Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
JCI Insight ; 8(5)2023 03 08.
Article En | MEDLINE | ID: mdl-36883566

The adult mammalian heart has limited regenerative capacity, while the neonatal heart fully regenerates during the first week of life. Postnatal regeneration is mainly driven by proliferation of preexisting cardiomyocytes and supported by proregenerative macrophages and angiogenesis. Although the process of regeneration has been well studied in the neonatal mouse, the molecular mechanisms that define the switch between regenerative and nonregenerative cardiomyocytes are not well understood. Here, using in vivo and in vitro approaches, we identified the lncRNA Malat1 as a key player in postnatal cardiac regeneration. Malat1 deletion prevented heart regeneration in mice after myocardial infarction on postnatal day 3 associated with a decline in cardiomyocyte proliferation and reparative angiogenesis. Interestingly, Malat1 deficiency increased cardiomyocyte binucleation even in the absence of cardiac injury. Cardiomyocyte-specific deletion of Malat1 was sufficient to block regeneration, supporting a critical role of Malat1 in regulating cardiomyocyte proliferation and binucleation, a landmark of mature nonregenerative cardiomyocytes. In vitro, Malat1 deficiency induced binucleation and the expression of a maturation gene program. Finally, the loss of hnRNP U, an interaction partner of Malat1, induced similar features in vitro, suggesting that Malat1 regulates cardiomyocyte proliferation and binucleation by hnRNP U to control the regenerative window in the heart.


Heart , Heterogeneous-Nuclear Ribonucleoprotein U , Myocardial Infarction , Myocytes, Cardiac , RNA, Long Noncoding , Regeneration , Animals , Mice , Heart/physiology , Heart/physiopathology , Heart Injuries/genetics , Heart Injuries/metabolism , Heart Injuries/physiopathology , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Macrophages/metabolism , Macrophages/physiology , Mammals , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Regeneration/genetics , Regeneration/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
2.
Nat Commun ; 12(1): 681, 2021 01 29.
Article En | MEDLINE | ID: mdl-33514719

Endothelial cells play a critical role in the adaptation of tissues to injury. Tissue ischemia induced by infarction leads to profound changes in endothelial cell functions and can induce transition to a mesenchymal state. Here we explore the kinetics and individual cellular responses of endothelial cells after myocardial infarction by using single cell RNA sequencing. This study demonstrates a time dependent switch in endothelial cell proliferation and inflammation associated with transient changes in metabolic gene signatures. Trajectory analysis reveals that the majority of endothelial cells 3 to 7 days after myocardial infarction acquire a transient state, characterized by mesenchymal gene expression, which returns to baseline 14 days after injury. Lineage tracing, using the Cdh5-CreERT2;mT/mG mice followed by single cell RNA sequencing, confirms the transient mesenchymal transition and reveals additional hypoxic and inflammatory signatures of endothelial cells during early and late states after injury. These data suggest that endothelial cells undergo a transient mes-enchymal activation concomitant with a metabolic adaptation within the first days after myocardial infarction but do not acquire a long-term mesenchymal fate. This mesenchymal activation may facilitate endothelial cell migration and clonal expansion to regenerate the vascular network.


Endothelium/pathology , Epithelial-Mesenchymal Transition/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Animals , Cell Movement/genetics , Cell Plasticity/genetics , Cell Proliferation/genetics , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Endothelium/cytology , Genes, Reporter/genetics , Human Umbilical Vein Endothelial Cells , Humans , Luminescent Proteins/genetics , Male , Mice , Mice, Transgenic , Myocardium/cytology , RNA-Seq , Single-Cell Analysis
3.
Haematologica ; 105(1): 136-147, 2020 01.
Article En | MEDLINE | ID: mdl-31018977

The endosteal bone marrow niche and vascular endothelial cells provide sanctuaries for leukemic cells. In murine chronic myeloid leukemia (CML) CD44 on leukemia cells and E-selectin on bone marrow endothelium are essential mediators for the engraftment of leukemic stem cells. We hypothesized that non-adhesion of CML-initiating cells to E-selectin on the bone marrow endothelium may lead to superior eradication of leukemic stem cells in CML after treatment with imatinib than imatinib alone. Indeed, here we show that treatment with the E-selectin inhibitor GMI-1271 in combination with imatinib prolongs survival of mice with CML via decreased contact time of leukemia cells with bone marrow endothelium. Non-adhesion of BCR-ABL1+ cells leads to an increase of cell cycle progression and an increase of expression of the hematopoietic transcription factor and proto-oncogene Scl/Tal1 in leukemia-initiating cells. We implicate SCL/TAL1 as an indirect phosphorylation target of BCR-ABL1 and as a negative transcriptional regulator of CD44 expression. We show that increased SCL/TAL1 expression is associated with improved outcome in human CML. These data demonstrate the BCR-ABL1-specific, cell-intrinsic pathways leading to altered interactions with the vascular niche via the modulation of adhesion molecules - which could be exploited therapeutically in the future.


