Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 113(6): 681-691, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28453731

ABSTRACT

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.


Subject(s)
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
2.
Arterioscler Thromb Vasc Biol ; 36(7): 1425-33, 2016 07.
Article in English | MEDLINE | ID: mdl-27199445

ABSTRACT

OBJECTIVE: Jumonji C (JmjC) domain-containing proteins modify histone and nonhistone proteins thereby controlling cellular functions. However, the role of JmjC proteins in angiogenesis is largely unknown. Here, we characterize the expression of JmjC domain-containing proteins after inducing endothelial differentiation of murine embryonic stem cells and study the function of JmjC domain-only proteins in endothelial cell (EC) functions. APPROACH AND RESULTS: We identified a large number of JmjC domain-containing proteins regulated by endothelial differentiation of murine embryonic stem cells. Among the family of JmjC domain-only proteins, Jmjd8 was significantly upregulated on endothelial differentiation. Knockdown of Jmjd8 in ECs significantly decreased in vitro network formation and sprouting in the spheroid assay. JMJD8 is exclusively detectable in the cytoplasm, excluding a function as a histone-modifying enzyme. Mass spectrometry analysis revealed JMJD8-interacting proteins with known functions in cellular metabolism like pyruvate kinase M2. Accordingly, knockdown of pyruvate kinase M2 in human umbilical vein ECs decreased endothelial sprouting in the spheroid assay. Knockdown of JMJD8 caused a reduction of EC metabolism as measured by Seahorse Bioscience extracellular flux analysis. Conversely, overexpression of JMJD8 enhanced cellular oxygen consumption rate of ECs, reflecting an increased mitochondrial respiration. CONCLUSIONS: Jmjd8 is upregulated during endothelial differentiation and regulates endothelial sprouting and metabolism by interacting with pyruvate kinase M2.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Embryonic Stem Cells/enzymology , Endothelial Progenitor Cells/enzymology , Energy Metabolism , Human Umbilical Vein Endothelial Cells/enzymology , Jumonji Domain-Containing Histone Demethylases/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Pyruvate Kinase/metabolism , Thyroid Hormones/metabolism , Animals , Carrier Proteins/genetics , Cell Respiration , HEK293 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Membrane Proteins/genetics , Mice , Mitochondria/enzymology , Oxygen Consumption , Protein Binding , Pyruvate Kinase/genetics , RNA Interference , Signal Transduction , Thyroid Hormones/genetics , Time Factors , Transfection , Up-Regulation , Thyroid Hormone-Binding Proteins
3.
J Mol Cell Cardiol ; 88: 111-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26456066

ABSTRACT

MicroRNAs are endogenously expressed small noncoding RNAs that regulate gene expression. Laminar blood flow induces atheroprotective gene expression in endothelial cells (ECs) in part by upregulating the transcription factor KLF2. Here, we identified KLF2- and flow-responsive miRs that affect gene expression in ECs. Bioinformatic assessment of mRNA expression patterns identified the miR-30-5p seed sequence to be highly enriched in mRNAs that are downregulated by KLF2. Indeed, KLF2 overexpression and shear stress stimulation in vitro and in vivo increased the expression of miR-30-5p family members. Furthermore, we identified angiopoietin 2 (Ang2) as a target of miR-30. MiR-30 overexpression reduces Ang2 levels, whereas miR-30 inhibition by LNA-antimiRs induces Ang2 expression. Consistently, miR-30 reduced basal and TNF-α-induced expression of the inflammatory cell­cell adhesion molecules E-selectin, ICAM1 and VCAM1, which was rescued by stimulation with exogenous Ang2. In summary, KLF2 and shear stress increase the expression of the miR-30-5p family which acts in an anti-inflammatory manner in ECs by impairing the expression of Ang2 and inflammatory cell­cell adhesion molecules. The upregulation of miR-30-5p family members may contribute to the atheroprotective effects of shear stress.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , Stress, Mechanical , Vesicular Transport Proteins/genetics , Adenoviridae/genetics , Base Sequence , Computational Biology , E-Selectin/genetics , E-Selectin/metabolism , Gene Expression Regulation , Hemorheology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Kruppel-Like Transcription Factors/metabolism , Lentivirus/genetics , MicroRNAs/metabolism , Molecular Sequence Data , RNA, Messenger/metabolism , Signal Transduction , Transduction, Genetic , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vesicular Transport Proteins/metabolism
4.
Biochem Soc Trans ; 42(6): 1576-83, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25399573

