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










Publication year range
1.
Cells ; 11(23)2022 Nov 22.
Article in English | MEDLINE | ID: mdl-36496981

ABSTRACT

Bicuspid aortic valve (BAV) patients develop ascending aortic (AAo) dilation. The pathogenesis of BAV aortopathy (genetic vs. haemodynamic) remains unclear. This study aims to identify regional changes around the AAo wall in BAV patients with aortopathy, integrating molecular data and clinical imaging. BAV patients with aortopathy (n = 15) were prospectively recruited to surgically collect aortic tissue and measure molecular markers across the AAo circumference. Dilated (anterior/right) vs. non-dilated (posterior/left) circumferential segments were profiled for whole-genomic microRNAs (next-generation RNA sequencing, miRCURY LNA PCR), protein content (tandem mass spectrometry), and elastin fragmentation and degeneration (histomorphometric analysis). Integrated bioinformatic analyses of RNA sequencing and proteomic datasets identified five microRNAs (miR-128-3p, miR-210-3p, miR-150-5p, miR-199b-5p, and miR-21-5p) differentially expressed across the AAo circumference. Among them, three miRNAs (miR-128-3p, miR-150-5p, and miR-199b-5p) were predicted to have an effect on eight common target genes, whose expression was dysregulated, according to proteomic analyses, and involved in the vascular-endothelial growth-factor signalling, Hippo signalling, and arachidonic acid pathways. Decreased elastic fibre levels and elastic layer thickness were observed in the dilated segments. Additionally, in a subset of patients n = 6/15, a four-dimensional cardiac magnetic resonance (CMR) scan was performed. Interestingly, an increase in wall shear stress (WSS) was observed at the anterior/right wall segments, concomitantly with the differentially expressed miRNAs and decreased elastic fibres. This study identified new miRNAs involved in the BAV aortic wall and revealed the concomitant expressional dysregulation of miRNAs, proteins, and elastic fibres on the anterior/right wall in dilated BAV patients, corresponding to regions of elevated WSS.


Subject(s)
Aortic Diseases , Bicuspid Aortic Valve Disease , Heart Valve Diseases , MicroRNAs , Humans , Bicuspid Aortic Valve Disease/complications , Bicuspid Aortic Valve Disease/metabolism , Bicuspid Aortic Valve Disease/pathology , Heart Valve Diseases/diagnostic imaging , Heart Valve Diseases/genetics , Heart Valve Diseases/complications , Aortic Valve/pathology , Proteomics , Aortic Diseases/metabolism , Magnetic Resonance Imaging , MicroRNAs/genetics , MicroRNAs/metabolism
2.
J Mol Cell Cardiol ; 160: 56-70, 2021 11.
Article in English | MEDLINE | ID: mdl-33991529

ABSTRACT

N6-methyladenosine (m6A) is the most abundant and well-studied internal modification of messenger RNAs among the various RNA modifications in eukaryotic cells. Moreover, it is increasingly recognized to regulate non-coding RNAs. The dynamic and reversible nature of m6A is ensured by the precise and coordinated activity of specific proteins able to insert ("write"), bind ("read") or remove ("erase") the m6A modification from coding and non-coding RNA molecules. Mounting evidence suggests a pivotal role for m6A in prenatal and postnatal development and cardiovascular pathophysiology. In the present review we summarise and discuss the major functions played by m6A RNA methylation and its components particularly referring to the cardiovascular system. We present the methods used to study m6A and the most abundantly methylated RNA molecules. Finally, we highlight the possible involvement of the m6A mark in cardiovascular disease as well as the need for further studies to better describe the mechanisms of action and the potential therapeutic role of this RNA modification.


