Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
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.
Nucleic Acids Res ; 51(2): 870-890, 2023 01 25.
Article En | MEDLINE | ID: mdl-36620874

Hypoxia induces massive changes in alternative splicing (AS) to adapt cells to the lack of oxygen. Here, we identify the splicing factor SRSF6 as a key factor in the AS response to hypoxia. The SRSF6 level is strongly reduced in acute hypoxia, which serves a dual purpose: it allows for exon skipping and triggers the dispersal of nuclear speckles. Our data suggest that cells use dispersal of nuclear speckles to reprogram their gene expression during hypoxic adaptation and that SRSF6 plays an important role in cohesion of nuclear speckles. Down-regulation of SRSF6 is achieved through inclusion of a poison cassette exon (PCE) promoted by SRSF4. Removing the PCE 3' splice site using CRISPR/Cas9 abolishes SRSF6 reduction in hypoxia. Aberrantly high SRSF6 levels in hypoxia attenuate hypoxia-mediated AS and impair dispersal of nuclear speckles. As a consequence, proliferation and genomic instability are increased, while the stress response is suppressed. The SRSF4-PCE-SRSF6 hypoxia axis is active in different cancer types, and high SRSF6 expression in hypoxic tumors correlates with a poor prognosis. We propose that the ultra-conserved PCE of SRSF6 acts as a tumor suppressor and that its inclusion in hypoxia is crucial to reduce SRSF6 levels. This may prevent tumor cells from entering the metastatic route of hypoxia adaptation.


Cell Hypoxia , Nuclear Speckles , RNA Splicing , Serine-Arginine Splicing Factors , Humans , Alternative Splicing , Exons/genetics , Phosphoproteins/genetics , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , HeLa Cells
3.
Noncoding RNA ; 8(4)2022 Jun 30.
Article En | MEDLINE | ID: mdl-35893232

Besides transcription, RNA decay accounts for a large proportion of regulated gene expression and is paramount for cellular functions. Classical RNA surveillance pathways, like nonsense-mediated decay (NMD), are also implicated in the turnover of non-mutant transcripts. Whereas numerous protein factors have been assigned to distinct RNA decay pathways, the contribution of long non-coding RNAs (lncRNAs) to RNA turnover remains unknown. Here we identify the lncRNA CALA as a potent regulator of RNA turnover in endothelial cells. We demonstrate that CALA forms cytoplasmic ribonucleoprotein complexes with G3BP1 and regulates endothelial cell functions. A detailed characterization of these G3BP1-positive complexes by mass spectrometry identifies UPF1 and numerous other NMD factors having cytoplasmic G3BP1-association that is CALA-dependent. Importantly, CALA silencing impairs degradation of NMD target transcripts, establishing CALA as a non-coding regulator of RNA steady-state levels in the endothelium.

4.
EMBO Rep ; 23(6): e54157, 2022 06 07.
Article En | MEDLINE | ID: mdl-35527520

Vascular integrity is essential for organ homeostasis to prevent edema formation and infiltration of inflammatory cells. Long non-coding RNAs (lncRNAs) are important regulators of gene expression and often expressed in a cell type-specific manner. By screening for endothelial-enriched lncRNAs, we identified the undescribed lncRNA NTRAS to control endothelial cell functions. Silencing of NTRAS induces endothelial cell dysfunction in vitro and increases vascular permeability and lethality in mice. Biochemical analysis revealed that NTRAS, through its CA-dinucleotide repeat motif, sequesters the splicing regulator hnRNPL to control alternative splicing of tight junction protein 1 (TJP1; also named zona occludens 1, ZO-1) pre-mRNA. Deletion of the hnRNPL binding motif in mice (Ntras∆CA/∆CA ) significantly repressed TJP1 exon 20 usage, favoring expression of the TJP1α- isoform, which augments permeability of the endothelial monolayer. Ntras∆CA/∆CA mice further showed reduced retinal vessel growth and increased vascular permeability and myocarditis. In summary, this study demonstrates that NTRAS is an essential gatekeeper of vascular integrity.