Fusion Proteins, bcr-abl , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Animals , Bone Marrow , E-Selectin/genetics , Endothelial Cells , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Proto-Oncogene Mas , T-Cell Acute Lymphocytic Leukemia Protein 1
4.
Kidney Int ; 95(3): 540-562, 2019 03.
Article En | MEDLINE | ID: mdl-30712922

Biglycan, a small leucine-rich proteoglycan, acts as a danger signal and is classically thought to promote macrophage recruitment via Toll-like receptors (TLR) 2 and 4. We have recently shown that biglycan signaling through TLR 2/4 and the CD14 co-receptor regulates inflammation, suggesting that TLR co-receptors may determine whether biglycan-TLR signaling is pro- or anti-inflammatory. Here, we sought to identify other co-receptors and characterize their impact on biglycan-TLR signaling. We found a marked increase in the number of autophagic macrophages in mice stably overexpressing soluble biglycan. In vitro, stimulation of murine macrophages with biglycan triggered autophagosome formation and enhanced the flux of autophagy markers. Soluble biglycan also promoted autophagy in human peripheral blood macrophages. Using macrophages from mice lacking TLR2 and/or TLR4, CD14, or CD44, we demonstrated that the pro-autophagy signal required TLR4 interaction with CD44, a receptor involved in adhesion, migration, lymphocyte activation, and angiogenesis. In vivo, transient overexpression of circulating biglycan at the onset of renal ischemia/reperfusion injury (IRI) enhanced M1 macrophage recruitment into the kidneys of Cd44+/+ and Cd44-/- mice but not Cd14-/- mice. The biglycan-CD44 interaction increased M1 autophagy and the number of renal M2 macrophages and reduced tubular damage following IRI. Thus, CD44 is a novel signaling co-receptor for biglycan, an interaction that is required for TLR4-CD44-dependent pro-autophagic activity in macrophages. Interfering with the interaction between biglycan and specific TLR co-receptors could represent a promising therapeutic intervention to curtail kidney inflammation and damage.


Acute Kidney Injury/immunology , Biglycan/metabolism , Hyaluronan Receptors/metabolism , Macrophages/immunology , Reperfusion Injury/immunology , Acute Kidney Injury/pathology , Animals , Autophagosomes/immunology , Autophagosomes/metabolism , Autophagy/immunology , Biglycan/genetics , Biglycan/immunology , Cells, Cultured , Disease Models, Animal , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Kidney Tubules/blood supply , Kidney Tubules/immunology , Kidney Tubules/pathology , Macrophage Activation , Mice , Mice, Knockout , Primary Cell Culture , Reperfusion Injury/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
5.
Circ Res ; 122(5): 670-677, 2018 03 02.
Article En | MEDLINE | ID: mdl-29358229

RATIONALE: Vascularization is critical to maintain organ function. Although many molecular pathways were shown to control vessel growth, the genuine process of capillary formation under different conditions is unclear. OBJECTIVE: Here, we elucidated whether clonal expansion contributes to vessel growth by using Confetti mice for genetic tracing of clonally expanding endothelial cells (ECs). METHODS AND RESULTS: In postnatal retina angiogenesis, we predominantly observed random distribution of fluorescence labeled ECs indicative of random integration or cell mixing. However, in models of pathophysiological angiogenesis (retinopathy of prematurity), as well as ischemia-induced angiogenesis in limbs and hearts, clonally expanded ECs were significantly more abundant (≤69%). Inhibition of VEGFR2 (vascular endothelial growth factor receptor 2) reduced clonal expansion after ischemia. To determine the mechanism underlying clonal expansion in vivo, we assessed gene expression specifically in clonally expanded ECs selected by laser capture microscopy. Clonally expanded ECs showed an enrichment of genes involved in endothelial-to-mesenchymal transition. Moreover, hypoxia-induced clonal expansion and endothelial-to-mesenchymal transition in ECs in vitro suggesting that hypoxia-enhanced endothelial-to-mesenchymal transition might contribute to vessel growth under ischemia. CONCLUSIONS: Our data suggest that neovascularization after ischemia is partially mediated by clonal expansion of ECs. Identification of the pathways that control clonal expansion may provide novel tools to augment therapeutic neovascularization or treat pathological angiogenesis.