Subject(s)
Adenosine/analogs & derivatives , Cardiovascular Diseases/metabolism , Cardiovascular System/embryology , Cardiovascular System/growth & development , Transcriptome/genetics , Adenosine/genetics , Adenosine/metabolism , Animals , Biomarkers/metabolism , Cardiovascular System/metabolism , Homeostasis/genetics , Humans , Methylation , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism
4.
Mol Ther ; 29(7): 2239-2252, 2021 07 07.
Article in English | MEDLINE | ID: mdl-33744469

ABSTRACT

MicroRNAs (miRNAs) regulate gene expression by post-transcriptional inhibition of target genes. Proangiogenic small extracellular vesicles (sEVs; popularly identified with the name "exosomes") with a composite cargo of miRNAs are secreted by cultured stem cells and present in human biological fluids. Lipid nanoparticles (LNPs) represent an advanced platform for clinically approved delivery of RNA therapeutics. In this study, we aimed to (1) identify the miRNAs responsible for sEV-induced angiogenesis; (2) develop the prototype of bioinspired "artificial exosomes" (AEs) combining LNPs with a proangiogenic miRNA, and (3) validate the angiogenic potential of the bioinspired AEs. We previously reported that human sEVs from bone marrow (BM)-CD34+ cells and pericardial fluid (PF) are proangiogenic. Here, we have shown that sEVs secreted from saphenous vein pericytes and BM mesenchymal stem cells also promote angiogenesis. Analysis of miRNA datasets available in-house or datamined from GEO identified the let-7 family as common miRNA signature of the proangiogenic sEVs. LNPs with either hsa-let-7b-5p or cyanine 5 (Cy5)-conjugated Caenorhabditis elegans miR-39 (Cy5-cel-miR-39; control miRNA) were prepared using microfluidic micromixing. let-7b-5p-AEs did not cause toxicity and transferred functionally active let-7b-5p to recipient endothelial cells (ECs). let-7b-AEs also improved EC survival under hypoxia and angiogenesis in vitro and in vivo. Bioinspired proangiogenic AEs could be further developed into innovative nanomedicine products targeting ischemic diseases.


Subject(s)
Exosomes/metabolism , Extracellular Vesicles/metabolism , Liposomes/chemistry , MicroRNAs/metabolism , Nanoparticles/chemistry , Neovascularization, Physiologic , Pericardial Fluid/physiology , Animals , Exosomes/genetics , Extracellular Vesicles/genetics , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Mice , MicroRNAs/genetics
6.
Mol Ther Nucleic Acids ; 17: 49-62, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31220779

ABSTRACT

MicroRNA-15a (miR-15a) and miR-16, which are transcribed from the miR-15a/miR-16-1 cluster, inhibit post-ischemic angiogenesis. MicroRNA (miRNA) binding to mRNA coding sequences (CDSs) is a newly emerging mechanism of gene expression regulation. We aimed to (1) identify new mediators of the anti-angiogenic action of miR-15a and -16, (2) develop an adenovirus (Ad)-based miR-15a/16 decoy system carrying a luciferase reporter (Luc) to both sense and inhibit miR-15a/16 activity, and (3) investigate Ad.Luc-Decoy-15a/16 therapeutic potential in a mouse limb ischemia (LI) model. LI increased miR-15a and -16 expression in mouse muscular endothelial cells (ECs). The miRNAs also increased in cultured human umbilical vein ECs (HUVECs) exposed to serum starvation, but not hypoxia. Using bioinformatic tools and luciferase activity assays, we characterized miR-15a and -16 binding to Tie2 CDS. In HUVECs, miR-15a or -16 overexpression reduced Tie2 at the protein, but not the mRNA, level. Conversely, miR-15a or -16 inhibition improved angiogenesis in a Tie2-dependent manner. Local Ad.Luc-Decoy-15a/16 delivery increased Tie2 levels in ischemic skeletal muscle and improved post-LI angiogenesis and perfusion recovery, with reduced toe necrosis. Bioluminescent imaging (in vivo imaging system [IVIS]) provided evidence that the Ad.Luc-Decoy-15a/16 system responds to miR-15a/16 increases. In conclusion, we have provided novel mechanistic evidence of the therapeutic potential of local miR-15a/16 inhibition in LI.