RNA, Long Noncoding , Alternative Splicing , Animals , Endothelial Cells/metabolism , Mice , Permeability , Protein Isoforms/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tight Junctions/metabolism
5.
Circ Res ; 130(1): 67-79, 2022 01 07.
Article En | MEDLINE | ID: mdl-34789007

BACKGROUND: Circular RNAs (circRNAs) are generated by back splicing of mostly mRNAs and are gaining increasing attention as a novel class of regulatory RNAs that control various cellular functions. However, their physiological roles and functional conservation in vivo are rarely addressed, given the inherent challenges of their genetic inactivation. Here, we aimed to identify locus conserved circRNAs in mice and humans, which can be genetically deleted due to retained intronic elements not contained in the mRNA host gene to eventually address functional conservation. METHODS AND RESULTS: Combining published endothelial RNA-sequencing data sets with circRNAs of the circATLAS databank, we identified locus-conserved circRNA retaining intronic elements between mice and humans. CRISPR/Cas9 mediated genetic depletion of the top expressed circRNA cZfp292 resulted in an altered endothelial morphology and aberrant flow alignment in the aorta in vivo. Consistently, depletion of cZNF292 in endothelial cells in vitro abolished laminar flow-induced alterations in cell orientation, paxillin localization and focal adhesion organization. Mechanistically, we identified the protein SDOS (syndesmos) to specifically interact with cZNF292 in endothelial cells by RNA-affinity purification and subsequent mass spectrometry analysis. Silencing of SDOS or its protein binding partner Syndecan-4, or mutation of the SDOS-cZNF292 binding site, prevented laminar flow-induced cytoskeletal reorganization thereby recapitulating cZfp292 knockout phenotypes. CONCLUSIONS: Together, our data reveal a hitherto unknown role of cZNF292/cZfp292 in endothelial flow responses, which influences endothelial shape.


DNA-Binding Proteins , Endothelial Cells , Endothelium, Vascular , RNA, Circular , Transcription Factors , Animals , Humans , Mice , Blood Circulation , DNA-Binding Proteins/genetics , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Protein Binding , RNA, Circular/genetics , RNA, Circular/metabolism , Syndecan-4/metabolism , Transcription Factors/genetics
6.
Am J Respir Crit Care Med ; 202(10): 1445-1457, 2020 11 15.
Article En | MEDLINE | ID: mdl-32634060

Rationale: Long noncoding RNAs (lncRNAs) are emerging as important regulators of diverse biological functions. Their role in pulmonary arterial hypertension (PAH) remains to be explored.Objectives: To elucidate the role of TYKRIL (tyrosine kinase receptor-inducing lncRNA) as a regulator of p53/ PDGFRß (platelet-derived growth factor receptor ß) signaling pathway and to investigate its role in PAH.Methods: Pericytes and pulmonary arterial smooth muscle cells exposed to hypoxia and derived from patients with idiopathic PAH were analyzed with RNA sequencing. TYKRIL knockdown was performed in above-mentioned human primary cells and in precision-cut lung slices derived from patients with PAH.Measurements and Main Results: Using RNA sequencing data, TYKRIL was identified to be consistently upregulated in pericytes and pulmonary arterial smooth muscles cells exposed to hypoxia and derived from patients with idiopathic PAH. TYKRIL knockdown reversed the proproliferative (n = 3) and antiapoptotic (n = 3) phenotype induced under hypoxic and idiopathic PAH conditions. Owing to the poor species conservation of TYKRIL, ex vivo studies were performed in precision-cut lung slices from patients with PAH. Knockdown of TYKRIL in precision-cut lung slices decreased the vascular remodeling (n = 5). The number of proliferating cell nuclear antigen-positive cells in the vessels was decreased and the number of terminal deoxynucleotide transferase-mediated dUTP nick end label-positive cells in the vessels was increased in the LNA (locked nucleic acid)-treated group compared with control. Expression of PDGFRß, a key player in PAH, was found to strongly correlate with TYKRIL expression in the patient samples (n = 12), and TYKRIL knockdown decreased PDGFRß expression (n = 3). From the transcription factor-screening array, it was observed that TYKRIL knockdown increased the p53 activity, a known repressor of PDGFRß. RNA immunoprecipitation using various p53 mutants demonstrated that TYKRIL binds to the N-terminal of p53 (an important region for p300 interaction with p53). The proximity ligation assay revealed that TYKRIL interferes with the p53-p300 interaction (n = 3) and regulates p53 nuclear translocation.Conclusions: TYKRIL plays an important role in PAH by regulating the p53/PDGFRß axis.