Cell Lineage , Endothelium, Vascular/cytology , Neovascularization, Physiologic , Retinal Vessels/physiology , Retinopathy of Prematurity/pathology , Transcriptome , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Retinal Vessels/cytology , Retinal Vessels/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Cardiovasc Res ; 113(6): 681-691, 2017 May 01.
Article En | MEDLINE | ID: mdl-28453731

AIMS: Vessel maturation involves the recruitment of mural cells such as pericytes and smooth muscle cells. Laminar shear stress is a major trigger for vessel maturation, but the molecular mechanisms by which shear stress affects recruitment of pericytes are unclear. MicroRNAs (miRs) are small non-coding RNAs, which post-transcriptionally control gene expression. The aim of the present study was to unveil the mechanism by which shear stress-regulated microRNAs contribute to vessel maturation. METHODS AND RESULTS: Here, we show that laminar shear stress increased miR-27a and miR-27b expression in vitro and in ex vivo in mouse femoral artery explants. Overexpression of miR-27b in endothelial cells increased pericyte adhesion and pericyte recruitment in vitro. In vitro barrier function of endothelial-pericyte co-cultures was augmented by miR-27b overexpression, whereas inhibition of miR-27a/b reduced adhesion and pericyte coverage and decreased barrier functions. In vivo, pharmacological inhibition of miR-27a/b by locked nucleic acid antisense oligonucleotides significantly reduced pericyte coverage and increased water content in the murine uterus. MiR-27b overexpression repressed semaphorins (SEMA), which mediate repulsive signals, and the vessel destabilizing human but not mouse Angiopoietin-2 (Ang-2). Silencing of SEMA6A and SEMA6D rescued the reduced pericyte adhesion by miR-27 inhibition. Furthermore, inhibition of SEMA6D increased barrier function of an endothelial-pericyte co-culture in vitro. CONCLUSION: The present study demonstrates for the first time that shear stress-regulated miR-27b promotes the interaction of endothelial cells with pericytes, partly by repressing SEMA6A and SEMA6D.


Brain/blood supply , Cell Communication , Cell Movement , Endothelial Cells/metabolism , Mechanotransduction, Cellular , Microvessels/metabolism , Pericytes/metabolism , Semaphorins/metabolism , Animals , Cells, Cultured , Coculture Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Interference , Semaphorins/genetics , Stress, Mechanical , Transfection
7.
Proc Natl Acad Sci U S A ; 114(15): 3993-3998, 2017 04 11.
Article En | MEDLINE | ID: mdl-28348240

Endothelial cells (ECs) not only are important for oxygen delivery but also act as a paracrine source for signals that determine the balance between tissue regeneration and fibrosis. Here we show that genetic inactivation of flow-induced transcription factor Krüppel-like factor 2 (KLF2) in ECs results in reduced liver damage and augmentation of hepatocyte proliferation after chronic liver injury by treatment with carbon tetrachloride (CCl4). Serum levels of GLDH3 and ALT were significantly reduced in CCl4-treated EC-specific KLF2-deficient mice. In contrast, transgenic overexpression of KLF2 in liver sinusoidal ECs reduced hepatocyte proliferation. KLF2 induced activin A expression and secretion from endothelial cells in vitro and in vivo, which inhibited hepatocyte proliferation. However, loss or gain of KLF2 expression did not change capillary density and liver fibrosis, but significantly affected hepatocyte proliferation. Taken together, the data demonstrate that KLF2 induces an antiproliferative secretome, including activin A, which attenuates liver regeneration.


Activins/metabolism , Kruppel-Like Transcription Factors/genetics , Liver Regeneration/physiology , Liver/cytology , Activins/genetics , Animals , Carbon Tetrachloride/toxicity , Cell Proliferation , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Endothelial Cells/metabolism , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/physiology , Human Umbilical Vein Endothelial Cells , Humans , Kruppel-Like Transcription Factors/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice, Transgenic
8.
Nat Med ; 22(10): 1140-1150, 2016 10.
Article En | MEDLINE | ID: mdl-27595325