7.
Arterioscler Thromb Vasc Biol ; 38(9): 2117-2125, 2018 09.
Article in English | MEDLINE | ID: mdl-30354255

ABSTRACT

Objective- The NTs (neurotrophins), BDNF (brain-derived neurotrophic factor) and NT-3 promote vascular development and angiogenesis. This study investigated the contribution of endogenous NTs in embryonic stem cell (ESC) vascular differentiation and the potential of exogenous BDNF to improve the process of ESC differentiation to endothelial cells (ECs). Approach and Results- Mouse ESCs were differentiated into vascular cells using a 2-dimensional embryoid body (EB) model. Supplementation of either BDNF or NT-3 increased EC progenitors' abundance at day 7 and enlarged the peripheral vascular plexus with ECs and SM22α+ (smooth muscle 22 alpha-positive) smooth muscle cells by day 13. Conversely, inhibition of either BDNF or NT-3 receptor signaling reduced ECs, without affecting smooth muscle cells spread. This suggests that during vascular development, endogenous NTs are especially relevant for endothelial differentiation. At mechanistic level, we have identified that BDNF-driven ESC-endothelial differentiation is mediated by a pathway encompassing the transcriptional repressor EZH2 (enhancer of zeste homolog 2), microRNA-214 (miR-214), and eNOS (endothelial nitric oxide synthase). It was known that eNOS, which is needed for endothelial differentiation, can be transcriptionally repressed by EZH2. In turn, miR-214 targets EZH2 for inhibition. We newly found that in ESC-ECs, BDNF increases miR-214 expression, reduces EZH2 occupancy of the eNOS promoter, and increases eNOS expression. Moreover, we found that NRP-1 (neuropilin 1), KDR (kinase insert domain receptor), and pCas130 (p130 Crk-associated substrate kinase), which reportedly induce definitive endothelial differentiation of pluripotent cells, were increased in BDNF-conditioned ESC-EC. Mechanistically, miR-214 mediated the BDNF-induced expressional changes, contributing to BDNF-driven endothelial differentiation. Finally, BDNF-conditioned ESC-ECs promoted angiogenesis in vitro and in vivo. Conclusions- BDNF promotes ESC-endothelial differentiation acting via miR-214.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Cell Differentiation , Embryonic Stem Cells/physiology , Endothelial Cells/physiology , Enhancer of Zeste Homolog 2 Protein/metabolism , MicroRNAs/metabolism , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Differentiation/drug effects , Cell Line , Crk-Associated Substrate Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Immunophilins/metabolism , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Growth Factors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Circ Res ; 118(1): 38-47, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26472816

ABSTRACT

RATIONALE: Several lines of evidence indicate that the regulation of microRNA (miRNA) levels by different stimuli may contribute to the modulation of stimulus-induced responses. The miR-17-92 cluster has been linked to tumor development and angiogenesis, but its role in vascular endothelial growth factor-induced endothelial cell (EC) functions is unclear and its regulation is unknown. OBJECTIVE: The purpose of this study was to elucidate the mechanism by which VEGF regulates the expression of miR-17-92 cluster in ECs and determine its contribution to the regulation of endothelial angiogenic functions, both in vitro and in vivo. This was done by analyzing the effect of postnatal inactivation of miR-17-92 cluster in the endothelium (miR-17-92 iEC-KO mice) on developmental retinal angiogenesis, VEGF-induced ear angiogenesis, and tumor angiogenesis. METHODS AND RESULTS: Here, we show that Erk/Elk1 activation on VEGF stimulation of ECs is responsible for Elk-1-mediated transcription activation (chromatin immunoprecipitation analysis) of the miR-17-92 cluster. Furthermore, we demonstrate that VEGF-mediated upregulation of the miR-17-92 cluster in vitro is necessary for EC proliferation and angiogenic sprouting. Finally, we provide genetic evidence that miR-17-92 iEC-KO mice have blunted physiological retinal angiogenesis during development and diminished VEGF-induced ear angiogenesis and tumor angiogenesis. Computational analysis and rescue experiments show that PTEN (phosphatase and tensin homolog) is a target of the miR-17-92 cluster and is a crucial mediator of miR-17-92-induced EC proliferation. However, the angiogenic transcriptional program is reduced when miR-17-92 is inhibited. CONCLUSIONS: Taken together, our results indicate that VEGF-induced miR-17-92 cluster expression contributes to the angiogenic switch of ECs and participates in the regulation of angiogenesis.