Gene Expression , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Protein-Tyrosine Kinases/genetics , RNA, Long Noncoding , Receptor, Platelet-Derived Growth Factor beta/genetics , Signal Transduction/genetics , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged
7.
Circ Res ; 126(9): 1127-1145, 2020 04 24.
Article En | MEDLINE | ID: mdl-32324505

The advent of deep sequencing technologies led to the identification of a considerable amount of noncoding RNA transcripts, which are increasingly recognized for their functions in controlling cardiovascular diseases. MicroRNAs have already been studied for a decade, leading to the identification of several vasculoprotective and detrimental species, which might be considered for therapeutic targeting. Other noncoding RNAs such as circular RNAs, YRNAs, or long noncoding RNAs are currently gaining increasing attention, and first studies provide insights into their functions as mediators or antagonists of vascular diseases in vivo. The present review article will provide an overview of the different types of noncoding RNAs controlling the vasculature and focus on the developing field of long noncoding RNAs.


MicroRNAs/genetics , RNA, Circular/genetics , RNA, Long Noncoding/genetics , Vascular Diseases/genetics , Animals , Gene Expression Regulation , Humans , MicroRNAs/metabolism , RNA, Circular/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Vascular Diseases/metabolism , Vascular Diseases/pathology , Vascular Remodeling
8.
Nat Commun ; 11(1): 2039, 2020 04 27.
Article En | MEDLINE | ID: mdl-32341350

Long non-coding RNAs (lncRNAs) contribute to cardiac (patho)physiology. Aging is the major risk factor for cardiovascular disease with cardiomyocyte apoptosis as one underlying cause. Here, we report the identification of the aging-regulated lncRNA Sarrah (ENSMUST00000140003) that is anti-apoptotic in cardiomyocytes. Importantly, loss of SARRAH (OXCT1-AS1) in human engineered heart tissue results in impaired contractile force development. SARRAH directly binds to the promoters of genes downregulated after SARRAH silencing via RNA-DNA triple helix formation and cardiomyocytes lacking the triple helix forming domain of Sarrah show an increase in apoptosis. One of the direct SARRAH targets is NRF2, and restoration of NRF2 levels after SARRAH silencing partially rescues the reduction in cell viability. Overexpression of Sarrah in mice shows better recovery of cardiac contractile function after AMI compared to control mice. In summary, we identified the anti-apoptotic evolutionary conserved lncRNA Sarrah, which is downregulated by aging, as a regulator of cardiomyocyte survival.


Apoptosis , Myocardial Infarction/genetics , Myocytes, Cardiac/cytology , RNA, Long Noncoding/genetics , Aging , Animals , Carrier Proteins/genetics , Cell Survival , Coenzyme A-Transferases/genetics , Disease Models, Animal , Gene Silencing , Humans , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , RNA, Antisense/genetics , RNA, Small Interfering/genetics , p300-CBP Transcription Factors/genetics
9.
Vascul Pharmacol ; 114: 13-22, 2019 03.
Article En | MEDLINE | ID: mdl-30910127