Adenosine-to-inosine (A-to-I) RNA editing, which is catalyzed by a family of adenosine deaminase acting on RNA (ADAR) enzymes, is important in the epitranscriptomic regulation of RNA metabolism. However, the role of A-to-I RNA editing in vascular disease is unknown. Here we show that cathepsin S mRNA (CTSS), which encodes a cysteine protease associated with angiogenesis and atherosclerosis, is highly edited in human endothelial cells. The 3' untranslated region (3' UTR) of the CTSS transcript contains two inverted repeats, the AluJo and AluSx+ regions, which form a long stem-loop structure that is recognized by ADAR1 as a substrate for editing. RNA editing enables the recruitment of the stabilizing RNA-binding protein human antigen R (HuR; encoded by ELAVL1) to the 3' UTR of the CTSS transcript, thereby controlling CTSS mRNA stability and expression. In endothelial cells, ADAR1 overexpression or treatment of cells with hypoxia or with the inflammatory cytokines interferon-γ and tumor-necrosis-factor-α induces CTSS RNA editing and consequently increases cathepsin S expression. ADAR1 levels and the extent of CTSS RNA editing are associated with changes in cathepsin S levels in patients with atherosclerotic vascular diseases, including subclinical atherosclerosis, coronary artery disease, aortic aneurysms and advanced carotid atherosclerotic disease. These results reveal a previously unrecognized role of RNA editing in gene expression in human atherosclerotic vascular diseases.


Adenosine Deaminase/genetics , Atherosclerosis/genetics , Cathepsins/genetics , ELAV-Like Protein 1/genetics , RNA Editing/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , 3' Untranslated Regions , Adenosine/metabolism , Adult , Aged , Aged, 80 and over , Aortic Aneurysm/genetics , Carotid Artery Diseases/genetics , Coronary Artery Disease/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation , Gene Knock-In Techniques , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia/genetics , Immunoblotting , Inosine/metabolism , Interferon-gamma/pharmacology , Male , Middle Aged , RNA Editing/drug effects , RNA Processing, Post-Transcriptional/drug effects , RNA Processing, Post-Transcriptional/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Tumor Necrosis Factor-alpha/pharmacology
9.
FEBS Lett ; 589(20 Pt B): 3182-8, 2015 Oct 07.
Article En | MEDLINE | ID: mdl-26348397

By transporting regulatory RNAs like microRNAs, extracellular vesicles provide a novel layer of intercellular gene regulation. However, the underlying secretory pathways and the mechanisms of cargo selection are poorly understood. Rab GTPases are central coordinators of membrane trafficking with distinct members of this family being responsible for specific transport pathways. Here we identified a vesicular export mechanism for miR-143, induced by the shear stress responsive transcription factor KLF2, and demonstrate its dependency on Rab7a/Rab27b in endothelial cells.


Extracellular Vesicles/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/metabolism , rab GTP-Binding Proteins/metabolism , Blotting, Western , Cells, Cultured , Extracellular Vesicles/genetics , Gene Expression , HeLa Cells , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/genetics , Microscopy, Confocal , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stress, Mechanical , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
10.
Arterioscler Thromb Vasc Biol ; 35(1): 137-45, 2015 Jan.
Article En | MEDLINE | ID: mdl-25359860

OBJECTIVE: Cellular metabolism was recently shown to regulate endothelial cell phenotype profoundly. Whether the atheroprotective biomechanical stimulus elicited by laminar shear stress modulates endothelial cell metabolism is not known. APPROACH AND RESULTS: Here, we show that laminar flow exposure reduced glucose uptake and mitochondrial content in endothelium. Shear stress-mediated reduction of endothelial metabolism was reversed by silencing the flow-sensitive transcription factor Krüppel-like factor 2 (KLF2). Endothelial-specific deletion of KLF2 in mice induced glucose uptake in endothelial cells of perfused hearts. KLF2 overexpression recapitulates the inhibitory effects on endothelial glycolysis elicited by laminar flow, as measured by Seahorse flux analysis and glucose uptake measurements. RNA sequencing showed that shear stress reduced the expression of key glycolytic enzymes, such as 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase-3 (PFKFB3), phosphofructokinase-1, and hexokinase 2 in a KLF2-dependent manner. Moreover, KLF2 represses PFKFB3 promoter activity. PFKFB3 knockdown reduced glycolysis, and overexpression increased glycolysis and partially reversed the KLF2-mediated reduction in glycolysis. Furthermore, PFKFB3 overexpression reversed KLF2-mediated reduction in angiogenic sprouting and network formation. CONCLUSIONS: Our data demonstrate that shear stress-mediated repression of endothelial cell metabolism via KLF2 and PFKFB3 controls endothelial cell phenotype.