Subject(s)
Endothelium, Vascular/metabolism , MAP Kinase Signaling System/physiology , MicroRNAs/biosynthesis , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Endothelium, Vascular/drug effects , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , MicroRNAs/genetics , Neovascularization, Physiologic/drug effects
9.
Curr Vasc Pharmacol ; 13(3): 291-304, 2015.
Article in English | MEDLINE | ID: mdl-26156264

ABSTRACT

Atherosclerosis (also known as arteriosclerotic vascular disease) is a chronic inflammatory disease of the arterial wall, characterized by the formation of lipid-laden lesions. The activation of endothelial cells at atherosclerotic lesion-prone sites in the arterial tree results in the up-regulation of cell adhesion molecules and chemokines, which mediate the recruitment of circulating monocytes. Accumulation of monocytes and monocyte-derived phagocytes in the wall of large arteries leads to chronic inflammation and the development and progression of atherosclerosis. The lesion experiences the following steps: foam cell formation, fatty streak accumulation, migration and proliferation of vascular smooth muscle cells, and fibrous cap formation. Finally, the rupture of the unstable fibrous cap causes thrombosis in complications of advanced lesions that leads to unstable coronary syndromes, myocardial infarction and stroke. MicroRNAs have recently emerged as a novel class of gene regulators at the post-transcriptional level. Several functions of vascular cells, such as cell differentiation, contraction, migration, proliferation and inflammation that are involved in angiogenesis, neointimal formation and lipid metabolism underlying various vascular diseases, have been found to be regulated by microRNAs and are described in the present review as well as their potential therapeutic application.


Subject(s)
Atherosclerosis/drug therapy , MicroRNAs/therapeutic use , Vascular Diseases/drug therapy , Animals , Atherosclerosis/pathology , Humans , MicroRNAs/genetics , MicroRNAs/physiology , Muscle, Smooth, Vascular/pathology , Vascular Diseases/pathology
10.
FASEB J ; 29(2): 597-610, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25392271

ABSTRACT

Atherosclerosis is the major cause of death and disability in diabetic and obese subjects with insulin resistance. Akt2, a phosphoinositide-dependent serine-threonine protein kinase, is highly express in insulin-responsive tissues; however, its role during the progression of atherosclerosis remains unknown. Thus, we aimed to investigate the contribution of Akt2 during the progression of atherosclerosis. We found that germ-line Akt2-deficient mice develop similar atherosclerotic plaques as wild-type mice despite higher plasma lipids and glucose levels. It is noteworthy that transplantation of bone marrow cells isolated from Akt2(-/-) mice to Ldlr(-/-) mice results in marked reduction of the progression of atherosclerosis compared with Ldlr(-/-) mice transplanted with wild-type bone marrow cells. In vitro studies indicate that Akt2 is required for macrophage migration in response to proatherogenic cytokines (monocyte chemotactic protein-1 and macrophage colony-stimulating factor). Moreover, Akt2(-/-) macrophages accumulate less cholesterol and have an alternative activated or M2-type phenotype when stimulated with proinflammatory cytokines. Together, these results provide evidence that macrophage Akt2 regulates migration, the inflammatory response and cholesterol metabolism and suggest that targeting Akt2 in macrophages might be beneficial for treating atherosclerosis.