The advent of deep sequencing technologies recently unraveled the complexity of the human genome: Although almost entirely transcribed, only a very minor part of our genome actually accounts for protein coding exons and most is considered non-coding. Among the non-coding transcripts, long non-coding RNAs (lncRNAs) constitute a rather heterogeneous group of linear as well as circular RNAs (circRNAs). LncRNAs act via multiple mechanisms and several lncRNAs were shown to be involved in vascular development, growth and remodeling. For example, the lncRNAs PUNISHER, MALAT1, MEG3, and GATA6-AS regulate vessel formation in vivo, whereas lincRNA-p21 controls smooth muscle cell function and neointima formation. For several other lncRNAs (e.g. SENCR, SMILR, and HypERlnc) functional roles in smooth muscle cells/pericytes have been described in vitro. Less information is available with respect to the function of circRNAs. Here most studies report on expression profiles but some circRNAs (e.g. cANRIL or cZNF292) may also play critical roles in smooth muscle or endothelial cells in vitro. This review summarizes the current knowledge of lncRNA and circRNA functions in vascular biology and disease and discusses their potential use as biomarkers.


Atherosclerosis/metabolism , Blood Vessels/metabolism , RNA, Long Noncoding/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Blood Vessels/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation , Genetic Markers , Humans , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , RNA, Long Noncoding/genetics , Signal Transduction
10.
Circulation ; 139(10): 1320-1334, 2019 03 05.
Article En | MEDLINE | ID: mdl-30586743

BACKGROUND: The majority of the human genome comprises noncoding sequences, which are in part transcribed as long noncoding RNAs (lncRNAs). lncRNAs exhibit multiple functions, including the epigenetic control of gene expression. In this study, the effect of the lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) on atherosclerosis was examined. METHODS: The effect of MALAT1 on atherosclerosis was determined in apolipoprotein E-deficient (Apoe-/-) MALAT1-deficient (Malat1-/-) mice that were fed with a high-fat diet and by studying the regulation of MALAT1 in human plaques. RESULTS: Apoe-/- Malat1-/- mice that were fed a high-fat diet showed increased plaque size and infiltration of inflammatory CD45+ cells compared with Apoe-/- Malat1+/+ control mice. Bone marrow transplantation of Apoe-/- Malat1-/- bone marrow cells in Apoe-/- Malat1+/+ mice enhanced atherosclerotic lesion formation, which suggests that hematopoietic cells mediate the proatherosclerotic phenotype. Indeed, bone marrow cells isolated from Malat1-/- mice showed increased adhesion to endothelial cells and elevated levels of proinflammatory mediators. Moreover, myeloid cells of Malat1-/- mice displayed enhanced adhesion to atherosclerotic arteries in vivo. The anti-inflammatory effects of MALAT1 were attributed in part to reduction of the microRNA miR-503. MALAT1 expression was further significantly decreased in human plaques compared with normal arteries and was lower in symptomatic versus asymptomatic patients. Lower levels of MALAT1 in human plaques were associated with a worse prognosis. CONCLUSIONS: Reduced levels of MALAT1 augment atherosclerotic lesion formation in mice and are associated with human atherosclerotic disease. The proatherosclerotic effects observed in Malat1-/- mice were mainly caused by enhanced accumulation of hematopoietic cells.


Aorta/metabolism , Aortitis/metabolism , Atherosclerosis/metabolism , Bone Marrow Cells/metabolism , Hematopoiesis , Plaque, Atherosclerotic , RNA, Long Noncoding/metabolism , Animals , Aorta/pathology , Aortitis/genetics , Aortitis/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Case-Control Studies , Disease Models, Animal , Down-Regulation , Humans , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , Signal Transduction
11.
Nat Commun ; 9(1): 237, 2018 01 16.
Article En | MEDLINE | ID: mdl-29339785