Endothelial Cells/enzymology , Energy Metabolism , Kruppel-Like Transcription Factors/metabolism , Mechanotransduction, Cellular , Phosphofructokinase-2/metabolism , Animals , Biomechanical Phenomena , Cells, Cultured , Down-Regulation , Glucose/metabolism , Glycolysis , Human Umbilical Vein Endothelial Cells/enzymology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice, Knockout , Mitochondria/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Phenotype , Phosphofructokinase-2/genetics , Promoter Regions, Genetic , RNA Interference , Regional Blood Flow , Stress, Mechanical , Time Factors , Transfection
12.
Circ Res ; 114(9): 1389-97, 2014 Apr 25.
Article En | MEDLINE | ID: mdl-24602777

RATIONALE: The human genome harbors a large number of sequences encoding for RNAs that are not translated but control cellular functions by distinct mechanisms. The expression and function of the longer transcripts namely the long noncoding RNAs in the vasculature are largely unknown. OBJECTIVE: Here, we characterized the expression of long noncoding RNAs in human endothelial cells and elucidated the function of the highly expressed metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). METHODS AND RESULTS: Endothelial cells of different origin express relative high levels of the conserved long noncoding RNAs MALAT1, taurine upregulated gene 1 (TUG1), maternally expressed 3 (MEG3), linc00657, and linc00493. MALAT1 was significantly increased by hypoxia and controls a phenotypic switch in endothelial cells. Silencing of MALAT1 by small interfering RNAs or GapmeRs induced a promigratory response and increased basal sprouting and migration, whereas proliferation of endothelial cells was inhibited. When angiogenesis was further stimulated by vascular endothelial growth factor, MALAT1 small interfering RNAs induced discontinuous sprouts indicative of defective proliferation of stalk cells. In vivo studies confirmed that genetic ablation of MALAT1 inhibited proliferation of endothelial cells and reduced neonatal retina vascularization. Pharmacological inhibition of MALAT1 by GapmeRs reduced blood flow recovery and capillary density after hindlimb ischemia. Gene expression profiling followed by confirmatory quantitative reverse transcriptase-polymerase chain reaction demonstrated that silencing of MALAT1 impaired the expression of various cell cycle regulators. CONCLUSIONS: Silencing of MALAT1 tips the balance from a proliferative to a migratory endothelial cell phenotype in vitro, and its genetic deletion or pharmacological inhibition reduces vascular growth in vivo.


Endothelial Cells/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , RNA, Long Noncoding/metabolism , Retinal Neovascularization/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/genetics , Ischemia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Oligonucleotides/genetics , Oligonucleotides/metabolism , RNA Interference , RNA, Long Noncoding/genetics , Retinal Neovascularization/genetics , Retinal Neovascularization/physiopathology , Signal Transduction , Transfection
13.
Basic Res Cardiol ; 109(2): 404, 2014 Mar.
Article En | MEDLINE | ID: mdl-24522833

ß1-Integrins are essential for angiogenesis. The mechanisms regulating integrin function in endothelial cells (EC) and their contribution to angiogenesis remain elusive. Brag2 is a guanine nucleotide exchange factor for the small Arf-GTPases Arf5 and Arf6. The role of Brag2 in EC and angiogenesis and the underlying molecular mechanisms remain unclear. siRNA-mediated Brag2-silencing reduced EC angiogenic sprouting and migration. Brag2-siRNA transfection differentially affected α5ß1- and αVß3-integrin function: specifically, Brag2-silencing increased focal/fibrillar adhesions and adhesion on ß1-integrin ligands (fibronectin and collagen), while reducing the adhesion on the αVß3-integrin ligand, vitronectin. Consistent with these results, Brag2-silencing enhanced surface expression of α5ß1-integrin, while reducing surface expression of αVß3-integrin. Mechanistically, Brag2-mediated αVß3-integrin-recycling and ß1-integrin endocytosis and specifically of the active/matrix-bound α5ß1-integrin present in fibrillar/focal adhesions (FA), suggesting that Brag2 contributes to the disassembly of FA via ß1-integrin endocytosis. Arf5 and Arf6 are promoting downstream of Brag2 angiogenic sprouting, ß1-integrin endocytosis and the regulation of FA. In vivo silencing of the Brag2-orthologues in zebrafish embryos using morpholinos perturbed vascular development. Furthermore, in vivo intravitreal injection of plasmids containing Brag2-shRNA reduced pathological ischemia-induced retinal and choroidal neovascularization. These data reveal that Brag2 is essential for developmental and pathological angiogenesis by promoting EC sprouting through regulation of adhesion by mediating ß1-integrin internalization and link for the first time the process of ß1-integrin endocytosis with angiogenesis.