Subject(s)
Atherosclerosis/physiopathology , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Animals , Blood Glucose/metabolism , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Cell Movement , Cholesterol/metabolism , Cytokines/metabolism , Disease Progression , Inflammation , Insulin/chemistry , Leukocytes/cytology , Lipids/blood , Lipoproteins, LDL/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Plaque, Atherosclerotic , Receptors, LDL/genetics
11.
J Cell Mol Med ; 18(6): 1104-12, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24636235

ABSTRACT

Wound healing is a well-regulated but complex process that involves haemostasis, inflammation, proliferation and maturation. Recent reports suggest that microRNAs (miRs) play important roles in dermal wound healing. In fact, miR deregulation has been linked with impaired wound repair. miR-155 has been shown to be induced by inflammatory mediators and plays a central regulatory role in immune responses. We have investigated the potential role of miR-155 in wound healing. By creating punch wounds in the skin of mice, we found an increased expression of miR-155 in wound tissue when compared with healthy skin. Interestingly, analysis of wounds of mice lacking the expression of miR-155 (miR-155(-/-) ) revealed an increased wound closure when compared with wild-type animals. Also, the accelerated wound closing correlated with elevated numbers of macrophages in wounded tissue. Gene expression analysis of wounds tissue and macrophages isolated from miR-155(-/-) mice that were treated with interleukin-4 demonstrated an increased expression of miR-155 targets (BCL6, RhoA and SHIP1) as well as, the finding in inflammatory zone-1 (FIZZ1) gene, when compared with WT mice. Moreover, the up-regulated levels of FIZZ1 in the wound tissue of miR-155(-/-) mice correlated with an increased deposition of type-1 collagens, a phenomenon known to be beneficial in wound closure. Our data indicate that the absence of miR-155 has beneficial effects in the wound healing process.


Subject(s)
Dermis/metabolism , Dermis/pathology , MicroRNAs/physiology , Wound Healing/genetics , Animals , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Dermis/injuries , Humans , Immunoenzyme Techniques , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
J Cell Sci ; 127(Pt 6): 1169-78, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24463821

ABSTRACT

MicroRNA-149 (miR-149) is located within the first intron of the glypican-1 (GPC1) gene. GPC1 is a low affinity receptor for fibroblast growth factor (FGF2) that enhances FGF2 binding to its receptor (FGFR1), subsequently promoting FGF2-FGFR1 activation and signaling. Using bioinformatic approaches, both GPC1 and FGFR1 were identified and subsequently validated as targets for miR-149 (both the mature strand, miR-149, and the passenger strand, miR-149*) in endothelial cells (ECs). As a consequence of their targeting activity towards GPC1 and FGFR1, both miR-149 and miR-149* regulated FGF2 signaling and FGF2-induced responses in ECs, namely proliferation, migration and cord formation. Moreover, lentiviral overexpression of miR-149 reduced in vivo tumor-induced neovascularization. Importantly, FGF2 transcriptionally stimulated the expression of miR-149 independently of its host gene, therefore assuring the steady state of FGF2-induced responses through the regulation of the GPC1-FGFR1 binary complex in ECs.


Subject(s)
Fibroblast Growth Factor 2/physiology , Glypicans/genetics , Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/physiology , Neovascularization, Physiologic , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Cells, Cultured , Gene Expression , Glypicans/metabolism , Humans , Male , Mice , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , RNA Interference , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
13.
Curr Vasc Pharmacol ; 2013 May 13.
Article in English | MEDLINE | ID: mdl-23713860

ABSTRACT

Atherosclerosis (also known as arteriosclerotic vascular disease) is a chronic inflammatory disease of the arterial wall, characterized by the formation of lipid-laden lesions. The activation of endothelial cells at atherosclerotic lesion-prone sites in the arterial tree results in the up-regulation of cell adhesion molecules and chemokines, which mediate the recruitment of circulating monocytes. Accumulation of monocytes and monocyte-derived phagocytes in the wall of large arteries leads to chronic inflammation and the development and progression of atherosclerosis. The lesion experiences the following steps: foam cell formation, fatty streak accumulation, migration and proliferation of vascular smooth muscle cells, and fibrous cap formation. Finally, the rupture of the unstable fibrous cap causes thrombosis in complications of advanced lesions that lead to unstable coronary syndromes, myocardial infarction and stroke. MicroRNAs have recently emerged as a novel class of gene regulators at the post-transcriptional level. Several functions of vascular cells, such as cell differentiation, contraction, migration, proliferation and inflammation that are involved in angiogenesis, neointimal formation and lipid metabolism underlying various vascular diseases, have been found to be regulated by microRNAs and are described in the present review as well as their potential therapeutic application.