ABSTRACT

The blood vasculature is constantly adapting to meet the demand from tissue. In so doing, branches may form, reorganize or regress. These complex processes employ integration of multiple signalling cascades, some of them being restricted to endothelial and mural cells and, hence, suitable for targeting of the vasculature. Both genetic and drug targeting experiments have demonstrated the requirement for the vascular endothelial growth factor (VEGF) system, the Delta-like-Notch system and the transforming growth factor ß (TGFß)/bone morphogenetic protein (BMP) cascades in vascular development. Although several of these signalling cascades in part converge into common downstream components, they differ in temporal and spatial regulation and expression. For example, the pro-angiogenic VEGFA is secreted by cells in need of oxygen, presented to the basal side of the endothelium, whereas BMP9 and BMP10 are supplied via the bloodstream in constant interaction with the apical side to suppress angiogenesis. Delta-like 4 (DLL4), on the other hand, is provided as an endothelial membrane bound ligand. In the present article, we discuss recent data on the integration of these pathways in the process of sprouting angiogenesis and vascular patterning and malformation.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Neovascularization, Physiologic , Receptors, Notch/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Humans
5.
Int J Radiat Biol ; 90(9): 778-89, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24913294

ABSTRACT

PURPOSE: To investigate the effects of cranial irradiation on the neurovascular niche in the young brain. Disruption of this niche has previously been observed in the adult rat brain after irradiation. MATERIALS AND METHODS: We subjected postnatal day 14 (P14) mice to a single dose of 8 Gy whole brain irradiation and measured the distance between microvessels and either neural progenitor cells (doublecortin-positive, DCX(+)) or proliferating cells (Ki-67(+)) in the dorsal hippocampal subgranular zone (SGZ) 6 hours, 1 week and 7 weeks post-irradiation. In addition, pericyte coverage of microvessels in the SGZ was measured. RESULTS: DCX(+) and Ki-67(+) cells were located closer to microvessels in the adult brain compared to young, still growing brains, constituting new information on normal development. We found an increased distance between microvessels and DCX(+) cells 6 h post-irradiation and between microvessels and Ki-67(+) cells 1 week post-irradiation. Furthermore, pericyte coverage was transiently decreased by 17% 6 h post-irradiation. CONCLUSIONS: The hippocampal neurovascular niche in the young, growing brain is transiently disrupted by irradiation. It remains to be elucidated what role these transient changes play in the apparently permanent ablation of hippocampal neurogenesis previously demonstrated in the same model.


Subject(s)
Brain/radiation effects , Hippocampus/radiation effects , Neurogenesis/physiology , Animals , Brain/growth & development , Cell Death , Cell Proliferation , Dentate Gyrus/radiation effects , Doublecortin Protein , Hippocampus/growth & development , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Microcirculation , Neurons/radiation effects , Particle Accelerators , Pericytes/cytology , Radiotherapy , Rats , Time Factors
6.
Nature ; 495(7439): 107-10, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23426265

ABSTRACT

Ageing is the predominant risk factor for cardiovascular diseases and contributes to a significantly worse outcome in patients with acute myocardial infarction. MicroRNAs (miRNAs) have emerged as crucial regulators of cardiovascular function and some miRNAs have key roles in ageing. We propose that altered expression of miRNAs in the heart during ageing contributes to the age-dependent decline in cardiac function. Here we show that miR-34a is induced in the ageing heart and that in vivo silencing or genetic deletion of miR-34a reduces age-associated cardiomyocyte cell death. Moreover, miR-34a inhibition reduces cell death and fibrosis following acute myocardial infarction and improves recovery of myocardial function. Mechanistically, we identified PNUTS (also known as PPP1R10) as a novel direct miR-34a target, which reduces telomere shortening, DNA damage responses and cardiomyocyte apoptosis, and improves functional recovery after acute myocardial infarction. Together, these results identify age-induced expression of miR-34a and inhibition of its target PNUTS as a key mechanism that regulates cardiac contractile function during ageing and after acute myocardial infarction, by inducing DNA damage responses and telomere attrition.


Subject(s)
Aging/physiology , Gene Expression Regulation , Heart/physiology , MicroRNAs/genetics , Myocardium/metabolism , Aging/genetics , Aging/pathology , Animals , Apoptosis , DNA Damage , Fibrosis/genetics , Fibrosis/pathology , Gene Deletion , Gene Knockout Techniques , Genetic Therapy , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Substrate Specificity , Telomere/genetics , Telomere/metabolism
7.
Circ Res ; 112(6): 924-34, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23362312

ABSTRACT

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.


Subject(s)
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
8.
Arterioscler Thromb Vasc Biol ; 33(3): 533-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23288173

ABSTRACT

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.