Cell Adhesion/physiology , Guanine Nucleotide Exchange Factors/genetics , Integrin beta1/metabolism , Integrin beta3/metabolism , Neovascularization, Pathologic/physiopathology , Retinopathy of Prematurity/physiopathology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Animals, Genetically Modified , COS Cells , Cell Movement/physiology , Chlorocebus aethiops , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/physiopathology , Disease Models, Animal , Guanine Nucleotide Exchange Factors/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , RNA, Small Interfering/genetics , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
14.
Circ Res ; 113(7): 856-62, 2013 Sep 13.
Article En | MEDLINE | ID: mdl-23856522

RATIONALE: The developmental role of the H3K27 demethylases Jmjd3, especially its epigenetic regulation at target genes in response to upstream developmental signaling, is unclear. OBJECTIVE: To determine the role of Jmjd3 during mesoderm and cardiovascular lineage commitment. METHODS AND RESULTS: Ablation of Jmjd3 in mouse embryonic stem cells does not affect the maintenance of pluripotency and self-renewal but compromised mesoderm and subsequent endothelial and cardiac differentiation. Jmjd3 reduces H3K27me3 marks at the Brachyury promoter and facilitates the recruitment of ß-catenin, which is critical for Wnt signal-induced mesoderm differentiation. CONCLUSIONS: These data demonstrate that Jmjd3 is required for mesoderm differentiation and cardiovascular lineage commitment.


Cell Differentiation , Embryonic Stem Cells/metabolism , Endothelium, Vascular/cytology , Jumonji Domain-Containing Histone Demethylases/metabolism , Mesoderm/cytology , Myocytes, Cardiac/cytology , Animals , Cell Line , Cell Lineage , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Fetal Proteins/genetics , Fetal Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Mesoderm/metabolism , Mice , Mutation , Promoter Regions, Genetic , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
15.
Arterioscler Thromb Vasc Biol ; 33(3): 533-43, 2013 Mar.
Article En | MEDLINE | ID: mdl-23288173

OBJECTIVE: Histone deacetylases (HDACs) modulate gene expression by deacetylation of histone and nonhistone proteins. Several HDACs control angiogenesis, but the role of HDAC9 is unclear. METHODS AND RESULTS: Here, we analyzed the function of HDAC9 in angiogenesis and its involvement in regulating microRNAs. In vitro, silencing of HDAC9 reduces endothelial cell tube formation and sprouting. Furthermore, HDAC9 silencing decreases vessel formation in a spheroid-based Matrigel plug assay in mice and disturbs vascular patterning in zebrafish embryos. Genetic deletion of HDAC9 reduces retinal vessel outgrowth and impairs blood flow recovery after hindlimb ischemia. Consistently, overexpression of HDAC9 increases endothelial cell sprouting, whereas mutant constructs lacking the catalytic domain, the nuclear localization sequence, or sumoylation site show no effect. To determine the mechanism underlying the proangiogenic effect of HDAC9, we measured the expression of the microRNA (miR)-17-92 cluster, which is known for its antiangiogenic activity. We demonstrate that silencing of HDAC9 in endothelial cells increases the expression of miR-17-92. Inhibition of miR-17-20a rescues the sprouting defects induced by HDAC9 silencing in vitro and blocking miR-17 expression partially reverses the disturbed vascular patterning of HDAC9 knockdown in zebrafish embryos. CONCLUSIONS: We found that HDAC9 promotes angiogenesis and transcriptionally represses the miR-17-92 cluster.


Histone Deacetylases/metabolism , Human Umbilical Vein Endothelial Cells/enzymology , Ischemia/enzymology , MicroRNAs/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Repressor Proteins/metabolism , Retinal Neovascularization/enzymology , Zebrafish Proteins/metabolism , Animals , Disease Models, Animal , Gene Knockdown Techniques , HEK293 Cells , Hindlimb , Histone Deacetylases/deficiency , Histone Deacetylases/genetics , Humans , Ischemia/genetics , Ischemia/physiopathology , Mice , Mice, Knockout , MicroRNAs/genetics , Mutation , Neovascularization, Physiologic/genetics , RNA Interference , RNA, Long Noncoding , Regional Blood Flow , Repressor Proteins/deficiency , Repressor Proteins/genetics , Retinal Neovascularization/genetics , Retinal Neovascularization/physiopathology , Transfection , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
16.
Circ Res ; 112(6): 924-34, 2013 Mar 15.
Article En | MEDLINE | ID: mdl-23362312