14.
Pharmacol Res ; 75: 15-27, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23603154

ABSTRACT

Endothelial cell dysfunction is a term which implies the dysregulation of normal endothelial cell functions, including impairment of the barrier functions, control of vascular tone, disturbance of proliferative, migratory and morphogenic capacities of endothelial cells, as well as control of leukocyte trafficking. MicroRNAs are short non-coding RNAs that have emerged as critical regulators of gene expression acting predominantly at the post-transcriptional level. This review summarizes the latest insights in the identification of endothelial-specific microRNAs and their targets, as well as their roles in controlling endothelial cell functions in both autocrine and paracrine manner. In addition, we discuss the therapeutic potential for the treatment of endothelial cell dysfunction and associated vascular pathophysiological conditions.


Subject(s)
Cardiovascular Diseases/therapy , Endothelial Cells , Endothelium, Vascular/physiopathology , Gene Targeting/methods , Genetic Therapy/methods , MicroRNAs/genetics , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Cell Differentiation/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gene Expression Regulation , Hemodynamics/genetics , Hemodynamics/physiology , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis
15.
Mol Cell Biol ; 33(11): 2339-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23547260

ABSTRACT

hsa-miR-33a and hsa-miR-33b, intronic microRNAs (miRNAs) located within the sterol regulatory element-binding protein 2 and 1 genes (Srebp-2 and -1), respectively, have recently been shown to regulate lipid homeostasis in concert with their host genes. Although the functional role of miR-33a and -b has been highly investigated, the role of their passenger strands, miR-33a* and -b*, remains unclear. Here, we demonstrate that miR-33a* and -b* accumulate to steady-state levels in human, mouse, and nonhuman primate tissues and share a similar lipid metabolism target gene network as their sister strands. Analogous to miR-33, miR-33* represses key enzymes involved in cholesterol efflux (ABCA1 and NPC1), fatty acid metabolism (CROT and CPT1a), and insulin signaling (IRS2). Moreover, miR-33* also targets key transcriptional regulators of lipid metabolism, including SRC1, SRC3, NFYC, and RIP140. Importantly, inhibition of either miR-33 or miR-33* rescues target gene expression in cells overexpressing pre-miR-33. Consistent with this, overexpression of miR-33* reduces fatty acid oxidation in human hepatic cells. Altogether, these data support a regulatory role for the miRNA* species and suggest that miR-33 regulates lipid metabolism through both arms of the miR-33/miR-33* duplex.


Subject(s)
Gene Expression Regulation , Lipid Metabolism/genetics , MicroRNAs/genetics , 3' Untranslated Regions , Animals , Base Sequence , Cell Line , Conserved Sequence , Fatty Acids/metabolism , Humans , Liver/cytology , Liver/physiology , Macaca mulatta/genetics , Macrophages/cytology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism
16.
FASEB J ; 27(4): 1460-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23271053

ABSTRACT

MicroRNA cluster miR-17-92 has been implicated in cardiovascular development and function, yet its precise mechanisms of action in these contexts are uncertain. This study aimed to investigate the role of miR-17-92 in morphogenesis and function of cardiac and smooth muscle tissues. To do so, a mouse model of conditional overexpression of miR-17-92 in cardiac and smooth muscle tissues was generated. Extensive cardiac functional studies identified a dose-dependent induction of dilated, hypertrophic cardiomyopathy, and arrhythmia inducibility in transgenic animals, which correlated with premature mortality (98.3 ± 42.5 d, P<0.0001). Expression analyses revealed the abundance of Pten transcript, a known miR-17-92 target, to be inversely correlated with miR-17-92 expression levels and heart size. In addition, we demonstrated through 3'-UTR luciferase assays and expression analyses that Connexin43 (Cx43) is a novel direct target of miR-19a/b and its expression is suppressed in transgenic hearts. Taken together, these data demonstrate that dysregulated expression of miR-17-92 during cardiovascular morphogenesis results in a lethal cardiomyopathy, possibly in part through direct repression of Pten and Cx43. This study highlights the importance of miR-17-92 in both normal and pathological functions of the heart, and provides a model that may serve as a useful platform to test novel antiarrhythmic therapeutics.


