-mediated hypoxia-inducible factor-1alpha knockdown abolishes hypoxia-induced apelin expression. Finally, microinterfering RNA-mediated apelin or APJ receptor knockdown inhibits both hypoxia-induced endothelial cell proliferation in vitro and hypoxia-induced vessel regeneration in the caudal fin regeneration of Fli-1 transgenic zebrafish. The hypoxia-induced apelin expression may, thus, provide a new mechanism involved in adaptive physiological and pathophysiological response of vascular cells to low oxygen level.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation , Endothelium, Vascular/metabolism , Hypoxia/metabolism , Muscle, Smooth, Vascular/metabolism , Neovascularization, Physiologic/physiology , Regeneration/immunology , Adipokines , Amino Acid Sequence , Animals , Apelin , Carrier Proteins/genetics , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Endothelium, Vascular/cytology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Transfection , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
Stem Cells ; 26(9): 2257-65, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18599809

ABSTRACT

Defined growth conditions are essential for many applications of human embryonic stem cells (hESC). Most defined media are presently used in combination with Matrigel, a partially defined extracellular matrix (ECM) extract from mouse sarcoma. Here, we defined ECM requirements of hESC by analyzing integrin expression and ECM production and determined integrin function using blocking antibodies. hESC expressed all major ECM proteins and corresponding integrins. We then systematically replaced Matrigel with defined medium supplements and ECM proteins. Cells attached efficiently to natural human vitronectin, fibronectin, and Matrigel but poorly to laminin + entactin and collagen IV. Integrin-blocking antibodies demonstrated that alphaVbeta5 integrins mediated adhesion to vitronectin, alpha5beta1 mediated adhesion to fibronectin, and alpha6beta1 mediated adhesion to laminin + entactin. Fibronectin in feeder cell-conditioned medium partially supported growth on all natural matrices, but in defined, nonconditioned medium only Matrigel or (natural and recombinant) vitronectin was effective. Recombinant vitronectin was the only defined functional alternative to Matrigel, supporting sustained self-renewal and pluripotency in three independent hESC lines.


Subject(s)
Embryonic Stem Cells/cytology , Receptors, Vitronectin/physiology , Vitronectin/pharmacology , Animals , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Collagen , Culture Media , Drug Combinations , Embryonic Stem Cells/metabolism , Extracellular Matrix Proteins/biosynthesis , Humans , Laminin , Mice , Proteoglycans , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...