RATIONALE: Polarity proteins are involved in the apico-basal orientation of epithelial cells, but relatively little is known regarding their function in mesenchymal cells. OBJECTIVE: We hypothesized that polarity proteins also contribute to endothelial processes like angiogenesis. METHODS AND RESULTS: Screening of endothelial cells revealed high expression of the polarity protein Scribble (Scrib). On fibronectin-coated carriers Scrib siRNA (siScrib) blocked directed but not random migration of human umbilical vein endothelial cells and led to an increased number and disturbed orientation of cellular lamellipodia. Coimmunoprecipitation/mass spectrometry and glutathione S-transferase (GST) pulldown assays identified integrin α5 as a novel Scrib interacting protein. By total internal reflection fluorescence (TIRF) microscopy, Scrib and integrin α5 colocalize at the basal plasma membrane of endothelial cells. Western blot and fluorescence activated cell sorting (FACS) analysis revealed that silencing of Scrib reduced the protein amount and surface expression of integrin α5 whereas surface expression of integrin αV was unaffected. Moreover, in contrast to fibronectin, the ligand of integrin α5, directional migration on collagen mediated by collagen-binding integrins was unaffected by siScrib. Mechanistically, Scrib supported integrin α5 recycling and protein stability by blocking its interaction with Rab7a, its translocation into lysosomes, and its subsequent degradation by pepstatin-sensitive proteases. In siScrib-treated cells, reinduction of the wild-type protein but not of PSD95, Dlg, ZO-1 (PDZ), or leucine rich repeat domain mutants restored integrin α5 abundance and directional cell migration. The downregulation of Scrib function in Tg(kdrl:EGFP)(s843) transgenic zebrafish embryos delayed the angiogenesis of intersegmental vessels. CONCLUSIONS: Scrib is a novel regulator of integrin α5 turnover and sorting, which is required for oriented cell migration and sprouting angiogenesis.


Cell Movement/physiology , Cell Polarity/physiology , Human Umbilical Vein Endothelial Cells/physiology , Integrin alpha5/metabolism , Membrane Proteins/physiology , Neovascularization, Physiologic/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Migration Assays , Cell Movement/drug effects , Endothelial Cells/physiology , Humans , Integrin alphaV/metabolism , Membrane Proteins/antagonists & inhibitors , Mice , RNA, Small Interfering/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors
17.
Circ Res ; 111(7): 854-62, 2012 Sep 14.
Article En | MEDLINE | ID: mdl-22821930

RATIONALE: Cell therapy is a promising option for the treatment of acute or chronic myocardial ischemia. The intracoronary infusion of cells imposes the potential risk of cell clotting, which may be prevented by the addition of anticoagulants. However, a comprehensive analysis of the effects of anticoagulants on the function of the cells is missing. OBJECTIVE: Here, we investigated the effects of heparin and the thrombin inhibitor bivalirudin on bone marrow-derived mononuclear cell (BMC) functional activity and homing capacity. METHODS AND RESULTS: Heparin, but not bivalirudin profoundly and dose-dependently inhibited basal and stromal cell-derived factor 1 (SDF-1)-induced BMC migration. Incubation of BMCs with 20 U/mL heparin for 30 minutes abrogated SDF-1-induced BMC invasion (16±8% of control; P<0.01), whereas no effects on apoptosis or colony formation were observed (80±33% and 100±44% of control, respectively). Pretreatment of BMCs with heparin significantly reduced the homing of the injected cells in a mouse ear-wound model (69±10% of control; P<0.05). In contrast, bivalirudin did not inhibit in vivo homing of BMCs. Mechanistically, heparin binds to both, the chemoattractant SDF-1 and its receptor, chemokine receptor 4 (CXCR4), blocking CXCR4 internalization as well as SDF-1/CXCR4 signaling after SDF-1 stimulation. CONCLUSIONS: Heparin blocks SDF-1/CXCR4 signaling by binding to the ligand as well as the receptor, thereby interfering with migration and homing of BMCs. In contrast, the thrombin inhibitor bivalirudin did not interfere with BMC homing or SDF-1/CXCR4 signaling. These findings suggest that bivalirudin but not heparin might be recommended as an anticoagulant for intracoronary infusion of BMCs for cell therapy after cardiac ischemia.


Bone Marrow Cells/cytology , Cell- and Tissue-Based Therapy , Chemokine CXCL12/drug effects , Heparin/pharmacology , Leukocytes, Mononuclear/drug effects , Myocardial Infarction/therapy , Receptors, CXCR4/drug effects , Signal Transduction/drug effects , Animals , Anticoagulants/pharmacology , Antithrombins/pharmacology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chemokine CXCL12/physiology , Disease Models, Animal , Female , Hirudins/pharmacology , Humans , In Vitro Techniques , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/physiology , Mice , Mice, Inbred Strains , Peptide Fragments/pharmacology , Receptors, CXCR4/physiology , Recombinant Proteins/pharmacology , Signal Transduction/physiology
18.
J Am Coll Cardiol ; 59(23): 2107-17, 2012 Jun 05.
Article En | MEDLINE | ID: mdl-22651868