Impaired or excessive growth of endothelial cells contributes to several diseases. However, the functional involvement of regulatory long non-coding RNAs in these processes is not well defined. Here, we show that the long non-coding antisense transcript of GATA6 (GATA6-AS) interacts with the epigenetic regulator LOXL2 to regulate endothelial gene expression via changes in histone methylation. Using RNA deep sequencing, we find that GATA6-AS is upregulated in endothelial cells during hypoxia. Silencing of GATA6-AS diminishes TGF-ß2-induced endothelial-mesenchymal transition in vitro and promotes formation of blood vessels in mice. We identify LOXL2, known to remove activating H3K4me3 chromatin marks, as a GATA6-AS-associated protein, and reveal a set of angiogenesis-related genes that are inversely regulated by LOXL2 and GATA6-AS silencing. As GATA6-AS silencing reduces H3K4me3 methylation of two of these genes, periostin and cyclooxygenase-2, we conclude that GATA6-AS acts as negative regulator of nuclear LOXL2 function.


Amino Acid Oxidoreductases/metabolism , Endothelial Cells/metabolism , GATA6 Transcription Factor/genetics , Gene Expression Regulation/genetics , Hypoxia/genetics , Neovascularization, Physiologic/genetics , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , Animals , Cell Adhesion Molecules/genetics , Cyclooxygenase 2/genetics , Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Gene Silencing , Histone Code/genetics , Histones/metabolism , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Methylation , Mice
14.
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
15.
J Am Coll Cardiol ; 67(10): 1214-1226, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26965544

Recent studies suggest that the majority of the human genome is transcribed, but only about 2% accounts for protein-coding exons. Long noncoding RNAs (lncRNAs) constitute a heterogenic class of RNAs that includes, for example, intergenic lncRNAs, antisense transcripts, and enhancer RNAs. Moreover, alternative splicing can lead to the formation of circular RNAs. In support of putative functions, GWAS for cardiovascular diseases have shown predictive single-nucleotide polymorphisms in lncRNAs, such as the 9p21 susceptibility locus that encodes the lncRNA antisense noncoding RNA in the INK4 locus (ANRIL). Many lncRNAs are regulated during disease. For example, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and myocardial infarction-associated transcript (MIAT) were shown to affect endothelial cell functions and diabetic retinopathy, whereas lincRNA-p21 controls neointima formation. In the heart, several lncRNAs were shown to act as microRNA sponges and to control ischemia-reperfusion injury or act as epigenetic regulators. In this review, the authors summarize the current understanding of lncRNA functions and their role as biomarkers in cardiovascular diseases.


Gene Expression Regulation , Myocardial Infarction/genetics , RNA, Long Noncoding/genetics , RNA/genetics , Biomarkers/blood , Genetic Predisposition to Disease , Humans , Myocardial Infarction/blood , RNA, Long Noncoding/blood
16.
Circ Res ; 117(10): 884-90, 2015 Oct 23.
Article En | MEDLINE | ID: mdl-26377962

RATIONALE: Circular RNAs (circRNAs) are noncoding RNAs generated by back splicing. Back splicing has been considered a rare event, but recent studies suggest that circRNAs are widely expressed. However, the expression, regulation, and function of circRNAs in vascular cells is still unknown. OBJECTIVE: Here, we characterize the expression, regulation, and function of circRNAs in endothelial cells. METHODS AND RESULTS: Endothelial circRNAs were identified by computational analysis of ribo-minus RNA generated from human umbilical venous endothelial cells cultured under normoxic or hypoxic conditions. Selected circRNAs were biochemically characterized, and we found that the majority of them lacks polyadenylation, is resistant to RNase R digestion and localized to the cytoplasm. We further validated the hypoxia-induced circRNAs cZNF292, cAFF1, and cDENND4C, as well as the downregulated cTHSD1 by reverse transcription polymerase chain reaction in cultured endothelial cells. Cloning of cZNF292 validated the predicted back splicing of exon 4 to a new alternative exon 1A. Silencing of cZNF292 inhibited cZNF292 expression and reduced tube formation and spheroid sprouting of endothelial cells in vitro. The expression of pre-mRNA or mRNA of the host gene was not affected by silencing of cZNF292. No validated microRNA-binding sites for cZNF292 were detected in Argonaute high-throughput sequencing of RNA isolated by cross-linking and immunoprecipitation data sets, suggesting that cZNF292 does not act as a microRNA sponge. CONCLUSIONS: We show that the majority of the selected endothelial circRNAs fulfill all criteria of bona fide circRNAs. The circRNA cZNF292 exhibits proangiogenic activities in vitro. These data suggest that endothelial circRNAs are regulated by hypoxia and have biological functions.