Subject(s)
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
9.
Blood ; 119(6): 1607-16, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22184411

ABSTRACT

MicroRNAs (miRs) are small RNAs that regulate gene expression at the posttranscriptional level. miR-27 is expressed in endothelial cells, but the specific functions of miR-27b and its family member miR-27a are largely unknown. Here we demonstrate that overexpression of miR-27a and miR-27b significantly increased endothelial cell sprouting. Inhibition of both miR-27a and miR-27b impaired endothelial cell sprout formation and induced endothelial cell repulsion in vitro. In vivo, inhibition of miR-27a/b decreased the number of perfused vessels in Matrigel plugs and impaired embryonic vessel formation in zebrafish. Mechanistically, miR-27 regulated the expression of the angiogenesis inhibitor semaphorin 6A (SEMA6A) in vitro and in vivo and targeted the 3'-untranslated region of SEMA6A. Silencing of SEMA6A partially reversed the inhibition of endothelial cell sprouting and abrogated the repulsion of endothelial cells mediated by miR-27a/b inhibition, indicating that SEMA6A is a functionally relevant miR-27 downstream target regulating endothelial cell repulsion. In summary, we show that miR-27a/b promotes angiogenesis by targeting the angiogenesis inhibitor SEMA6A, which controls repulsion of neighboring endothelial cells.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Semaphorins/genetics , 3' Untranslated Regions/genetics , Animals , Blood Vessels/embryology , Blood Vessels/metabolism , Blotting, Western , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endothelial Cells/physiology , Gene Expression , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Neovascularization, Physiologic/physiology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Semaphorins/metabolism , Transfection , Zebrafish/embryology , Zebrafish/genetics
10.
EMBO J ; 30(20): 4142-56, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21847094

ABSTRACT

Histone deacetylases (HDACs) deacetylate histones and non-histone proteins, thereby affecting protein activity and gene expression. The regulation and function of the cytoplasmic class IIb HDAC6 in endothelial cells (ECs) is largely unexplored. Here, we demonstrate that HDAC6 is upregulated by hypoxia and is essential for angiogenesis. Silencing of HDAC6 in ECs decreases sprouting and migration in vitro and formation of functional vascular networks in matrigel plugs in vivo. HDAC6 regulates zebrafish vessel formation, and HDAC6-deficient mice showed a reduced formation of perfused vessels in matrigel plugs. Consistently, overexpression of wild-type HDAC6 increases sprouting from spheroids. HDAC6 function requires the catalytic activity but is independent of ubiquitin binding and deacetylation of α-tubulin. Instead, we found that HDAC6 interacts with and deacetylates the actin-remodelling protein cortactin in ECs, which is essential for zebrafish vessel formation and which mediates the angiogenic effect of HDAC6. In summary, we show that HDAC6 is necessary for angiogenesis in vivo and in vitro, involving the interaction and deacetylation of cortactin that regulates EC migration and sprouting.


Subject(s)
Cell Movement , Cortactin/metabolism , Histone Deacetylases/metabolism , Neovascularization, Physiologic , Tubulin/metabolism , Zebrafish Proteins/metabolism , Acetylation , Animals , Carcinoma, Lewis Lung/enzymology , Cells, Cultured , Endothelial Cells/metabolism , Female , Histone Deacetylase 6 , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, Knockout
11.
Blood ; 113(22): 5669-79, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19351956

ABSTRACT

Class IIa histone deacetylases (HDACs) are signal-responsive regulators of gene expression involved in vascular homeostasis. To investigate the differential role of class IIa HDACs for the regulation of angiogenesis, we used siRNA to specifically suppress the individual HDAC isoenzymes. Silencing of HDAC5 exhibited a unique pro-angiogenic effect evidenced by increased endothelial cell migration, sprouting, and tube formation. Consistently, overexpression of HDAC5 decreased sprout formation, indicating that HDAC5 is a negative regulator of angiogenesis. The antiangiogenic activity of HDAC5 was independent of myocyte enhancer factor-2 binding and its deacetylase activity but required a nuclear localization indicating that HDAC5 might affect the transcriptional regulation of gene expression. To identify putative HDAC5 targets, we performed microarray expression analysis. Silencing of HDAC5 increased the expression of fibroblast growth factor 2 (FGF2) and angiogenic guidance factors, including Slit2. Antagonization of FGF2 or Slit2 reduced sprout induction in response to HDAC5 siRNA. Chromatin immunoprecipitation assays demonstrate that HDAC5 binds to the promoter of FGF2 and Slit2. In summary, HDAC5 represses angiogenic genes, such as FGF2 and Slit2, which causally contribute to capillary-like sprouting of endothelial cells. The derepression of angiogenic genes by HDAC5 inactivation may provide a useful therapeutic target for induction of angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , Histone Deacetylases/physiology , Neovascularization, Physiologic/genetics , Angiogenesis Inhibitors/antagonists & inhibitors , Angiogenesis Inhibitors/physiology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/physiology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/physiology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Models, Biological , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...