OBJECTIVES: This study evaluated the regulation and function of micro-RNAs (miRs) in bone marrow-mononuclear cells (BMCs). BACKGROUND: Although cell therapy with BMCs may represent a therapeutic option to treat patients with heart disease, the impaired functionality of patient-derived cells remains a major challenge. Small noncoding miRs post-transcriptionally control gene expression patterns and play crucial roles in modulating cell survival and function. METHODS: Micro-RNAs were detected by miR profiling in BMCs isolated from healthy volunteers (n = 6) or from patients with myocardial infarction (n = 6), and the results were confirmed by polymerase chain reaction (PCR) in a larger cohort (n = 37). The function of selected miRs was determined by gain-of-function studies in vitro and by locked nuclear acid (LNA) modified inhibitors in vitro and in vivo. RESULTS: We identified several miRs that are up-regulated in BMCs from patients with myocardial infarction compared with BMCs from healthy controls, including the pro-apoptotic and antiproliferative miR-34a and the hypoxia-controlled miR-210. Inhibition of miR-34 by LNA-34a significantly reduced miR-34a expression and blocked hydrogen peroxide-induced cell death of BMC in vitro, whereas overexpression of miR-34a reduced the survival of BMCs in vitro. Pre-treatment of BMCs with LNA-34a ex vivo significantly increased the therapeutic benefit of transplanted BMCs in mice after acute myocardial infarction (AMI). CONCLUSIONS: These results demonstrate that cardiovascular disease modulates the miR expression of BMCs in humans. Reducing the expression of the pro-apoptotic miR-34a improves the survival of BMCs in vitro and enhances the therapeutic benefit of cell therapy in mice after AMI.


Bone Marrow Cells/cytology , Cell Survival/genetics , Gene Expression Regulation , MicroRNAs/genetics , Myocardial Infarction/genetics , Aged , Analysis of Variance , Animals , Blotting, Western , Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/therapy , Case-Control Studies , Cells, Cultured , Cohort Studies , Disease Models, Animal , Female , Humans , Male , Mice , Middle Aged , Myocardial Infarction/mortality , Myocardial Infarction/therapy , Polymerase Chain Reaction/methods , Reference Values
19.
J Biol Chem ; 286(39): 34335-45, 2011 Sep 30.
Article En | MEDLINE | ID: mdl-21795701

The semaphorins are a family of secreted or membrane-bound proteins that are known to guide axons in the developing nervous system. Genetic evidence revealed that a class III semaphorin, semaphorin 3E (Sema3E), and its receptor Plexin-D1 also control the vascular patterning during development. At the molecular level, we have recently shown that Sema3E acts on Plexin-D1 expressed in endothelial cells, thus initiating a novel antiangiogenic signaling pathway that results in the retraction of filopodia in endothelial tip cells. Sema3E induces the rapid disassembly of integrin-mediated adhesive structures, thereby inhibiting endothelial cell adhesion to the extracellular matrix. This process requires the activation of small GTPase Arf6 (ADP-ribosylation factor 6), which regulates intracellular trafficking of ß1 integrin. However, the molecular mechanisms by which Sema3E-Plexin-D1 activates Arf6 remained to be identified. Here we show that GEP100 (guanine nucleotide exchange protein 100)/Brag2, a guanine nucleotide exchange factor for Arf6, mediates Sema3E-induced Arf6 activation in endothelial cells. We provide evidence that upon activation by Sema3E, Plexin-D1 recruits phosphatidylinositol-4-phosphate 5-kinase, and its enzymatic lipid product, phosphatidylinositol 4,5-bisphosphate, binds to the pleckstrin homology domain of GEP100. Phosphatidylinositol 4,5-bisphosphate binding to GEP100 enhances its guanine nucleotide exchange factor activity toward Arf6, thus resulting in the disassembly of integrin-mediated focal adhesions and endothelial cell collapse. Our present study reveals a novel phospholipid-regulated antiangiogenic signaling pathway whereby Sema3E activates Arf6 through Plexin-D1 and consequently controls integrin-mediated endothelial cell attachment to the extracellular matrix and migration.


ADP-Ribosylation Factors/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Endothelial Cells/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Semaphorins/metabolism , Signal Transduction/physiology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Animals , COS Cells , Cell Adhesion/physiology , Cell Adhesion Molecules, Neuronal/genetics , Cell Movement/physiology , Chlorocebus aethiops , Endothelial Cells/cytology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Integrins/genetics , Integrins/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins , Phosphotransferases (Alcohol Group Acceptor)/genetics , Semaphorins/genetics
...