Carrier Proteins/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Nerve Tissue Proteins/genetics , RNA/genetics , Carrier Proteins/metabolism , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Cluster Analysis , Computer Simulation , Exons , Gene Expression Profiling , Gene Expression Regulation , Humans , Neovascularization, Physiologic , Nerve Tissue Proteins/metabolism , RNA/metabolism , RNA Interference , RNA Splicing , RNA, Circular , Reverse Transcriptase Polymerase Chain Reaction , Transfection
17.
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
19.
Int J Med Microbiol ; 302(4-5): 221-4, 2012 Oct.
Article En | MEDLINE | ID: mdl-22964417

The parasitic unicellular trypanosomatids are responsible for several fatal diseases in humans and livestock. Regarding their biochemistry and molecular biology, they possess a multitude of special features such as polycistronic transcription of protein-coding genes. The resulting long primary transcripts need to be processed by coupled trans-splicing and polyadenylation reactions, thereby generating mature mRNAs. Catalyzed by a large ribonucleoprotein complex termed the spliceosome, trans-splicing attaches a 39-nucleotide leader sequence, which is derived from the Spliced Leader (SL) RNA, to each protein-coding gene. Recent genome-wide studies demonstrated that alternative trans-splicing increases mRNA and protein diversity in these organisms. In this mini-review we give an overview of the current state of research on trans-splicing.


RNA, Messenger/metabolism , RNA, Protozoan/metabolism , Trans-Splicing , Trypanosoma/genetics , Alternative Splicing , Polyadenylation , Protein Binding , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Protozoan/genetics , RNA, Spliced Leader/genetics , RNA, Spliced Leader/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Transcription, Genetic , Trypanosoma/metabolism
20.
RNA Biol ; 8(1): 90-100, 2011.
Article En | MEDLINE | ID: mdl-21282982

Pre-mRNA splicing in trypanosomes requires the SMN-mediated assembly of small nuclear ribonucleoproteins (snRNPs). In contrast to higher eukaryotes, the cellular localization of snRNP biogenesis and the involvement of nuclear-cytoplasmic trafficking in trypanosomes are controversial. By using RNAi knockdown of SMN in T. brucei to investigate its functional role in snRNP assembly, we found dramatic changes in the steady-state levels of snRNAs and snRNPs: The SL RNA accumulates, whereas U1, U4, and U5 snRNA levels decrease, and Sm core assembly in particular of the SL RNA is strongly reduced. In addition, SMN depletion blocks U4/U6 di-snRNP formation; the variant Sm core of the U2 snRNP, however, still forms efficiently after SMN knockdown. Concerning the longstanding question, whether nuclear-cytoplasmic trafficking is involved in trypanosomal snRNP biogenesis, fluorescence in situ hybridization (FISH) and immunofluorescence assays revealed that the SL RNA genes and transcripts colocalize with SMN. Remarkably, SMN silencing leads to a nucleoplasmic accumulation of both SL RNA and the Sm proteins. In sum, our data demonstrate an essential and snRNA-selective role of SMN in snRNP biogenesis in vivo and strongly argue for a nucleoplasmic Sm core assembly of the SL RNP.


RNA, Protozoan/metabolism , RNA, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins/metabolism , Trypanosoma brucei brucei/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Loci , In Situ Hybridization, Fluorescence , RNA Interference , RNA Splicing , RNA, Protozoan/genetics , RNA, Small Nuclear/analysis , RNA, Spliced Leader/metabolism , SMN Complex Proteins/genetics , Trypanosoma brucei brucei/metabolism
...