Subject(s)
Arrhythmias, Cardiac/genetics , Cardiomyopathy, Hypertrophic/genetics , MicroRNAs/genetics , Animals , Arrhythmias, Cardiac/physiopathology , Cardiomyopathy, Hypertrophic/mortality , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Connexin 43/genetics , Connexin 43/metabolism , Disease Models, Animal , Heart Defects, Congenital/genetics , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
17.
Cell Cycle ; 11(5): 922-33, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22333591

ABSTRACT

Cholesterol metabolism is tightly regulated at the cellular level and is essential for cellular growth. microRNAs (miRNAs), a class of noncoding RNAs, have emerged as critical regulators of gene expression, acting predominantly at posttranscriptional level. Recent work from our group and others has shown that hsa-miR-33a and hsa-miR-33b, miRNAs located within intronic sequences of the Srebp genes, regulate cholesterol and fatty acid metabolism in concert with their host genes. Here, we show that hsa-miR-33 family members modulate the expression of genes involved in cell cycle regulation and cell proliferation. MiR-33 inhibits the expression of the cyclin-dependent kinase 6 (CDK6) and cyclin D1 (CCND1), thereby reducing cell proliferation and cell cycle progression. Overexpression of miR-33 induces a significant G 1 cell cycle arrest in Huh7 and A549 cell lines. Most importantly, inhibition of miR-33 expression using 2'fluoro/methoxyethyl-modified (2'F/MOE-modified) phosphorothioate backbone antisense oligonucleotides improves liver regeneration after partial hepatectomy (PH) in mice, suggesting an important role for miR-33 in regulating hepatocyte proliferation during liver regeneration. Altogether, these results suggest that Srebp/miR-33 locus may cooperate to regulate cell proliferation, cell cycle progression and may also be relevant to human liver regeneration.


Subject(s)
MicroRNAs/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase 6/metabolism , G1 Phase Cell Cycle Checkpoints , HeLa Cells , Humans , Liver Regeneration/drug effects , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Phosphates/chemistry
18.
Arterioscler Thromb Vasc Biol ; 31(11): 2595-606, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21885851

ABSTRACT

OBJECTIVE: MicroRNAs play key roles in modulating a variety of cellular processes by posttranscriptional regulation of their target genes. Vascular endothelial growth factor (VEGF), VEGF receptor-2 (VEGFR2), and fibroblast growth factor receptor-1 (FGFR1) were identified by bioinformatic approaches and subsequently validated as targets of microRNA (miR)-16 and miR-424 in endothelial cells (ECs). METHODS AND RESULTS: Mimetics of these microRNAs reduced VEGF, VEGFR2, and FGFR1 expression, whereas specific antagonists enhanced their expression. Expression of mature miR-16 and miR-424 was upregulated on VEGF or basic fibroblast growth factor (bFGF) treatment. This upregulation was accompanied by a parallel increase in primary transcript (pri-miR)-16-1 and pri-miR-16-2 but not in pri-miR-424 levels, indicating a VEGF/bFGF-dependent transcriptional and posttranscriptional regulation of miR-16 and miR-424, respectively. Reduced expression of VEGFR2 and FGFR1 by miR-16 or miR-424 overexpression regulated VEGF and bFGF signaling through these receptors, thereby affecting the activity of downstream components of the pathways. Functionally, miR-16 or miR-424 overexpression reduced proliferation, migration, and cord formation of ECs in vitro, and lentiviral overexpression of miR-16 reduced the ability of ECs to form blood vessels in vivo. CONCLUSION: We conclude that these miRNAs fine-tune the expression of selected endothelial angiogenic mediators in response to these growth factors. Altogether, these findings suggest that miR-16 and miR-424 play important roles in regulating cell-intrinsic angiogenic activity of ECs.


Subject(s)
Endothelium, Vascular/physiology , MicroRNAs/physiology , Neovascularization, Physiologic/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology , 3' Untranslated Regions/physiology , Animals , Cattle , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Mice , Mice, SCID , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/physiology
19.
Am J Physiol Cell Physiol ; 300(3): C683-91, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21160032

ABSTRACT

The nitric oxide (NO)-soluble guanylate cyclase (sGC) pathway exerts most of its cellular actions through the activation of the cGMP-dependent protein kinase (PKG). Accumulation of extracellular matrix is one of the main structural changes in pathological conditions characterized by a decreased activity of this pathway, such as hypertension, diabetes, or aging, and it is a well-known fact that extracellular matrix proteins modulate cell phenotype through the interaction with membrane receptors such as integrins. The objectives of this study were 1) to evaluate whether extracellular matrix proteins, particularly fibronectin (FN), modulate PKG expression in contractile cells, 2) to analyze the mechanisms involved, and 3) to evaluate the functional consequences. FN increased type I PKG (PKG-I) protein content in human mesangial cells, an effect dependent on the interaction with ß(1)-integrin. The FN upregulation of PKG-I protein content was due to increased mRNA expression, determined by augmented transcriptional activity of the PKG-I promoter region. Akt and the transcription factor CCAAT enhancer-binding protein (C/EBP) mediated the genesis of these changes. FN also increased PKG-I in another type of contractile cell, rat vascular smooth muscle cells (RVSMC). Tirofiban, a pharmacological analog of FN, increased PKG-I protein content in RVSMC and rat aortic walls and magnified the hypotensive effect of dibutyryl cGMP in conscious Wistar rats. The present results provide evidence of a mechanism able to increase PKG-I protein content in contractile cells. Elucidation of this novel mechanism provides a rationale for future pharmacotherapy in certain vascular diseases.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cyclic GMP-Dependent Protein Kinases/biosynthesis , Fibronectins/physiology , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Transcriptional Activation/physiology , Up-Regulation/physiology , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/genetics , Fibronectins/metabolism , Humans , Male , Mesangial Cells/cytology , Mesangial Cells/enzymology , Mesangial Cells/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
20.
Hypertension ; 56(3): 484-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20679183

ABSTRACT

The aim of the present experiments was to evaluate the differences in arterial pressure between H-Ras lacking mice and control mice and to analyze the mechanisms involved in the genesis of the differences. H-Ras lacking mice and mouse embryonic fibroblasts from these animals were used. Blood pressure was measured using 3 different methods: direct intraarterial measurement in anesthetized animals, tail-cuff sphygmomanometer, and radiotelemetry. H-Ras lacking mice showed lower blood pressure than control animals. Moreover, the aorta protein content of endothelial nitric oxide synthase, soluble guanylyl cyclase, and cyclic guanosine monophosphate-dependent protein kinase was higher in H-Ras knockout mice than in control animals. The activity of these enzymes was increased, because urinary nitrite excretion, sodium nitroprusside-stimulated vascular cyclic guanosine monophosphate synthesis, and phosphorylated vasoactive-stimulated phosphoprotein in aortic tissue increased in these animals. Furthermore, mouse embryonic fibroblasts from H-Ras lacking mice showed higher cyclic guanosine monophosphate-dependent protein kinase promoter activity than control cells. These results strongly support the upregulation of the nitric oxide-cyclic guanosine monophosphate pathway in H-Ras-deficient mice. Moreover, they suggest that H-Ras pathway could be considered as a therapeutic target for hypertension treatment.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Hypotension/metabolism , Nitric Oxide/metabolism , ras Proteins/metabolism , Animals , Aorta/metabolism , Blood Pressure/physiology , Blotting, Northern , Blotting, Western , Cells, Cultured , Cyclic GMP/genetics , Cyclic GMP-Dependent Protein Kinases/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Hypotension/genetics , Immunohistochemistry , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , Soluble Guanylyl Cyclase , Statistics, Nonparametric , Up-Regulation/physiology , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...